1
|
Wrona MV, Ghosh R, Coll K, Chun C, Yousefzadeh MJ. The 3 I's of immunity and aging: immunosenescence, inflammaging, and immune resilience. FRONTIERS IN AGING 2024; 5:1490302. [PMID: 39478807 PMCID: PMC11521913 DOI: 10.3389/fragi.2024.1490302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Accepted: 09/23/2024] [Indexed: 11/02/2024]
Abstract
As we age, our immune system's ability to effectively respond to pathogens declines, a phenomenon known as immunosenescence. This age-related deterioration affects both innate and adaptive immunity, compromising immune function and leading to chronic inflammation that accelerates aging. Immunosenescence is characterized by alterations in immune cell populations and impaired functionality, resulting in increased susceptibility to infections, diminished vaccine efficacy, and higher prevalence of age-related diseases. Chronic low-grade inflammation further exacerbates these issues, contributing to a decline in overall health and resilience. This review delves into the characteristics of immunosenescence and examines the various intrinsic and extrinsic factors contributing to immune aging and how the hallmarks of aging and cell fates can play a crucial role in this process. Additionally, it discusses the impact of sex, age, social determinants, and gut microbiota health on immune aging, illustrating the complex interplay of these factors in altering immune function. Furthermore, the concept of immune resilience is explored, focusing on the metrics for assessing immune health and identifying strategies to enhance immune function. These strategies include lifestyle interventions such as diet, regular physical activity, stress management, and the use of gerotherapeutics and other approaches. Understanding and mitigating the effects of immunosenescence are crucial for developing interventions that support robust immune responses in aged individuals.
Collapse
Affiliation(s)
- Marianna V. Wrona
- Columbia University in the City of New York, New York, NY, United States
| | - Rituparna Ghosh
- Columbia Center for Human Longevity, Columbia University Medical Center, New York, NY, United States
- Columbia Center for Translational Immunology, Columbia University Medical Center, New York, NY, United States
- Department of Medicine, Columbia University Medical Center, New York, NY, United States
| | - Kaitlyn Coll
- Florida International University, Miami, FL, United States
| | - Connor Chun
- Bronx High School of Science, New York, NY, United States
| | - Matthew J. Yousefzadeh
- Columbia University in the City of New York, New York, NY, United States
- Columbia Center for Human Longevity, Columbia University Medical Center, New York, NY, United States
- Columbia Center for Translational Immunology, Columbia University Medical Center, New York, NY, United States
- Department of Medicine, Columbia University Medical Center, New York, NY, United States
| |
Collapse
|
2
|
Herzog CMS, Goeminne LJE, Poganik JR, Barzilai N, Belsky DW, Betts-LaCroix J, Chen BH, Chen M, Cohen AA, Cummings SR, Fedichev PO, Ferrucci L, Fleming A, Fortney K, Furman D, Gorbunova V, Higgins-Chen A, Hood L, Horvath S, Justice JN, Kiel DP, Kuchel GA, Lasky-Su J, LeBrasseur NK, Maier AB, Schilling B, Sebastiano V, Slagboom PE, Snyder MP, Verdin E, Widschwendter M, Zhavoronkov A, Moqri M, Gladyshev VN. Challenges and recommendations for the translation of biomarkers of aging. NATURE AGING 2024; 4:1372-1383. [PMID: 39285015 DOI: 10.1038/s43587-024-00683-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Accepted: 07/12/2024] [Indexed: 10/01/2024]
Abstract
Biomarkers of aging (BOA) are quantitative parameters that predict biological age and ideally its changes in response to interventions. In recent years, many promising molecular and omic BOA have emerged with an enormous potential for translational geroscience and improving healthspan. However, clinical translation remains limited, in part due to the gap between preclinical research and the application of BOA in clinical research and other translational settings. We surveyed experts in these areas to better understand current challenges for the translation of aging biomarkers. We identified six key barriers to clinical translation and developed guidance for the field to overcome them. Core recommendations include linking BOA to clinically actionable insights, improving affordability and availability to broad populations and validation of biomarkers that are robust and responsive at the level of individuals. Our work provides key insights and practical recommendations to overcome barriers impeding clinical translation of BOA.
Collapse
Affiliation(s)
- Chiara M S Herzog
- European Translational Oncology Prevention and Screening Institute, Universität Innsbruck, Innsbruck, Austria
| | - Ludger J E Goeminne
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Jesse R Poganik
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Nir Barzilai
- Institute for Aging Research, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Daniel W Belsky
- Department of Epidemiology, Butler Columbia Aging Center, Mailman School of Public Health, Columbia University, New York, NY, USA
| | | | - Brian H Chen
- San Francisco Coordinating Center, California Pacific Medical Center Research Institute, San Francisco, CA, USA
- Department of Epidemiology and Biostatistics, University of California, San Francisco, CA, USA
| | | | - Alan A Cohen
- Department of Environmental Health Sciences, Butler Columbia Aging Center, Mailman School of Public Health, Columbia University, New York, NY, USA
| | - Steven R Cummings
- San Francisco Coordinating Center, California Pacific Medical Center Research Institute, San Francisco, CA, USA
- Department of Epidemiology and Biostatistics, University of California, San Francisco, CA, USA
| | | | | | | | | | - David Furman
- Buck Institute for Research on Aging, Novato, CA, USA
- Stanford 1000 Immunomes Project, Stanford School of Medicine, Stanford, CA, USA
- The National Scientific and Research Council, Austral University, Buenos Aires, Argentina
| | - Vera Gorbunova
- Departments of Biology and Medicine, University of Rochester, Rochester, NY, USA
| | | | - Lee Hood
- Buck Institute for Research on Aging, Novato, CA, USA
- Phenome Health, Seattle, WA, USA
| | | | - Jamie N Justice
- XPRIZE Foundation, Culver City, CA, USA
- Department of Internal Medicine, Section on Gerontology and Geriatric Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Douglas P Kiel
- Hinda and Arthur Marcus Institute for Aging Research, Hebrew SeniorLife, Roslindale, MA, USA
- Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| | - George A Kuchel
- University of Connecticut School of Medicine, @UConnAging, Farmington, CT, USA
| | - Jessica Lasky-Su
- Department of Network Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Nathan K LeBrasseur
- Department of Physical Medicine and Rehabilitation, Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, USA
| | - Andrea B Maier
- Healthy Longevity Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Centre for Healthy Longevity, @AgeSingapore, National University Health System, Singapore, Singapore
- Department of Human Movement Sciences, @AgeAmsterdam, Amsterdam Movement Sciences, Faculty of Behavioural and Movement Sciences, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | | | - Vittorio Sebastiano
- Department of Obstetrics and Gynecology, School of Medicine, Stanford University, Stanford, CA, USA
| | - P Eline Slagboom
- Section of Molecular Epidemiology, Department of Biomedical Data Sciences, Leiden University Medical Center, Leiden, the Netherlands
| | - Michael P Snyder
- Department of Genetics, School of Medicine, Stanford University, Stanford, CA, USA
| | - Eric Verdin
- Buck Institute for Research on Aging, Novato, CA, USA
| | - Martin Widschwendter
- European Translational Oncology Prevention and Screening Institute, Universität Innsbruck, Innsbruck, Austria
- Department of Women's Cancer, EGA Institute for Women's Health, University College London, London, UK
- Department of Women's and Children's Health, Division of Obstetrics and Gynaecology, Karolinska Institutet, Stockholm, Sweden
| | | | - Mahdi Moqri
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
- Department of Genetics, School of Medicine, Stanford University, Stanford, CA, USA.
| | - Vadim N Gladyshev
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
3
|
Alvarez-Kuglen M, Ninomiya K, Qin H, Rodriguez D, Fiengo L, Farhy C, Hsu WM, Kirk B, Havas A, Feng GS, Roberts AJ, Anderson RM, Serrano M, Adams PD, Sharpee TO, Terskikh AV. ImAge quantitates aging and rejuvenation. NATURE AGING 2024; 4:1308-1327. [PMID: 39210148 DOI: 10.1038/s43587-024-00685-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Accepted: 07/11/2024] [Indexed: 09/04/2024]
Abstract
For efficient, cost-effective and personalized healthcare, biomarkers that capture aspects of functional, biological aging, thus predicting disease risk and lifespan more accurately and reliably than chronological age, are essential. We developed an imaging-based chromatin and epigenetic age (ImAge) that captures intrinsic age-related trajectories of the spatial organization of chromatin and epigenetic marks in single nuclei, in mice. We show that such trajectories readily emerge as principal changes in each individual dataset without regression on chronological age, and that ImAge can be computed using several epigenetic marks and DNA labeling. We find that interventions known to affect biological aging induce corresponding effects on ImAge, including increased ImAge upon chemotherapy treatment and decreased ImAge upon caloric restriction and partial reprogramming by transient OSKM expression in liver and skeletal muscle. Further, ImAge readouts from chronologically identical mice inversely correlated with their locomotor activity, suggesting that ImAge may capture elements of biological and functional age. In sum, we developed ImAge, an imaging-based biomarker of aging with single-cell resolution rooted in the analysis of spatial organization of epigenetic marks.
Collapse
Affiliation(s)
| | - Kenta Ninomiya
- Harry Perkins Institute of Medical Research, The University of Western Australia, Perth, Western Australia, Australia
| | - Haodong Qin
- Department of Physics, University of California San Diego, La Jolla, CA, USA
| | | | | | - Chen Farhy
- Sanford Burnham Prebys, La Jolla, CA, USA
| | - Wei-Mien Hsu
- Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Brian Kirk
- Sanford Burnham Prebys, La Jolla, CA, USA
| | | | - Gen-Sheng Feng
- School of Medicine, Univerity of California San Diego, La Jolla, CA, USA
| | | | - Rozalyn M Anderson
- University of Wisconsin, Madison, WI, USA
- GRECC, William S Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Manuel Serrano
- Institute for Research in Biomedicine (IRB Barcelona), Barcelona, Spain
- Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
- Altos Labs, Cambridge Institute of Science, Granta Park, UK
| | | | | | - Alexey V Terskikh
- Harry Perkins Institute of Medical Research, The University of Western Australia, Perth, Western Australia, Australia.
- The Scintillon Research Institute, San Diego, CA, USA.
| |
Collapse
|
4
|
Wang S, Rao Z, Cao R, Blaes AH, Coresh J, Deo R, Dubin R, Joshu CE, Lehallier B, Lutsey PL, Pankow JS, Post WS, Rotter JI, Sedaghat S, Tang W, Thyagarajan B, Walker KA, Ganz P, Platz EA, Guan W, Prizment A. Development, characterization, and replication of proteomic aging clocks: Analysis of 2 population-based cohorts. PLoS Med 2024; 21:e1004464. [PMID: 39316596 PMCID: PMC11460707 DOI: 10.1371/journal.pmed.1004464] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 10/08/2024] [Accepted: 08/22/2024] [Indexed: 09/26/2024] Open
Abstract
BACKGROUND Biological age may be estimated by proteomic aging clocks (PACs). Previous published PACs were constructed either in smaller studies or mainly in white individuals, and they used proteomic measures from only one-time point. In this study, we created de novo PACs and compared their performance to published PACs at 2 different time points in the Atherosclerosis Risk in Communities (ARIC) study of white and black participants (around 75% white and 25% black). MEDTHODS AND FINDINGS A total of 4,712 plasma proteins were measured using SomaScan in blood samples collected in 1990 to 1992 from 11,761 midlife participants (aged 46 to 70 years) and in 2011 to 2013 from 5,183 late-life participants (aged 66 to 90 years). The de novo ARIC PACs were constructed by training them against chronological age using elastic net regression in two-thirds of healthy participants in midlife and late life and validated in the remaining one-third of healthy participants at the corresponding time point. We also computed 3 published PACs. We estimated age acceleration for each PAC as residuals after regressing each PAC on chronological age. We also calculated the change in age acceleration from midlife to late life. We examined the associations of age acceleration and change in age acceleration with mortality through 2019 from all-cause, cardiovascular disease (CVD), cancer, and lower respiratory disease (LRD) using Cox proportional hazards regression in participants (irrespective of health) after excluding the training set. The model was adjusted for chronological age, smoking, body mass index (BMI), and other confounders. We externally validated the midlife PAC using the Multi-Ethnic Study of Atherosclerosis (MESA) Exam 1 data. The ARIC PACs had a slightly stronger correlation with chronological age than published PACs in healthy participants at each time point. Associations with mortality were similar for the ARIC PACs and published PACs. For late-life and midlife age acceleration for the ARIC PACs, respectively, hazard ratios (HRs) per 1 standard deviation were 1.65 and 1.38 (both p < 0.001) for all-cause mortality, 1.37 and 1.20 (both p < 0.001) for CVD mortality, 1.21 (p = 0.028) and 1.04 (p = 0.280) for cancer mortality, and 1.68 and 1.36 (both p < 0.001) for LRD mortality. For the change in age acceleration, HRs for all-cause, CVD, and LRD mortality were comparable to the HRs for late-life age acceleration. The association between the change in age acceleration and cancer mortality was not significant. The external validation of the midlife PAC in MESA showed significant associations with mortality, as observed for midlife participants in ARIC. The main limitation is that our PACs were constructed in midlife and late-life participants. It is unknown whether these PACs could be applied to young individuals. CONCLUSIONS In this longitudinal study, we found that the ARIC PACs and published PACs were similarly associated with an increased risk of mortality. These findings suggested that PACs show promise as biomarkers of biological age. PACs may be serve as tools to predict mortality and evaluate the effect of anti-aging lifestyle and therapeutic interventions.
Collapse
Affiliation(s)
- Shuo Wang
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Zexi Rao
- Division of Biostatistics and Health Data Science, School of Public Health, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Rui Cao
- Division of Biostatistics and Health Data Science, School of Public Health, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Anne H. Blaes
- Division of Hematology, Oncology and Transplantation, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Josef Coresh
- Departments of Population Health and Medicine, New York University Glossman School of Medicine, New York, New York, United States of America
| | - Rajat Deo
- Division of Cardiovascular Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Ruth Dubin
- Division of Nephrology, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
| | - Corinne E. Joshu
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, United States of America
- Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, Maryland, United States of America
| | | | - Pamela L. Lutsey
- Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - James S. Pankow
- Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Wendy S. Post
- Division of Cardiology, Department of Medicine, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Jerome I. Rotter
- Institute for Translational Genomics and Population Sciences, The Lundquist Institute for Biomedical Innovation; Department of Pediatrics, Harbor-UCLA Medical Center, Torrance, California, United States of America
| | - Sanaz Sedaghat
- Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Weihong Tang
- Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Bharat Thyagarajan
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Keenan A. Walker
- Laboratory of Behavioral Neuroscience, National Institute on Aging, National Institutes of Health, Baltimore, Maryland, United States of America
| | - Peter Ganz
- Department of Medicine, University of California, San Francisco, California, United States of America
| | - Elizabeth A. Platz
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, United States of America
- Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, Maryland, United States of America
| | - Weihua Guan
- Division of Biostatistics and Health Data Science, School of Public Health, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Anna Prizment
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, Minnesota, United States of America
| |
Collapse
|
5
|
Pugashetti JV, Kim JS, Bose S, Adegunsoye A, Linderholm AL, Chen CH, Strek ME, Flaherty KR, Murray S, Newton CA, Alqalyoobi S, Ma SF, Mychaleckyj JC, Bowler RP, Han MK, Curtis JL, Martinez FJ, Smith JA, Noth I, Oldham JM. Biological Age, Chronological Age, and Survival in Pulmonary Fibrosis: A Causal Mediation Analysis. Am J Respir Crit Care Med 2024; 210:639-647. [PMID: 38843133 PMCID: PMC11389564 DOI: 10.1164/rccm.202310-1887oc] [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: 10/27/2023] [Accepted: 06/05/2024] [Indexed: 06/30/2024] Open
Abstract
Rationale: Accelerated biological aging has been implicated in the development of interstitial lung disease (ILD) and other diseases of aging but remains poorly understood. Objectives: To identify plasma proteins that mediate the relationship between chronological age and survival association in patients with ILD. Methods: Causal mediation analysis was performed to identify plasma proteins that mediated the chronological age-survival relationship in an idiopathic pulmonary fibrosis discovery cohort. Proteins mediating this relationship after adjustment for false discovery were advanced for testing in an independent ILD validation cohort and explored in a chronic obstructive pulmonary disease cohort. A proteomic-based measure of biological age was constructed and survival analysis performed, assessing the impact of biological age and peripheral blood telomere length on the chronological age-survival relationship. Measurements and Main Results: Twenty-two proteins mediated the chronological age-survival relationship after adjustment for false discovery in the idiopathic pulmonary fibrosis discovery cohort (n = 874), with 19 remaining significant mediators of this relationship in the ILD validation cohort (n = 983) and one mediating this relationship in the chronic obstructive pulmonary disease cohort. Latent transforming growth factor-β binding protein 2 and ectodysplasin A2 receptor showed the strongest mediation across cohorts. A proteomic measure of biological age completely attenuated the chronological age-survival association and better discriminated survival than chronological age. Results were robust to adjustment for peripheral blood telomere length, which did not mediate the chronological age-survival relationship. Conclusions: Molecular measures of aging completely mediate the relationship between chronological age and survival, suggesting that chronological age has no direct effect on ILD survival.
Collapse
Affiliation(s)
| | - John S Kim
- Division of Pulmonary and Critical Care Medicine and
| | | | - Ayodeji Adegunsoye
- Section of Pulmonary and Critical Care Medicine, University of Chicago, Chicago, Illinois
| | - Angela L Linderholm
- Division of Pulmonary, Critical Care and Sleep Medicine, University of California, Davis, Sacramento, California
| | - Ching-Hsien Chen
- Division of Pulmonary, Critical Care and Sleep Medicine, University of California, Davis, Sacramento, California
| | - Mary E Strek
- Section of Pulmonary and Critical Care Medicine, University of Chicago, Chicago, Illinois
| | - Kevin R Flaherty
- Division of Pulmonary and Critical Care Medicine
- Pulmonary Fibrosis Foundation, Chicago, Illinois
| | | | - Chad A Newton
- Pulmonary Fibrosis Foundation, Chicago, Illinois
- Division of Pulmonary and Critical Care, University of Texas Southwestern, Dallas, Texas
| | - Shehabaldin Alqalyoobi
- Division of Pulmonary and Critical Care, East Carolina University, Greenville, North Carolina
| | - Shwu-Fan Ma
- Division of Pulmonary and Critical Care Medicine and
| | - Josyf C Mychaleckyj
- School of Public Health Sciences, University of Virginia, Charlottesville, Virginia
| | - Russell P Bowler
- Division of Pulmonary Medicine, National Jewish Health, Denver, Colorado
| | - MeiLan K Han
- Division of Pulmonary and Critical Care Medicine
| | - Jeffrey L Curtis
- Division of Pulmonary and Critical Care Medicine
- Medical Service, Veterans Affairs Ann Arbor Healthcare System, Ann Arbor, Michigan; and
| | - Fernando J Martinez
- Division of Pulmonary and Critical Care Medicine, Weill Cornell Medical Center, New York, New York
| | - Jennifer A Smith
- Department of Epidemiology, University of Michigan, Ann Arbor, Michigan
| | - Imre Noth
- Division of Pulmonary and Critical Care Medicine and
| | - Justin M Oldham
- Division of Pulmonary and Critical Care Medicine
- Department of Epidemiology, University of Michigan, Ann Arbor, Michigan
| |
Collapse
|
6
|
Rezaeian AH, Wei W. Molecular signaling and clinical implications in the human aging-cancer cycle. Semin Cancer Biol 2024; 106-107:28-42. [PMID: 39197809 DOI: 10.1016/j.semcancer.2024.08.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 08/07/2024] [Accepted: 08/09/2024] [Indexed: 09/01/2024]
Abstract
It is well documented that aging is associated with cancer, and likewise, cancer survivors display accelerated aging. As the number of aging individuals and cancer survivors continues to grow, it raises additional concerns across society. Therefore, unraveling the molecular mechanisms of aging in tissues is essential to developing effective therapies to fight the aging and cancer diseases in cancer survivors and cancer patients. Indeed, cellular senescence is a critical response, or a natural barrier to suppress the transition of normal cells into cancer cells, however, hypoxia which is physiologically required to maintain the stem cell niche, is increased by aging and inhibits senescence in tissues. Interestingly, oxygen restriction or hypoxia increases longevity and slows the aging process in humans, but hypoxia can also drive angiogenesis to facilitate cancer progression. In addition, cancer treatment is considered as one of the major reasons that drive cellular senescence, subsequently followed by accelerated aging. Several clinical trials have recently evaluated inhibitors to eliminate senescent cells. However, some mechanisms of aging typically can also retard cancer cell growth and progression, which might require careful strategy for better clinical outcomes. Here we describe the molecular regulation of aging and cancer in crosstalk with DNA damage and hypoxia signaling pathways in cancer patients and cancer survivors. We also update several therapeutic strategies that might be critical in reversing the cancer treatment-associated aging process.
Collapse
Affiliation(s)
- Abdol-Hossein Rezaeian
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, United States.
| | - Wenyi Wei
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, United States.
| |
Collapse
|
7
|
Marttila S, Rajić S, Ciantar J, Mak JKL, Junttila IS, Kummola L, Hägg S, Raitoharju E, Kananen L. Biological aging of different blood cell types. GeroScience 2024:10.1007/s11357-024-01287-w. [PMID: 39060678 DOI: 10.1007/s11357-024-01287-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Accepted: 07/10/2024] [Indexed: 07/28/2024] Open
Abstract
Biological age (BA) captures detrimental age-related changes. The best-known and most-used BA indicators include DNA methylation-based epigenetic clocks and telomere length (TL). The most common biological sample material for epidemiological aging studies, whole blood, is composed of different cell types. We aimed to compare differences in BAs between blood cell types and assessed the BA indicators' cell type-specific associations with chronological age (CA). An analysis of DNA methylation-based BA indicators, including TL, methylation level at cg16867657 in ELOVL2, as well as the Hannum, Horvath, DNAmPhenoAge, and DunedinPACE epigenetic clocks, was performed on 428 biological samples of 12 blood cell types. BA values were different in the majority of the pairwise comparisons between cell types, as well as in comparison to whole blood (p < 0.05). DNAmPhenoAge showed the largest cell type differences, up to 44.5 years and DNA methylation-based TL showed the lowest differences. T cells generally had the "youngest" BA values, with differences across subsets, whereas monocytes had the "oldest" values. All BA indicators, except DunedinPACE, strongly correlated with CA within a cell type. Some differences such as DNAmPhenoAge-difference between naïve CD4 + T cells and monocytes were constant regardless of the blood donor's CA (range 20-80 years), while for DunedinPACE they were not. In conclusion, DNA methylation-based indicators of BA exhibit cell type-specific characteristics. Our results have implications for understanding the molecular mechanisms underlying epigenetic clocks and underscore the importance of considering cell composition when utilizing them as indicators for the success of aging interventions.
Collapse
Affiliation(s)
- Saara Marttila
- Molecular Epidemiology (MOLE), Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland.
- Gerontology Research Center, Tampere University, Tampere, Finland.
- Tays Research Services, Wellbeing Services County of Pirkanmaa, Tampere University Hospital, Tampere, Finland.
| | - Sonja Rajić
- Molecular Epidemiology (MOLE), Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Joanna Ciantar
- Molecular Epidemiology (MOLE), Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Jonathan K L Mak
- Department of Medical Epidemiology and Biostatistics, Karolinska Institute, Stockholm, Sweden
- Department of Pharmacology and Pharmacy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Ilkka S Junttila
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Fimlab Laboratories, Tampere, Finland
- Northern Finland Laboratory Centre (NordLab), Oulu, Finland
- Research Unit of Biomedicine, University of Oulu, Oulu, Finland
| | - Laura Kummola
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Sara Hägg
- Department of Medical Epidemiology and Biostatistics, Karolinska Institute, Stockholm, Sweden
| | - Emma Raitoharju
- Molecular Epidemiology (MOLE), Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Tays Research Services, Wellbeing Services County of Pirkanmaa, Tampere University Hospital, Tampere, Finland
| | - Laura Kananen
- Gerontology Research Center, Tampere University, Tampere, Finland.
- Department of Medical Epidemiology and Biostatistics, Karolinska Institute, Stockholm, Sweden.
- Faculty of Social Sciences (Health Sciences), Tampere University, Tampere, Finland.
- Department of Neurobiology, Care Sciences and Society (NVS), Karolinska Institute, Stockholm, Sweden.
| |
Collapse
|
8
|
Yusri K, Kumar S, Fong S, Gruber J, Sorrentino V. Towards Healthy Longevity: Comprehensive Insights from Molecular Targets and Biomarkers to Biological Clocks. Int J Mol Sci 2024; 25:6793. [PMID: 38928497 PMCID: PMC11203944 DOI: 10.3390/ijms25126793] [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: 05/23/2024] [Revised: 06/16/2024] [Accepted: 06/18/2024] [Indexed: 06/28/2024] Open
Abstract
Aging is a complex and time-dependent decline in physiological function that affects most organisms, leading to increased risk of age-related diseases. Investigating the molecular underpinnings of aging is crucial to identify geroprotectors, precisely quantify biological age, and propose healthy longevity approaches. This review explores pathways that are currently being investigated as intervention targets and aging biomarkers spanning molecular, cellular, and systemic dimensions. Interventions that target these hallmarks may ameliorate the aging process, with some progressing to clinical trials. Biomarkers of these hallmarks are used to estimate biological aging and risk of aging-associated disease. Utilizing aging biomarkers, biological aging clocks can be constructed that predict a state of abnormal aging, age-related diseases, and increased mortality. Biological age estimation can therefore provide the basis for a fine-grained risk stratification by predicting all-cause mortality well ahead of the onset of specific diseases, thus offering a window for intervention. Yet, despite technological advancements, challenges persist due to individual variability and the dynamic nature of these biomarkers. Addressing this requires longitudinal studies for robust biomarker identification. Overall, utilizing the hallmarks of aging to discover new drug targets and develop new biomarkers opens new frontiers in medicine. Prospects involve multi-omics integration, machine learning, and personalized approaches for targeted interventions, promising a healthier aging population.
Collapse
Affiliation(s)
- Khalishah Yusri
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117596, Singapore
- Healthy Longevity Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore
| | - Sanjay Kumar
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117596, Singapore
- Healthy Longevity Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore
| | - Sheng Fong
- Department of Geriatric Medicine, Singapore General Hospital, Singapore 169608, Singapore
- Clinical and Translational Sciences PhD Program, Duke-NUS Medical School, Singapore 169857, Singapore
| | - Jan Gruber
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117596, Singapore
- Healthy Longevity Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore
- Science Division, Yale-NUS College, Singapore 138527, Singapore
| | - Vincenzo Sorrentino
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117596, Singapore
- Healthy Longevity Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore
- Department of Medical Biochemistry, Amsterdam UMC, Amsterdam Gastroenterology Endocrinology Metabolism and Amsterdam Neuroscience Cellular & Molecular Mechanisms, University of Amsterdam, Meibergdreef 9, 1105AZ Amsterdam, The Netherlands
| |
Collapse
|
9
|
Shim J, Fleisch E, Barata F. Circadian rhythm analysis using wearable-based accelerometry as a digital biomarker of aging and healthspan. NPJ Digit Med 2024; 7:146. [PMID: 38834756 DOI: 10.1038/s41746-024-01111-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Accepted: 04/12/2024] [Indexed: 06/06/2024] Open
Abstract
Recognizing the pivotal role of circadian rhythm in the human aging process and its scalability through wearables, we introduce CosinorAge, a digital biomarker of aging developed from wearable-derived circadian rhythmicity from 80,000 midlife and older adults in the UK and US. A one-year increase in CosinorAge corresponded to 8-12% higher all-cause and cause-specific mortality risks and 3-14% increased prospective incidences of age-related diseases. CosinorAge also captured a non-linear decline in resilience and physical functioning, evidenced by an 8-33% reduction in self-rated health and a 3-23% decline in health-related quality of life score, adjusting for covariates and multiple testing. The associations were robust in sensitivity analyses and external validation using an independent cohort from a disparate geographical region using a different wearable device. Moreover, we illustrated a heterogeneous impact of circadian parameters associated with biological aging, with young (<45 years) and fast agers experiencing a substantially delayed acrophase with a 25-minute difference in peak timing compared to slow agers, diminishing to a 7-minute difference in older adults (>65 years). We demonstrated a significant enhancement in the predictive performance when integrating circadian rhythmicity in the estimation of biological aging over physical activity. Our findings underscore CosinorAge's potential as a scalable, economic, and digital solution for promoting healthy longevity, elucidating the critical and multifaceted circadian rhythmicity in aging processes. Consequently, our research contributes to advancing preventive measures in digital medicine.
Collapse
Affiliation(s)
- Jinjoo Shim
- Centre for Digital Health Interventions, Department of Management, Technology, and Economics, ETH Zurich, Zurich, Switzerland.
| | - Elgar Fleisch
- Centre for Digital Health Interventions, Department of Management, Technology, and Economics, ETH Zurich, Zurich, Switzerland
- Centre for Digital Health Interventions, Institute of Technology Management, University of St. Gallen, St. Gallen, Switzerland
| | - Filipe Barata
- Centre for Digital Health Interventions, Department of Management, Technology, and Economics, ETH Zurich, Zurich, Switzerland
| |
Collapse
|
10
|
Zane F, MacMurray C, Guillermain C, Cansell C, Todd N, Rera M. Ageing as a two-phase process: theoretical framework. FRONTIERS IN AGING 2024; 5:1378351. [PMID: 38651031 PMCID: PMC11034523 DOI: 10.3389/fragi.2024.1378351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Accepted: 02/26/2024] [Indexed: 04/25/2024]
Abstract
Human ageing, along with the ageing of conventional model organisms, is depicted as a continuous and progressive decline of biological capabilities accompanied by an exponentially increasing mortality risk. However, not all organisms experience ageing identically and our understanding of the phenomenon is coloured by human-centric views. Ageing is multifaceted and influences a diverse range of species in varying ways. Some undergo swift declines post-reproduction, while others exhibit insubstantial changes throughout their existence. This vast array renders defining universally applicable "ageing attributes" a daunting task. It is nonetheless essential to recognize that not all ageing features are organism-specific. These common attributes have paved the way for identifying "hallmarks of ageing," processes that are intertwined with age, amplified during accelerated ageing, and manipulations of which can potentially modulate or even reverse the ageing process. Yet, a glaring observation is that individuals within a single population age at varying rates. To address this, demographers have coined the term 'frailty'. Concurrently, scientific advancements have ushered in the era of molecular clocks. These innovations enable a distinction between an individual's chronological age (time since birth) and biological age (physiological status and mortality risk). In 2011, the "Smurf" phenotype was unveiled in Drosophila, delineating an age-linked escalation in intestinal permeability that presages imminent mortality. It not only acts as a predictor of natural death but identifies individuals exhibiting traits normally described as age-related. Subsequent studies have revealed the phenotype in organisms like nematodes, zebrafish, and mice, invariably acting as a death predictor. Collectively, these findings have steered our conception of ageing towards a framework where ageing is not linear and continuous but marked by two distinct, necessary phases, discernible in vivo, courtesy of the Smurf phenotype. This framework includes a mathematical enunciation of longevity trends based on three experimentally measurable parameters. It facilitates a fresh perspective on the evolution of ageing as a function. In this article, we aim to delineate and explore the foundational principles of this innovative framework, emphasising its potential to reshape our understanding of ageing, challenge its conventional definitions, and recalibrate our comprehension of its evolutionary trajectory.
Collapse
Affiliation(s)
- Flaminia Zane
- Université Paris Cité, INSERM UMR U1284, Paris, France
| | | | | | - Céline Cansell
- Université Paris-Saclay, AgroParisTech, INRAE, UMR PNCA, Palaiseau, France
| | - Nicolas Todd
- Eco-Anthropologie (EA), Muséum National d’Histoire Naturelle, CNRS, Université de Paris, Musée de l’Homme, Paris, France
| | - Michael Rera
- Université Paris Cité, Institut Jacques Monod, CNRS UMR 7592, Paris, France
| |
Collapse
|
11
|
Chen R, Zhang S, Peng G, Meng W, Borchert G, Wang W, Yu Z, Liao H, Ge Z, He M, Zhu Z. Deep neural network-estimated age using optical coherence tomography predicts mortality. GeroScience 2024; 46:1703-1711. [PMID: 37733221 PMCID: PMC10828229 DOI: 10.1007/s11357-023-00920-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Accepted: 08/22/2023] [Indexed: 09/22/2023] Open
Abstract
The concept of biological age has emerged as a measurement that reflects physiological and functional decline with ageing. Here we aimed to develop a deep neural network (DNN) model that predicts biological age from optical coherence tomography (OCT). A total of 84,753 high-quality OCT images from 53,159 individuals in the UK Biobank were included, among which 12,631 3D-OCT images from 8,541 participants without any reported medical conditions at baseline were used to develop an age prediction model. For the remaining 44,618 participants, OCT age gap, the difference between the OCT-predicted age and chronological age, was calculated for each participant. Cox regression models assessed the association between OCT age gap and mortality. The DNN model predicted age with a mean absolute error of 3.27 years and showed a strong correlation of 0.85 with chronological age. After a median follow-up of 11.0 years (IQR 10.9-11.1 years), 2,429 deaths (5.44%) were recorded. For each 5-year increase in OCT age gap, there was an 8% increased mortality risk (hazard ratio [HR] = 1.08, CI:1.02-1.13, P = 0.004). Compared with an OCT age gap within ± 4 years, OCT age gap less than minus 4 years was associated with a 16% decreased mortality risk (HR = 0.84, CI: 0.75-0.94, P = 0.002) and OCT age gap more than 4 years showed an 18% increased risk of death incidence (HR = 1.18, CI: 1.02-1.37, P = 0.026). OCT imaging could serve as an ageing biomarker to predict biological age with high accuracy and the OCT age gap, defined as the difference between the OCT-predicted age and chronological age, can be used as a marker of the risk of mortality.
Collapse
Affiliation(s)
- Ruiye Chen
- Centre for Eye Research Australia; Ophthalmology, University of Melbourne, Melbourne, Australia
- Ophthalmology, Department of Surgery, University of Melbourne, Melbourne, Australia
| | - Shiran Zhang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou, China
| | - Guankai Peng
- Guangzhou Vision Tech Medical Technology Co., Ltd, GuangZhou, China
| | - Wei Meng
- Guangzhou Vision Tech Medical Technology Co., Ltd, GuangZhou, China
| | - Grace Borchert
- Centre for Eye Research Australia; Ophthalmology, University of Melbourne, Melbourne, Australia
| | - Wei Wang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou, China
| | - Zhen Yu
- Central Clinical School, Monash University, Melbourne, Australia
| | - Huan Liao
- Epigenetics and Neural Plasticity Laboratory, Florey Institute of Neuroscience and Mental Health, University of Melbourne, Melbourne, Australia
| | - Zongyuan Ge
- Faculty of IT, Monash University, Melbourne, Australia
- Monash Medical AI, Monash University, Melbourne, Australia
| | - Mingguang He
- Centre for Eye Research Australia; Ophthalmology, University of Melbourne, Melbourne, Australia.
- Ophthalmology, Department of Surgery, University of Melbourne, Melbourne, Australia.
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou, China.
| | - Zhuoting Zhu
- Centre for Eye Research Australia; Ophthalmology, University of Melbourne, Melbourne, Australia.
- Ophthalmology, Department of Surgery, University of Melbourne, Melbourne, Australia.
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou, China.
| |
Collapse
|
12
|
Aroke EN, Srinivasasainagendra V, Kottae P, Quinn TL, Wiggins AM, Hobson J, Kinnie K, Stoudmire T, Tiwari HK, Goodin BR. The Pace of Biological Aging Predicts Nonspecific Chronic Low Back Pain Severity. THE JOURNAL OF PAIN 2024; 25:974-983. [PMID: 37907115 PMCID: PMC10960701 DOI: 10.1016/j.jpain.2023.10.018] [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: 06/24/2023] [Revised: 10/15/2023] [Accepted: 10/21/2023] [Indexed: 11/02/2023]
Abstract
This study aimed to determine if and how the pace of biological aging was associated with nonspecific chronic low back pain (cLBP) and compare what measure of epigenetic age acceleration most strongly predicts cLBP outcomes. We used the Dunedin Pace of Aging from the Epigenome (DunedinPACE), Horvath's, Hannum's, and PhenoAge clocks to determine the pace of biological aging in 69 cLBP, and 49 pain-free controls (PFCs) adults, ages 18 to 85 years. On average, participants with cLBP had higher DunedinPACE (P < .001) but lower Horvath (P = .04) and Hannum (P = .02) accelerated epigenetic age than PFCs. There was no significant difference in PhenoAge acceleration between the cLBP and PFC groups (P = .97). DunedinPACE had the largest effect size (Cohen's d = .78) on group differences. In univariate regressions, a unit increase in DunedinPACE score was associated with 265.98 times higher odds of cLBP than the PFC group (P < .001). After controlling for sex, race, and body mass index (BMI), the odds ratio of cLBP to PFC group was 149.62 (P < .001). Furthermore, among participants with cLBP, DunedinPACE scores positively correlated with pain severity (rs = .385, P = .001) and interference (rs = .338, P = .005). Epigenetic age acceleration from Horvath, Hannum, and PhenoAge clocks were not significant predictors of cLBP. The odds of a faster pace of biological aging are higher among adults with cLBP, and this was associated with greater pain severity and disability. Future interventions to slow the pace of biological aging may improve cLBP outcomes. PERSPECTIVE: Accelerated epigenetic aging is common among adults with nonspecific cLBP. Higher DunedinPACE scores positively correlate with pain severity and interference, and better predict cLBP than other DNA methylation clocks. Interventions to slow the pace of biological aging may be viable targets for improving pain outcomes.
Collapse
Affiliation(s)
- Edwin N. Aroke
- School of Nursing, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Vinodh Srinivasasainagendra
- Department of Biostatistics, School of Public Health, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Pooja Kottae
- Department of Computer Science, College of Arts and Sciences, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Tammie L. Quinn
- Department of Psychology, College of Arts and Sciences, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Asia M. Wiggins
- Department of Psychology, College of Arts and Sciences, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Joanna Hobson
- Department of Psychology, College of Arts and Sciences, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Kiari Kinnie
- School of Nursing, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Tonya Stoudmire
- School of Nursing, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Hemant K. Tiwari
- Department of Biostatistics, School of Public Health, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Burel R. Goodin
- Department of Anesthesiology, School of Medicine, Washington University, St Louis, USA
| |
Collapse
|
13
|
Wang S, El Jurdi N, Thyagarajan B, Prizment A, Blaes AH. Accelerated Aging in Cancer Survivors: Cellular Senescence, Frailty, and Possible Opportunities for Interventions. Int J Mol Sci 2024; 25:3319. [PMID: 38542292 PMCID: PMC10970400 DOI: 10.3390/ijms25063319] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2024] [Revised: 03/08/2024] [Accepted: 03/12/2024] [Indexed: 06/02/2024] Open
Abstract
The population of cancer survivors has markedly increased due to the rapid improvements in cancer treatment. However, cancer survivors experience accelerated aging, which leads to chronic diseases and other age-related conditions, such as frailty. Those conditions may persist years after cancer diagnosis and treatment. Cellular senescence, a hallmark of aging, is one of the mechanisms that contribute to accelerated aging in cancer survivors. Several aging measures, including measures based on clinical markers and biomarkers, have been proposed to estimate the aging process, and some of them have shown associations with mortality and frailty in cancer survivors. Several anti-aging interventions, including lifestyle changes and anti-aging drugs, have been proposed. Future research, particularly in large-scale studies, is needed to determine the efficiency of these aging measures and anti-aging interventions before considering their application in clinics. This review focuses on the mechanisms of cellular senescence and accelerated aging in cancer survivors, assessment of the aging process using clinical markers and biomarkers, and the high prevalence of frailty in that population, as well as possible opportunities for anti-aging interventions. A deeper understanding of aging measures and anti-aging interventions in cancer survivors will contribute to the development of effective strategies to mitigate accelerated aging in cancer survivors and improve their quality of life.
Collapse
Affiliation(s)
- Shuo Wang
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN 55455, USA
| | - Najla El Jurdi
- Division of Hematology, Oncology and Transplantation, University of Minnesota, Minneapolis, MN 55455, USA
| | - Bharat Thyagarajan
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN 55455, USA
| | - Anna Prizment
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN 55455, USA
| | - Anne H. Blaes
- Division of Hematology, Oncology and Transplantation, University of Minnesota, Minneapolis, MN 55455, USA
| |
Collapse
|
14
|
Sluiskes MH, Goeman JJ, Beekman M, Slagboom PE, Putter H, Rodríguez-Girondo M. Clarifying the biological and statistical assumptions of cross-sectional biological age predictors: an elaborate illustration using synthetic and real data. BMC Med Res Methodol 2024; 24:58. [PMID: 38459475 PMCID: PMC10921716 DOI: 10.1186/s12874-024-02181-x] [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: 06/22/2023] [Accepted: 02/15/2024] [Indexed: 03/10/2024] Open
Abstract
BACKGROUND There is divergence in the rate at which people age. The concept of biological age is postulated to capture this variability, and hence to better represent an individual's true global physiological state than chronological age. Biological age predictors are often generated based on cross-sectional data, using biochemical or molecular markers as predictor variables. It is assumed that the difference between chronological and predicted biological age is informative of one's chronological age-independent aging divergence ∆. METHODS We investigated the statistical assumptions underlying the most popular cross-sectional biological age predictors, based on multiple linear regression, the Klemera-Doubal method or principal component analysis. We used synthetic and real data to illustrate the consequences if this assumption does not hold. RESULTS The most popular cross-sectional biological age predictors all use the same strong underlying assumption, namely that a candidate marker of aging's association with chronological age is directly informative of its association with the aging rate ∆. We called this the identical-association assumption and proved that it is untestable in a cross-sectional setting. If this assumption does not hold, weights assigned to candidate markers of aging are uninformative, and no more signal may be captured than if markers would have been assigned weights at random. CONCLUSIONS Cross-sectional methods for predicting biological age commonly use the untestable identical-association assumption, which previous literature in the field had never explicitly acknowledged. These methods have inherent limitations and may provide uninformative results, highlighting the importance of researchers exercising caution in the development and interpretation of cross-sectional biological age predictors.
Collapse
Affiliation(s)
- Marije H Sluiskes
- Medical Statistics, Department of Biomedical Data Sciences, Leiden University Medical Center, Leiden, the Netherlands.
| | - Jelle J Goeman
- Medical Statistics, Department of Biomedical Data Sciences, Leiden University Medical Center, Leiden, the Netherlands
| | - Marian Beekman
- Molecular Epidemiology, Department of Biomedical Data Sciences, Leiden University Medical Center, Leiden, the Netherlands
| | - P Eline Slagboom
- Molecular Epidemiology, Department of Biomedical Data Sciences, Leiden University Medical Center, Leiden, the Netherlands
- Max Planck Institute for the Biology of Ageing, Cologne, Germany
| | - Hein Putter
- Medical Statistics, Department of Biomedical Data Sciences, Leiden University Medical Center, Leiden, the Netherlands
| | - Mar Rodríguez-Girondo
- Medical Statistics, Department of Biomedical Data Sciences, Leiden University Medical Center, Leiden, the Netherlands
| |
Collapse
|
15
|
Moqri M, Herzog C, Poganik JR, Ying K, Justice JN, Belsky DW, Higgins-Chen AT, Chen BH, Cohen AA, Fuellen G, Hägg S, Marioni RE, Widschwendter M, Fortney K, Fedichev PO, Zhavoronkov A, Barzilai N, Lasky-Su J, Kiel DP, Kennedy BK, Cummings S, Slagboom PE, Verdin E, Maier AB, Sebastiano V, Snyder MP, Gladyshev VN, Horvath S, Ferrucci L. Validation of biomarkers of aging. Nat Med 2024; 30:360-372. [PMID: 38355974 PMCID: PMC11090477 DOI: 10.1038/s41591-023-02784-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Accepted: 12/19/2023] [Indexed: 02/16/2024]
Abstract
The search for biomarkers that quantify biological aging (particularly 'omic'-based biomarkers) has intensified in recent years. Such biomarkers could predict aging-related outcomes and could serve as surrogate endpoints for the evaluation of interventions promoting healthy aging and longevity. However, no consensus exists on how biomarkers of aging should be validated before their translation to the clinic. Here, we review current efforts to evaluate the predictive validity of omic biomarkers of aging in population studies, discuss challenges in comparability and generalizability and provide recommendations to facilitate future validation of biomarkers of aging. Finally, we discuss how systematic validation can accelerate clinical translation of biomarkers of aging and their use in gerotherapeutic clinical trials.
Collapse
Affiliation(s)
- Mahdi Moqri
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Department of Genetics, School of Medicine, Stanford University, Stanford, CA, USA
- Department of Obstetrics and Gynecology, School of Medicine, Stanford University, Stanford, CA, USA
| | - Chiara Herzog
- European Translational Oncology Prevention and Screening Institute, Universität Innsbruck, Innsbruck, Austria
| | - Jesse R Poganik
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Kejun Ying
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- T.H. Chan School of Public Health, Harvard University, Boston, MA, USA
| | - Jamie N Justice
- Department of Internal Medicine, Section on Gerontology and Geriatric Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Daniel W Belsky
- Department of Epidemiology, Butler Columbia Aging Center, Mailman School of Public Health, Columbia University, New York, NY, USA
| | | | - Brian H Chen
- Herbert Wertheim School of Public Health and Human Longevity Science, University of California San Diego, San Diego, CA, USA
| | - Alan A Cohen
- Department of Environmental Health Sciences, Butler Columbia Aging Center, Mailman School of Public Health, Columbia University, New York, NY, USA
| | - Georg Fuellen
- Institute for Biostatistics and Informatics in Medicine and Ageing Research, Rostock University Medical Center, Rostock, Germany
- School of Medicine, University College Dublin, Dublin, Ireland
| | - Sara Hägg
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Riccardo E Marioni
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
| | - Martin Widschwendter
- European Translational Oncology Prevention and Screening Institute, Universität Innsbruck, Innsbruck, Austria
- Department of Women's Cancer, EGA Institute for Women's Health, University College London, London, UK
- Department of Women's and Children's Health, Division of Obstetrics and Gynaecology, Karolinska Institutet, Stockholm, Sweden
| | | | | | | | - Nir Barzilai
- Institute for Aging Research, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Jessica Lasky-Su
- Department of Network Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Douglas P Kiel
- Musculoskeletal Research Center, Hinda and Arthur Marcus Institute for Aging Research and Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| | - Brian K Kennedy
- Healthy Longevity Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Healthy Longevity Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Centre for Healthy Longevity, @AgeSingapore, National University Health System, Singapore, Singapore
| | - Steven Cummings
- San Francisco Coordinating Center, California Pacific Medical Center Research Institute, San Francisco, CA, USA
- Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, CA, USA
| | - P Eline Slagboom
- Section of Molecular Epidemiology, Department of Biomedical Data Sciences, Leiden University Medical Center, Leiden, the Netherlands
| | - Eric Verdin
- Buck Institute for Research on Aging, Novato, CA, USA
| | - Andrea B Maier
- Healthy Longevity Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Centre for Healthy Longevity, @AgeSingapore, National University Health System, Singapore, Singapore
- Department of Human Movement Sciences, @AgeAmsterdam, Amsterdam Movement Sciences, Faculty of Behavioural and Movement Sciences, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Vittorio Sebastiano
- Department of Obstetrics and Gynecology, School of Medicine, Stanford University, Stanford, CA, USA
| | - Michael P Snyder
- Department of Genetics, School of Medicine, Stanford University, Stanford, CA, USA
| | - Vadim N Gladyshev
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| | | | | |
Collapse
|
16
|
Prosz A, Pipek O, Börcsök J, Palla G, Szallasi Z, Spisak S, Csabai I. Biologically informed deep learning for explainable epigenetic clocks. Sci Rep 2024; 14:1306. [PMID: 38225268 PMCID: PMC10789766 DOI: 10.1038/s41598-023-50495-5] [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: 01/17/2023] [Accepted: 12/20/2023] [Indexed: 01/17/2024] Open
Abstract
Ageing is often characterised by progressive accumulation of damage, and it is one of the most important risk factors for chronic disease development. Epigenetic mechanisms including DNA methylation could functionally contribute to organismal aging, however the key functions and biological processes may govern ageing are still not understood. Although age predictors called epigenetic clocks can accurately estimate the biological age of an individual based on cellular DNA methylation, their models have limited ability to explain the prediction algorithm behind and underlying key biological processes controlling ageing. Here we present XAI-AGE, a biologically informed, explainable deep neural network model for accurate biological age prediction across multiple tissue types. We show that XAI-AGE outperforms the first-generation age predictors and achieves similar results to deep learning-based models, while opening up the possibility to infer biologically meaningful insights of the activity of pathways and other abstract biological processes directly from the model.
Collapse
Affiliation(s)
- Aurel Prosz
- Danish Cancer Institute, Copenhagen, Denmark
| | - Orsolya Pipek
- Department of Physics of Complex Systems, ELTE Eötvös Loránd University, Budapest, Hungary
| | - Judit Börcsök
- Danish Cancer Institute, Copenhagen, Denmark
- Biotech Research & Innovation Centre (BRIC), University of Copenhagen, Copenhagen, Denmark
| | - Gergely Palla
- Department of Biological Physics, ELTE Eötvös Loránd University, Budapest, Hungary
- Health Services Management Training Centre, Semmelweis University, Budapest, Hungary
| | | | - Sandor Spisak
- Institute of Enzymology, HUN-REN Research Centre for Natural Sciences, Budapest, Hungary.
| | - István Csabai
- Department of Physics of Complex Systems, ELTE Eötvös Loránd University, Budapest, Hungary
| |
Collapse
|
17
|
Dutta S, Goodrich JM, Dolinoy DC, Ruden DM. Biological Aging Acceleration Due to Environmental Exposures: An Exciting New Direction in Toxicogenomics Research. Genes (Basel) 2023; 15:16. [PMID: 38275598 PMCID: PMC10815440 DOI: 10.3390/genes15010016] [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: 11/27/2023] [Revised: 12/14/2023] [Accepted: 12/20/2023] [Indexed: 01/27/2024] Open
Abstract
Biological clock technologies are designed to assess the acceleration of biological age (B-age) in diverse cell types, offering a distinctive opportunity in toxicogenomic research to explore the impact of environmental stressors, social challenges, and unhealthy lifestyles on health impairment. These clocks also play a role in identifying factors that can hinder aging and promote a healthy lifestyle. Over the past decade, researchers in epigenetics have developed testing methods that predict the chronological and biological age of organisms. These methods rely on assessing DNA methylation (DNAm) levels at specific CpG sites, RNA levels, and various biomolecules across multiple cell types, tissues, and entire organisms. Commonly known as 'biological clocks' (B-clocks), these estimators hold promise for gaining deeper insights into the pathways contributing to the development of age-related disorders. They also provide a foundation for devising biomedical or social interventions to prevent, reverse, or mitigate these disorders. This review article provides a concise overview of various epigenetic clocks and explores their susceptibility to environmental stressors.
Collapse
Affiliation(s)
- Sudipta Dutta
- Department of Veterinary Integrative Biosciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX 77843, USA;
| | - Jaclyn M. Goodrich
- Department of Environmental Health Sciences, School of Public Health, University of Michigan, Ann Arbor, MI 48109, USA; (J.M.G.); (D.C.D.)
| | - Dana C. Dolinoy
- Department of Environmental Health Sciences, School of Public Health, University of Michigan, Ann Arbor, MI 48109, USA; (J.M.G.); (D.C.D.)
- Department of Nutritional Sciences, School of Public Health, University of Michigan, Ann Arbor, MI 48109, USA
| | - Douglas M. Ruden
- C. S. Mott Center for Human Health and Development, Department of Obstetrics and Gynecology, Institute of Environmental Health Sciences, Wayne State University, Detroit, MI 48202, USA
| |
Collapse
|
18
|
Coelho-Júnior HJ, Calvani R, Picca A, Tosato M, Landi F, Marzetti E. Association of Physical Activity and Exercise with Physical Performance and Muscle Mass in Older Adults: Results from the Longevity Check-Up (Lookup) 7+ Project. J Clin Med 2023; 12:7521. [PMID: 38137590 PMCID: PMC10744185 DOI: 10.3390/jcm12247521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Revised: 11/28/2023] [Accepted: 12/02/2023] [Indexed: 12/24/2023] Open
Abstract
Regular engagement in physical activity (PA) or physical exercise (PE) is effective at improving physical performance and body composition in older adults. Less is known about the benefits that may be obtained through combining PA with PE and whether the effects of activity habits differ between men and women. This study cross-sectionally investigated the association of PA and/or PE with physical performance and anthropometric measures in a large and relatively unselected sample of older adults enrolled in the Longevity Check-up (Lookup) 7+ project. Participants were individuals 65 years and older living in the community who were recruited in unconventional settings across Italy. Adherence to PA or PE was operationalized as involvement in light walking or various types of exercise, respectively, at least twice weekly for a minimum of 30 min per session throughout the last 12 months. Physical performance measures included handgrip strength and five-time sit-to-stand (5STS) tests. Lower-limb muscle power and appendicular skeletal muscle mass (ASM) were estimated through validated equations. We analyzed data of 4119 participants, of whom 2222 (53.4%) were women. The mean age was 72.8 ± 5.8 years in men and 72.1 ± 5.4 years in women. Regular engagement in PA + PE was reported by 139 (7.3%) men and 100 (4.5%) women. Results indicated that regular walking activity and/or PE were significantly associated with better physical performance and greater ASM with sex-specific patterns. Associations were also influenced by the type of activity, physical performance assessment tool, and anthropometric parameters. Men engaged in PA + PE performed better on the 5STS test and had greater handgrip strength, ASM, and relative and specific muscle power than those practicing either PA or PE. In women, the combination of PA and PE was associated with greater handgrip strength. The findings of this study indicate that older adults regularly practicing PA + PE had better physical performance than those who only engaged in either modality. In men, the combination of PA and PE was also associated with greater ASM.
Collapse
Affiliation(s)
- Hélio José Coelho-Júnior
- Department of Geriatrics, Orthopedics and Rheumatology, Università Cattolica del Sacro Cuore, L.go F. Vito 1, 00168 Rome, Italy; (F.L.); (E.M.)
| | - Riccardo Calvani
- Department of Geriatrics, Orthopedics and Rheumatology, Università Cattolica del Sacro Cuore, L.go F. Vito 1, 00168 Rome, Italy; (F.L.); (E.M.)
- Fondazione Policlinico Universitario “A. Gemelli” IRCCS, Largo A. Gemelli 8, 00168 Rome, Italy; (A.P.); (M.T.)
| | - Anna Picca
- Fondazione Policlinico Universitario “A. Gemelli” IRCCS, Largo A. Gemelli 8, 00168 Rome, Italy; (A.P.); (M.T.)
- Department of Medicine and Surgery, LUM University, Str. Statale 100 km 18, 70100 Casamassima, Italy
| | - Matteo Tosato
- Fondazione Policlinico Universitario “A. Gemelli” IRCCS, Largo A. Gemelli 8, 00168 Rome, Italy; (A.P.); (M.T.)
| | - Francesco Landi
- Department of Geriatrics, Orthopedics and Rheumatology, Università Cattolica del Sacro Cuore, L.go F. Vito 1, 00168 Rome, Italy; (F.L.); (E.M.)
- Fondazione Policlinico Universitario “A. Gemelli” IRCCS, Largo A. Gemelli 8, 00168 Rome, Italy; (A.P.); (M.T.)
| | - Emanuele Marzetti
- Department of Geriatrics, Orthopedics and Rheumatology, Università Cattolica del Sacro Cuore, L.go F. Vito 1, 00168 Rome, Italy; (F.L.); (E.M.)
- Fondazione Policlinico Universitario “A. Gemelli” IRCCS, Largo A. Gemelli 8, 00168 Rome, Italy; (A.P.); (M.T.)
| |
Collapse
|
19
|
Jackson P, Kempf MC, Goodin BR, A. Hidalgo B, Aroke EN. Neighborhood Environment and Epigenetic Age: A Scoping Review. West J Nurs Res 2023; 45:1139-1149. [PMID: 37902222 PMCID: PMC10748459 DOI: 10.1177/01939459231208304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2023]
Abstract
BACKGROUND Interest in how the neighborhood environment impacts age-related health conditions has been increasing for decades. Epigenetic changes are environmentally derived modifications to the genome that alter the way genes function-thus altering health status. Epigenetic age, a biomarker for biological age, has been shown to be a useful predictor of several age-related health conditions. Consequently, its relation to the neighborhood environment has been the focus of a growing body of literature. OBJECTIVE We aimed to describe the scope of the evidence on the relationship between neighborhood environmental characteristics and epigenetic age. METHODS Using scoping review following methods established by Arksey and O'Malley, we first defined our research questions and searched the literature in PubMed, PsycINFO, and EMBASE. Next, we selected the literature to be included, and finally, we analyzed and summarized the information. RESULTS Nine articles met the inclusion criteria. Most studies examined deprivation as the neighborhood characteristic of interest. While all studies were observational in design, the articles included diverse participants, including men and women, adults and children, and multiple ethnicities. Results demonstrated a relationship between the neighborhood environment and epigenetic age, whether the characteristic of interest is socioeconomic or physical. CONCLUSIONS Overall, studies concluded there was a relationship between neighborhood characteristics and epigenetic age, whether the characteristic of interest was socioeconomic or physical. However, findings varied based on how the neighborhood characteristic and/or epigenetic age was measured. Furthermore, a paucity of investigations on physical characteristics was noticeable and warrants increased attention.
Collapse
Affiliation(s)
- Pamela Jackson
- School of Public Health, University of Alabama at Birmingham, Birmingham, AL, USA
| | | | - Burel R. Goodin
- Department of Anesthesiology, Washington University Pain Center, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
| | - Bertha A. Hidalgo
- School of Public Health, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Edwin N. Aroke
- School of Nursing, University of Alabama at Birmingham, Birmingham, AL, USA
| |
Collapse
|
20
|
Fernandez ME, Martinez-Romero J, Aon MA, Bernier M, Price NL, de Cabo R. How is Big Data reshaping preclinical aging research? Lab Anim (NY) 2023; 52:289-314. [PMID: 38017182 DOI: 10.1038/s41684-023-01286-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Accepted: 10/10/2023] [Indexed: 11/30/2023]
Abstract
The exponential scientific and technological progress during the past 30 years has favored the comprehensive characterization of aging processes with their multivariate nature, leading to the advent of Big Data in preclinical aging research. Spanning from molecular omics to organism-level deep phenotyping, Big Data demands large computational resources for storage and analysis, as well as new analytical tools and conceptual frameworks to gain novel insights leading to discovery. Systems biology has emerged as a paradigm that utilizes Big Data to gain insightful information enabling a better understanding of living organisms, visualized as multilayered networks of interacting molecules, cells, tissues and organs at different spatiotemporal scales. In this framework, where aging, health and disease represent emergent states from an evolving dynamic complex system, context given by, for example, strain, sex and feeding times, becomes paramount for defining the biological trajectory of an organism. Using bioinformatics and artificial intelligence, the systems biology approach is leading to remarkable advances in our understanding of the underlying mechanism of aging biology and assisting in creative experimental study designs in animal models. Future in-depth knowledge acquisition will depend on the ability to fully integrate information from different spatiotemporal scales in organisms, which will probably require the adoption of theories and methods from the field of complex systems. Here we review state-of-the-art approaches in preclinical research, with a focus on rodent models, that are leading to conceptual and/or technical advances in leveraging Big Data to understand basic aging biology and its full translational potential.
Collapse
Affiliation(s)
- Maria Emilia Fernandez
- Experimental Gerontology Section, Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Jorge Martinez-Romero
- Experimental Gerontology Section, Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
- Laboratory of Epidemiology and Population Science, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Miguel A Aon
- Experimental Gerontology Section, Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
- Laboratory of Cardiovascular Science, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Michel Bernier
- Experimental Gerontology Section, Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Nathan L Price
- Experimental Gerontology Section, Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Rafael de Cabo
- Experimental Gerontology Section, Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA.
| |
Collapse
|
21
|
Zhang Q. An interpretable biological age. THE LANCET. HEALTHY LONGEVITY 2023; 4:e662-e663. [PMID: 37944548 DOI: 10.1016/s2666-7568(23)00213-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Accepted: 10/10/2023] [Indexed: 11/12/2023] Open
|
22
|
Bortz J, Guariglia A, Klaric L, Tang D, Ward P, Geer M, Chadeau-Hyam M, Vuckovic D, Joshi PK. Biological age estimation using circulating blood biomarkers. Commun Biol 2023; 6:1089. [PMID: 37884697 PMCID: PMC10603148 DOI: 10.1038/s42003-023-05456-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Accepted: 10/13/2023] [Indexed: 10/28/2023] Open
Abstract
Biological age captures physiological deterioration better than chronological age and is amenable to interventions. Blood-based biomarkers have been identified as suitable candidates for biological age estimation. This study aims to improve biological age estimation using machine learning models and a feature-set of 60 circulating biomarkers available from the UK Biobank (n = 306,116). We implement an Elastic-Net derived Cox model with 25 selected biomarkers to predict mortality risk (C-Index = 0.778; 95% CI [0.767-0.788]), which outperforms the well-known blood-biomarker based PhenoAge model (C-Index = 0.750; 95% CI [0.739-0.761]), providing a C-Index lift of 0.028 representing an 11% relative increase in predictive value. Importantly, we then show that using common clinical assay panels, with few biomarkers, alongside imputation and the model derived on the full set of biomarkers, does not substantially degrade predictive accuracy from the theoretical maximum achievable for the available biomarkers. Biological age is estimated as the equivalent age within the same-sex population which corresponds to an individual's mortality risk. Values ranged between 20-years younger and 20-years older than individuals' chronological age, exposing the magnitude of ageing signals contained in blood markers. Thus, we demonstrate a practical and cost-efficient method of estimating an improved measure of Biological Age, available to the general population.
Collapse
Affiliation(s)
- Jordan Bortz
- Humanity Inc, Humanity, 177 Huntington Ave, Ste 1700, Humanity Inc - 91556, Boston, MA, 02115, USA.
- Department of Epidemiology and Biostatistics, School of Public Health, Faculty of Medicine, Imperial College London, London, UK.
| | - Andrea Guariglia
- Humanity Inc, Humanity, 177 Huntington Ave, Ste 1700, Humanity Inc - 91556, Boston, MA, 02115, USA
- Department of Epidemiology and Biostatistics, School of Public Health, Faculty of Medicine, Imperial College London, London, UK
| | - Lucija Klaric
- Humanity Inc, Humanity, 177 Huntington Ave, Ste 1700, Humanity Inc - 91556, Boston, MA, 02115, USA
| | - David Tang
- Department of Epidemiology and Biostatistics, School of Public Health, Faculty of Medicine, Imperial College London, London, UK
| | - Peter Ward
- Humanity Inc, Humanity, 177 Huntington Ave, Ste 1700, Humanity Inc - 91556, Boston, MA, 02115, USA
| | - Michael Geer
- Humanity Inc, Humanity, 177 Huntington Ave, Ste 1700, Humanity Inc - 91556, Boston, MA, 02115, USA
| | - Marc Chadeau-Hyam
- Department of Epidemiology and Biostatistics, School of Public Health, Faculty of Medicine, Imperial College London, London, UK
- MRC Centre for Environment and Health, School of Public Health, Imperial College London, London, UK
- NIHR-HPRU, Health Protection Research Unit in Chemical and Radiation Threats and Hazards, Public Health England and Imperial College London, London, UK
| | - Dragana Vuckovic
- Department of Epidemiology and Biostatistics, School of Public Health, Faculty of Medicine, Imperial College London, London, UK.
- NIHR-HPRU, Health Protection Research Unit in Chemical and Radiation Threats and Hazards, Public Health England and Imperial College London, London, UK.
| | - Peter K Joshi
- Humanity Inc, Humanity, 177 Huntington Ave, Ste 1700, Humanity Inc - 91556, Boston, MA, 02115, USA.
- Centre for Global Health Research, Usher Institute, University of Edinburgh, Edinburgh, UK.
| |
Collapse
|
23
|
Bousquet A, Sanderson K, O’Shea TM, Fry RC. Accelerated Aging and the Life Course of Individuals Born Preterm. CHILDREN (BASEL, SWITZERLAND) 2023; 10:1683. [PMID: 37892346 PMCID: PMC10605448 DOI: 10.3390/children10101683] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/02/2023] [Revised: 09/29/2023] [Accepted: 10/11/2023] [Indexed: 10/29/2023]
Abstract
Individuals born preterm have shorter lifespans and elevated rates of chronic illness that contribute to mortality risk when compared to individuals born at term. Emerging evidence suggests that individuals born preterm or of low birthweight also exhibit physiologic and cellular biomarkers of accelerated aging. It is unclear whether, and to what extent, accelerated aging contributes to a higher risk of chronic illness and mortality among individuals born preterm. Here, we review accelerated aging phenotypes in adults born preterm and biological pathways that appear to contribute to accelerated aging. We highlight biomarkers of accelerated aging and various resiliency factors, including both pharmacologic and non-pharmacologic factors, that might buffer the propensity for accelerated aging among individuals born preterm.
Collapse
Affiliation(s)
- Audrey Bousquet
- Department of Environmental Sciences and Engineering, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC 27599, USA; (A.B.); (R.C.F.)
| | - Keia Sanderson
- Department of Internal Medicine, School of Medicine, University of North Carolina, Chapel Hill, NC 27599, USA;
| | - T. Michael O’Shea
- Department of Pediatrics, School of Medicine, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Rebecca C. Fry
- Department of Environmental Sciences and Engineering, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC 27599, USA; (A.B.); (R.C.F.)
| |
Collapse
|
24
|
Kraus M, Stumpf UC, Keppler AM, Neuerburg C, Böcker W, Wackerhage H, Baumbach SF, Saller MM. Development of a Machine Learning-Based Model to Predict Timed-Up-and-Go Test in Older Adults. Geriatrics (Basel) 2023; 8:99. [PMID: 37887972 PMCID: PMC10606325 DOI: 10.3390/geriatrics8050099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 09/29/2023] [Accepted: 10/05/2023] [Indexed: 10/28/2023] Open
Abstract
INTRODUCTION The measurement of physical frailty in elderly patients with orthopedic impairments remains a challenge due to its subjectivity, unreliability, time-consuming nature, and limited applicability to uninjured individuals. Our study aims to address this gap by developing objective, multifactorial machine models that do not rely on mobility data and subsequently validating their predictive capacity concerning the Timed-up-and-Go test (TUG test) in orthogeriatric patients. METHODS We utilized 67 multifactorial non-mobility parameters in a pre-processing phase, employing six feature selection algorithms. Subsequently, these parameters were used to train four distinct machine learning algorithms, including a generalized linear model, a support vector machine, a random forest algorithm, and an extreme gradient boost algorithm. The primary goal was to predict the time required for the TUG test without relying on mobility data. RESULTS The random forest algorithm yielded the most accurate estimations of the TUG test time. The best-performing algorithm demonstrated a mean absolute error of 2.7 s, while the worst-performing algorithm exhibited an error of 7.8 s. The methodology used for variable selection appeared to exert minimal influence on the overall performance. It is essential to highlight that all the employed algorithms tended to overestimate the time for quick patients and underestimate it for slower patients. CONCLUSION Our findings demonstrate the feasibility of predicting the TUG test time using a machine learning model that does not depend on mobility data. This establishes a basis for identifying patients at risk automatically and objectively assessing the physical capacity of currently immobilized patients. Such advancements could significantly contribute to enhancing patient care and treatment planning in orthogeriatric settings.
Collapse
Affiliation(s)
- Moritz Kraus
- Department of Orthopedics and Trauma Surgery, Musculoskeletal University Center Munich, University Hospital of Ludwig-Maximilians-University (LMU), 81377 Munich, Germany; (U.C.S.); (A.M.K.); (C.N.); (W.B.); (S.F.B.); (M.M.S.)
| | - Ulla Cordula Stumpf
- Department of Orthopedics and Trauma Surgery, Musculoskeletal University Center Munich, University Hospital of Ludwig-Maximilians-University (LMU), 81377 Munich, Germany; (U.C.S.); (A.M.K.); (C.N.); (W.B.); (S.F.B.); (M.M.S.)
| | - Alexander Martin Keppler
- Department of Orthopedics and Trauma Surgery, Musculoskeletal University Center Munich, University Hospital of Ludwig-Maximilians-University (LMU), 81377 Munich, Germany; (U.C.S.); (A.M.K.); (C.N.); (W.B.); (S.F.B.); (M.M.S.)
| | - Carl Neuerburg
- Department of Orthopedics and Trauma Surgery, Musculoskeletal University Center Munich, University Hospital of Ludwig-Maximilians-University (LMU), 81377 Munich, Germany; (U.C.S.); (A.M.K.); (C.N.); (W.B.); (S.F.B.); (M.M.S.)
| | - Wolfgang Böcker
- Department of Orthopedics and Trauma Surgery, Musculoskeletal University Center Munich, University Hospital of Ludwig-Maximilians-University (LMU), 81377 Munich, Germany; (U.C.S.); (A.M.K.); (C.N.); (W.B.); (S.F.B.); (M.M.S.)
| | - Henning Wackerhage
- Faculty of Sport and Health Sciences, Technical University of Munich, 80809 Munich, Germany;
| | - Sebastian Felix Baumbach
- Department of Orthopedics and Trauma Surgery, Musculoskeletal University Center Munich, University Hospital of Ludwig-Maximilians-University (LMU), 81377 Munich, Germany; (U.C.S.); (A.M.K.); (C.N.); (W.B.); (S.F.B.); (M.M.S.)
| | - Maximilian Michael Saller
- Department of Orthopedics and Trauma Surgery, Musculoskeletal University Center Munich, University Hospital of Ludwig-Maximilians-University (LMU), 81377 Munich, Germany; (U.C.S.); (A.M.K.); (C.N.); (W.B.); (S.F.B.); (M.M.S.)
| |
Collapse
|
25
|
Wang S, Rao Z, Cao R, Blaes AH, Coresh J, Joshu CE, Lehallier B, Lutsey PL, Pankow JS, Sedaghat S, Tang W, Thyagarajan B, Walker KA, Ganz P, Platz EA, Guan W, Prizment A. Development and Characterization of Proteomic Aging Clocks in the Atherosclerosis Risk in Communities (ARIC) Study. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.09.06.23295174. [PMID: 37732184 PMCID: PMC10508816 DOI: 10.1101/2023.09.06.23295174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/22/2023]
Abstract
Biological age may be estimated by proteomic aging clocks (PACs). Previous published PACs were constructed either in smaller studies or mainly in White individuals, and they used proteomic measures from only one-time point. In the Atherosclerosis Risk in Communities (ARIC) study of about 12,000 persons followed for 30 years (around 75% White, 25% Black), we created de novo PACs and compared their performance to published PACs at two different time points. We measured 4,712 plasma proteins by SomaScan in 11,761 midlife participants, aged 46-70 years (1990-92), and 5,183 late-life pariticpants, aged 66-90 years (2011-13). All proteins were log2-transformed to correct for skewness. We created de novo PACs by training them against chronological age using elastic net regression in two-thirds of healthy participants in midlife and late life and compared their performance to three published PACs. We estimated age acceleration (by regressing each PAC on chronological age) and its change from midlife to late life. We examined their associations with mortality from all-cause, cardiovascular disease (CVD), cancer, and lower respiratory disease (LRD) using Cox proportional hazards regression in all remaining participants irrespective of health. The model was adjusted for chronological age, smoking, body mass index (BMI), and other confounders. The ARIC PACs had a slightly stronger correlation with chronological age than published PACs in healthy participants at each time point. Associations with mortality were similar for the ARIC and published PACs. For late-life and midlife age acceleration for the ARIC PACs, respectively, hazard ratios (HRs) per one standard deviation were 1.65 and 1.38 (both p<0.001) for all-cause mortality, 1.37 and 1.20 (both p<0.001) for CVD mortality, 1.21 (p=0.03) and 1.04 (p=0.19) for cancer mortality, and 1.46 and 1.68 (both p<0.001) for LRD mortality. For the change in age acceleration, HRs for all-cause, CVD, and LRD mortality were comparable to those observed for late-life age acceleration. The association between the change in age acceleration and cancer mortality was insignificant. In this prospective study, the ARIC and published PACs were similarly associated with an increased risk of mortality and advanced testing in relation to various age-related conditions in future studies is suggested.
Collapse
Affiliation(s)
- Shuo Wang
- Department of Laboratory Medicine and Pathology, Medical School, University of Minnesota, Minneapolis, MN
| | - Zexi Rao
- Division of Biostatistics, School of Public Health, University of Minnesota, Minneapolis, MN
| | - Rui Cao
- Division of Biostatistics, School of Public Health, University of Minnesota, Minneapolis, MN
| | - Anne H. Blaes
- Division of Hematology, Oncology and Transplantation, Medical School, University of Minnesota, Minneapolis, MN
| | - Josef Coresh
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
| | - Corinne E. Joshu
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
- Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD
| | - Benoit Lehallier
- Alkahest Inc, San Carlos, CA, United States, Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA
| | - Pamela L. Lutsey
- Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, Minneapolis, MN
| | - James S. Pankow
- Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, Minneapolis, MN
| | - Sanaz Sedaghat
- Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, Minneapolis, MN
| | - Weihong Tang
- Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, Minneapolis, MN
| | - Bharat Thyagarajan
- Department of Laboratory Medicine and Pathology, Medical School, University of Minnesota, Minneapolis, MN
| | - Keenan A. Walker
- Laboratory of Behavioral Neuroscience, Intramural Research Program, National Institute on Aging, Baltimore, MD
| | - Peter Ganz
- Division of Cardiology, Zuckerberg San Francisco General Hospital and Department of Medicine, University of California, San Francisco, CA
| | - Elizabeth A. Platz
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
- Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD
| | - Weihua Guan
- Division of Biostatistics, School of Public Health, University of Minnesota, Minneapolis, MN
| | - Anna Prizment
- Department of Laboratory Medicine and Pathology, Medical School, University of Minnesota, Minneapolis, MN
| |
Collapse
|
26
|
Zipple MN, Vogt CC, Sheehan MJ. Re-wilding model organisms: Opportunities to test causal mechanisms in social determinants of health and aging. Neurosci Biobehav Rev 2023; 152:105238. [PMID: 37225063 PMCID: PMC10527394 DOI: 10.1016/j.neubiorev.2023.105238] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 04/14/2023] [Accepted: 05/17/2023] [Indexed: 05/26/2023]
Abstract
Social experiences are strongly associated with individuals' health, aging, and survival in many mammalian taxa, including humans. Despite their role as models of many other physiological and developmental bases of health and aging, biomedical model organisms (particularly lab mice) remain an underutilized tool in resolving outstanding questions regarding social determinants of health and aging, including causality, context-dependence, reversibility, and effective interventions. This status is largely due to the constraints of standard laboratory conditions on animals' social lives. Even when kept in social housing, lab animals rarely experience social and physical environments that approach the richness, variability, and complexity they have evolved to navigate and benefit from. Here we argue that studying biomedical model organisms outside under complex, semi-natural social environments ("re-wilding") allows researchers to capture the methodological benefits of both field studies of wild animals and laboratory studies of model organisms. We review recent efforts to re-wild mice and highlight discoveries that have only been made possible by researchers studying mice under complex, manipulable social environments.
Collapse
Affiliation(s)
- Matthew N Zipple
- Laboratory for Animal Social Evolution and Recognition, Department of Neurobiology and Behavior, Cornell University, Ithaca, NY, USA.
| | - Caleb C Vogt
- Laboratory for Animal Social Evolution and Recognition, Department of Neurobiology and Behavior, Cornell University, Ithaca, NY, USA
| | - Michael J Sheehan
- Laboratory for Animal Social Evolution and Recognition, Department of Neurobiology and Behavior, Cornell University, Ithaca, NY, USA.
| |
Collapse
|
27
|
Moqri M, Herzog C, Poganik JR, Justice J, Belsky DW, Higgins-Chen A, Moskalev A, Fuellen G, Cohen AA, Bautmans I, Widschwendter M, Ding J, Fleming A, Mannick J, Han JDJ, Zhavoronkov A, Barzilai N, Kaeberlein M, Cummings S, Kennedy BK, Ferrucci L, Horvath S, Verdin E, Maier AB, Snyder MP, Sebastiano V, Gladyshev VN. Biomarkers of aging for the identification and evaluation of longevity interventions. Cell 2023; 186:3758-3775. [PMID: 37657418 PMCID: PMC11088934 DOI: 10.1016/j.cell.2023.08.003] [Citation(s) in RCA: 124] [Impact Index Per Article: 124.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 08/01/2023] [Accepted: 08/02/2023] [Indexed: 09/03/2023]
Abstract
With the rapid expansion of aging biology research, the identification and evaluation of longevity interventions in humans have become key goals of this field. Biomarkers of aging are critically important tools in achieving these objectives over realistic time frames. However, the current lack of standards and consensus on the properties of a reliable aging biomarker hinders their further development and validation for clinical applications. Here, we advance a framework for the terminology and characterization of biomarkers of aging, including classification and potential clinical use cases. We discuss validation steps and highlight ongoing challenges as potential areas in need of future research. This framework sets the stage for the development of valid biomarkers of aging and their ultimate utilization in clinical trials and practice.
Collapse
Affiliation(s)
- Mahdi Moqri
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA; Department of Genetics, School of Medicine, Stanford University, Stanford, CA, USA; Department of Obstetrics and Gynecology, School of Medicine, Stanford University, Stanford, CA, USA
| | - Chiara Herzog
- European Translational Oncology Prevention and Screening Institute, Universität Innsbruck, Innsbruck, Austria
| | - Jesse R Poganik
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Jamie Justice
- Department of Internal Medicine, Section on Gerontology and Geriatric Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Daniel W Belsky
- Department of Epidemiology, Butler Columbia Aging Center, Mailman School of Public Health, Columbia University, New York, NY, USA
| | | | - Alexey Moskalev
- Institute of Biogerontology, Lobachevsky University, Nizhny Novgorod, Russia
| | - Georg Fuellen
- Institute for Biostatistics and Informatics in Medicine and Ageing Research, Rostock University Medical Center, Rostock, Germany; School of Medicine, University College Dublin, Dublin, Ireland
| | - Alan A Cohen
- Department of Environmental Health Sciences, Butler Columbia Aging Center, Mailman School of Public Health, Columbia University, New York, NY, USA
| | - Ivan Bautmans
- Gerontology Department, Vrije Universiteit Brussel, Brussels, Belgium; Frailty in Ageing Research Department, Vrije Universiteit Brussel, Brussels, Belgium
| | - Martin Widschwendter
- European Translational Oncology Prevention and Screening Institute, Universität Innsbruck, Innsbruck, Austria; Department of Women's Cancer, EGA Institute for Women's Health, University College London, London, UK; Department of Women's and Children's Health, Division of Obstetrics and Gynaecology, Karolinska Institutet, Stockholm, Sweden
| | - Jingzhong Ding
- Gerontology and Geriatric Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | | | | | - Jing-Dong Jackie Han
- Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Center for Quantitative Biology, Peking University, Beijing, China
| | - Alex Zhavoronkov
- Insilico Medicine Hong Kong, Pak Shek Kok, New Territories, Hong Kong SAR, China
| | - Nir Barzilai
- Institute for Aging Research, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Matt Kaeberlein
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
| | - Steven Cummings
- San Francisco Coordinating Center, California Pacific Medical Center Research Institute, San Francisco, CA, USA; Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, CA, USA
| | - Brian K Kennedy
- Healthy Longevity Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | | | | | - Eric Verdin
- Buck Institute for Research on Aging, Novato, CA, USA
| | - Andrea B Maier
- Department of Human Movement Sciences, @AgeAmsterdam, Amsterdam Movement Sciences, Faculty of Behavioural and Movement Sciences, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands; Healthy Longevity Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Centre for Healthy Longevity, @AgeSingapore, National University Health System, Singapore, Singapore
| | - Michael P Snyder
- Department of Genetics, School of Medicine, Stanford University, Stanford, CA, USA.
| | - Vittorio Sebastiano
- Department of Obstetrics and Gynecology, School of Medicine, Stanford University, Stanford, CA, USA.
| | - Vadim N Gladyshev
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
28
|
Simpson DJ, Zhao Q, Olova NN, Dabrowski J, Xie X, Latorre‐Crespo E, Chandra T. Region-based epigenetic clock design improves RRBS-based age prediction. Aging Cell 2023; 22:e13866. [PMID: 37170475 PMCID: PMC10410054 DOI: 10.1111/acel.13866] [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: 02/08/2023] [Revised: 04/20/2023] [Accepted: 04/21/2023] [Indexed: 05/13/2023] Open
Abstract
Recent studies suggest that epigenetic rejuvenation can be achieved using drugs that mimic calorie restriction and techniques such as reprogramming-induced rejuvenation. To effectively test rejuvenation in vivo, mouse models are the safest alternative. However, we have found that the recent epigenetic clocks developed for mouse reduced-representation bisulphite sequencing (RRBS) data have significantly poor performance when applied to external datasets. We show that the sites captured and the coverage of key CpGs required for age prediction vary greatly between datasets, which likely contributes to the lack of transferability in RRBS clocks. To mitigate these coverage issues in RRBS-based age prediction, we present two novel design strategies that use average methylation over large regions rather than individual CpGs, whereby regions are defined by sliding windows (e.g. 5 kb), or density-based clustering of CpGs. We observe improved correlation and error in our regional blood clocks (RegBCs) compared to published individual-CpG-based techniques when applied to external datasets. The RegBCs are also more robust when applied to low coverage data and detect a negative age acceleration in mice undergoing calorie restriction. Our RegBCs offer a proof of principle that age prediction of RRBS datasets can be improved by accounting for multiple CpGs over a region, which negates the lack of read depth currently hindering individual-CpG-based approaches.
Collapse
Affiliation(s)
- Daniel J. Simpson
- MRC Human Genetics Unit, MRC Institute of Genetics and CancerUniversity of EdinburghEdinburghUK
| | - Qian Zhao
- MRC Human Genetics Unit, MRC Institute of Genetics and CancerUniversity of EdinburghEdinburghUK
| | - Nelly N. Olova
- MRC Human Genetics Unit, MRC Institute of Genetics and CancerUniversity of EdinburghEdinburghUK
| | - Jan Dabrowski
- MRC Human Genetics Unit, MRC Institute of Genetics and CancerUniversity of EdinburghEdinburghUK
| | - Xiaoxiao Xie
- MRC Human Genetics Unit, MRC Institute of Genetics and CancerUniversity of EdinburghEdinburghUK
| | - Eric Latorre‐Crespo
- MRC Human Genetics Unit, MRC Institute of Genetics and CancerUniversity of EdinburghEdinburghUK
| | - Tamir Chandra
- MRC Human Genetics Unit, MRC Institute of Genetics and CancerUniversity of EdinburghEdinburghUK
| |
Collapse
|
29
|
Bafei SEC, Shen C. Biomarkers selection and mathematical modeling in biological age estimation. NPJ AGING 2023; 9:13. [PMID: 37393295 DOI: 10.1038/s41514-023-00110-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/20/2022] [Accepted: 05/08/2023] [Indexed: 07/03/2023]
Abstract
Biological age (BA) is important for clinical monitoring and preventing aging-related disorders and disabilities. Clinical and/or cellular biomarkers are measured and integrated in years using mathematical models to display an individual's BA. To date, there is not yet a single or set of biomarker(s) and technique(s) that is validated as providing the BA that reflects the best real aging status of individuals. Herein, a comprehensive overview of aging biomarkers is provided and the potential of genetic variations as proxy indicators of the aging state is highlighted. A comprehensive overview of BA estimation methods is also provided as well as a discussion of their performances, advantages, limitations, and potential approaches to overcome these limitations.
Collapse
Affiliation(s)
- Solim Essomandan Clémence Bafei
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, 211166, Jiangsu, China
| | - Chong Shen
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, 211166, Jiangsu, China.
| |
Collapse
|
30
|
Shim J, Fleisch E, Barata F. Wearable-based accelerometer activity profile as digital biomarker of inflammation, biological age, and mortality using hierarchical clustering analysis in NHANES 2011-2014. Sci Rep 2023; 13:9326. [PMID: 37291134 PMCID: PMC10250365 DOI: 10.1038/s41598-023-36062-y] [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: 02/09/2023] [Accepted: 05/29/2023] [Indexed: 06/10/2023] Open
Abstract
Repeated disruptions in circadian rhythms are associated with implications for health outcomes and longevity. The utilization of wearable devices in quantifying circadian rhythm to elucidate its connection to longevity, through continuously collected data remains largely unstudied. In this work, we investigate a data-driven segmentation of the 24-h accelerometer activity profiles from wearables as a novel digital biomarker for longevity in 7,297 U.S. adults from the 2011-2014 National Health and Nutrition Examination Survey. Using hierarchical clustering, we identified five clusters and described them as follows: "High activity", "Low activity", "Mild circadian rhythm (CR) disruption", "Severe CR disruption", and "Very low activity". Young adults with extreme CR disturbance are seemingly healthy with few comorbid conditions, but in fact associated with higher white blood cell, neutrophils, and lymphocyte counts (0.05-0.07 log-unit, all p < 0.05) and accelerated biological aging (1.42 years, p < 0.001). Older adults with CR disruption are significantly associated with increased systemic inflammation indexes (0.09-0.12 log-unit, all p < 0.05), biological aging advance (1.28 years, p = 0.021), and all-cause mortality risk (HR = 1.58, p = 0.042). Our findings highlight the importance of circadian alignment on longevity across all ages and suggest that data from wearable accelerometers can help in identifying at-risk populations and personalize treatments for healthier aging.
Collapse
Affiliation(s)
- Jinjoo Shim
- Centre for Digital Health Interventions, Department of Management, Technology, and Economics, ETH Zurich, Zurich, Switzerland.
| | - Elgar Fleisch
- Centre for Digital Health Interventions, Department of Management, Technology, and Economics, ETH Zurich, Zurich, Switzerland
- Centre for Digital Health Interventions, Institute of Technology Management, University of St. Gallen, St. Gallen, Switzerland
| | - Filipe Barata
- Centre for Digital Health Interventions, Department of Management, Technology, and Economics, ETH Zurich, Zurich, Switzerland
| |
Collapse
|
31
|
Gao C, Amador C, Walker RM, Campbell A, Madden RA, Adams MJ, Bai X, Liu Y, Li M, Hayward C, Porteous DJ, Shen X, Evans KL, Haley CS, McIntosh AM, Navarro P, Zeng Y. Phenome-wide analyses identify an association between the parent-of-origin effects dependent methylome and the rate of aging in humans. Genome Biol 2023; 24:117. [PMID: 37189164 PMCID: PMC10184337 DOI: 10.1186/s13059-023-02953-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Accepted: 04/26/2023] [Indexed: 05/17/2023] Open
Abstract
BACKGROUND The variation in the rate at which humans age may be rooted in early events acting through the genomic regions that are influenced by such events and subsequently are related to health phenotypes in later life. The parent-of-origin-effect (POE)-regulated methylome includes regions enriched for genetically controlled imprinting effects (the typical type of POE) and regions influenced by environmental effects associated with parents (the atypical POE). This part of the methylome is heavily influenced by early events, making it a potential route connecting early exposures, the epigenome, and aging. We aim to test the association of POE-CpGs with early and later exposures and subsequently with health-related phenotypes and adult aging. RESULTS We perform a phenome-wide association analysis for the POE-influenced methylome using GS:SFHS (Ndiscovery = 5087, Nreplication = 4450). We identify and replicate 92 POE-CpG-phenotype associations. Most of the associations are contributed by the POE-CpGs belonging to the atypical class where the most strongly enriched associations are with aging (DNAmTL acceleration), intelligence, and parental (maternal) smoking exposure phenotypes. A proportion of the atypical POE-CpGs form co-methylation networks (modules) which are associated with these phenotypes, with one of the aging-associated modules displaying increased within-module methylation connectivity with age. The atypical POE-CpGs also display high levels of methylation heterogeneity, fast information loss with age, and a strong correlation with CpGs contained within epigenetic clocks. CONCLUSIONS These results identify the association between the atypical POE-influenced methylome and aging and provide new evidence for the "early development of origin" hypothesis for aging in humans.
Collapse
Affiliation(s)
- Chenhao Gao
- Faculty of Forensic Medicine, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
| | - Carmen Amador
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
| | - Rosie M Walker
- Centre for Clinical Brain Sciences, Chancellor's Building, 49 Little France Crescent, Edinburgh BioQuarter, Edinburgh, UK
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
- School of Psychology, University of Exeter, Perry Road, Exeter, UK
| | - Archie Campbell
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
| | | | - Mark J Adams
- Division of Psychiatry, University of Edinburgh, Edinburgh, UK
| | - Xiaomeng Bai
- Faculty of Forensic Medicine, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
| | - Ying Liu
- Faculty of Forensic Medicine, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
| | - Miaoxin Li
- Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
| | - Caroline Hayward
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
| | - David J Porteous
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
| | - Xueyi Shen
- Division of Psychiatry, University of Edinburgh, Edinburgh, UK
| | - Kathryn L Evans
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
| | - Chris S Haley
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
- Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, UK
| | | | - Pau Navarro
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK.
- Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, UK.
| | - Yanni Zeng
- Faculty of Forensic Medicine, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China.
- Guangdong Province Translational Forensic Medicine Engineering Technology Research Center, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China.
- Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China.
| |
Collapse
|
32
|
Lee EE, Adamowicz DH, Frangou S. An NIMH Workshop on Non-Affective Psychosis in Midlife and Beyond: Research Agenda on Phenomenology, Clinical Trajectories, Underlying Mechanisms, and Intervention Targets. Am J Geriatr Psychiatry 2023; 31:353-365. [PMID: 36858928 PMCID: PMC10990076 DOI: 10.1016/j.jagp.2023.01.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 01/05/2023] [Accepted: 01/23/2023] [Indexed: 02/05/2023]
Abstract
We present a review of the state of the research in the phenomenology, clinical trajectories, biological mechanisms, aging biomarkers, and treatments for middle-aged and older people with schizophrenia (PwS) discussed at the NIMH sponsored workshop "Non-affective Psychosis in Midlife and Beyond." The growing population of PwS has specific clinical needs that require tailored and mechanistically derived interventions. Differentiating between the effects of aging and disease progression is a key challenge of studying older PwS. This review of the workshop highlights the recent findings in this understudied clinical population and the critical gaps in knowledge and consensus for research priorities. This review showcases the major challenges and opportunities for research to advance clinical care for this growing and understudied population.
Collapse
Affiliation(s)
- Ellen E Lee
- Department of Psychiatry (EEL, DA), University of California San Diego, La Jolla, CA; Sam and Rose Stein Institute for Research on Aging (EEL, DA), University of California San Diego, La Jolla, CA; Desert-Pacific Mental Illness Research Education and Clinical Center, Veterans Affairs San Diego Healthcare System (EEL), San Diego, CA.
| | - David H Adamowicz
- Department of Psychiatry (EEL, DA), University of California San Diego, La Jolla, CA; Sam and Rose Stein Institute for Research on Aging (EEL, DA), University of California San Diego, La Jolla, CA
| | - Sophia Frangou
- Department of Psychiatry (SF), University of British Columbia, Vancouver, British Columbia, Canada; Icahn School of Medicine at Mount Sinai (SF), New York, NY
| |
Collapse
|
33
|
Adeodato CSR, Soares-Lima SC, Batista PV, Fagundes MCN, Camuzi D, Tavares SJO, Pinto LFR, Scelza MFZ. Interleukin 6 and Interleukin 1β hypomethylation and overexpression are common features of apical periodontitis: a case-control study with gingival tissue as control. Arch Oral Biol 2023; 150:105694. [PMID: 37043986 DOI: 10.1016/j.archoralbio.2023.105694] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 03/20/2023] [Accepted: 04/04/2023] [Indexed: 04/08/2023]
Abstract
OBJECTIVES Apical periodontitis is a periradicular tissue disorder that usually arises from infection by microorganisms in the root canal system resulting in local bone resorption. This usually involves the dysregulation of inflammatory mediators, which can be mediated by epigenetic mechanisms. Thus, the objective of this study was to evaluate Interleukin 6 (IL6) and Interleukin 1β (IL1β) and DNA methylation and gene expression levels in apical periodontitis. METHODS Gene expression was analyzed in 60 participants using quantitative polymerase chain reaction, while the methylation levels of IL6 and IL1β promoters were analyzed in 72 patients using pyrosequencing. All statistical analyzes were performed using the GraphPad Prism software version 8.0. The p value was considered statistically significant when < 0.05. RESULTS A significantly higher IL6 and IL1β expression levels were observed in cases relative to controls (fold-changes of 27.4 and 11.43, respectively, and p < 0.0001). By comparing the same groups, lower promoter methylation levels were observed for both genes in cases (methylation percentage delta relative to controls of -24.57% and -16.02%, respectively, and p < 0.0001). A significant inverse correlation between gene expression and promoter methylation was observed for both IL6 (p = 0.0002) and IL1β (p = 0.001). Neither IL6 expression nor promoter methylation were significantly associated with cases' age, smoking history, alcohol consumption history or sex. For IL1β, alcoholic cases showed lower methylation level relative to non-alcoholic cases (p = 0.01), while females showed higher methylation levels relative to males (p = 0.03). CONCLUSIONS Our data suggest a role for DNA methylation in IL6 and IL1β upregulation in apical periodontitis.
Collapse
Affiliation(s)
- Caroline Sousa Ribeiro Adeodato
- Post-graduation Program in Dentistry of Fluminense Federal University (UFF), Mario Santos Braga Street, no 28, 24020-140 Niteroi, RJ, Brazil
| | - Sheila Coelho Soares-Lima
- Molecular Carcinogenesis Program of National Cancer Institute (INCA), André Cavalcante Street, no 37, 20231-050 Rio de Janeiro, Brazil
| | - Paula Vieira Batista
- Molecular Carcinogenesis Program of National Cancer Institute (INCA), André Cavalcante Street, no 37, 20231-050 Rio de Janeiro, Brazil
| | - Marina Chianello Nicolau Fagundes
- Molecular Carcinogenesis Program of National Cancer Institute (INCA), André Cavalcante Street, no 37, 20231-050 Rio de Janeiro, Brazil
| | - Diego Camuzi
- Molecular Carcinogenesis Program of National Cancer Institute (INCA), André Cavalcante Street, no 37, 20231-050 Rio de Janeiro, Brazil
| | - Sandro Junio Oliveira Tavares
- Post-graduation Program in Dentistry of Fluminense Federal University (UFF), Mario Santos Braga Street, no 28, 24020-140 Niteroi, RJ, Brazil
| | - Luis Felipe Ribeiro Pinto
- Molecular Carcinogenesis Program of National Cancer Institute (INCA), André Cavalcante Street, no 37, 20231-050 Rio de Janeiro, Brazil; Biochemistry Department, Biology Institute, State University of Rio de Janeiro, Boulevard 28 de Setembro, 87 - Vila Isabel, 20511-010 Rio de Janeiro, Brazil
| | - Miriam Fatima Zaccaro Scelza
- Endodontics Department, Faculty of Dentistry, Fluminense Federal University (UFF), Mario Santos Braga Street, no 28, 24020-140 Niteroi, RJ, Brazil.
| |
Collapse
|
34
|
Hallmarks and Biomarkers of Skin Senescence: An Updated Review of Skin Senotherapeutics. Antioxidants (Basel) 2023; 12:antiox12020444. [PMID: 36830002 PMCID: PMC9952625 DOI: 10.3390/antiox12020444] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2023] [Revised: 02/06/2023] [Accepted: 02/07/2023] [Indexed: 02/12/2023] Open
Abstract
Aging is a complex process characterized by an ongoing decline in physiological functions, leading to degenerative diseases and an increased probability of death. Cellular senescence has been typically considered as an anti-proliferative process; however, the chronic accumulation of senescent cells contributes to tissue dysfunction and aging. In this review, we discuss some of the most important hallmarks and biomarkers of cellular senescence with a special focus on skin biomarkers, reactive oxygen species (ROS), and senotherapeutic strategies to eliminate or prevent senescence. Although most of them are not exclusive to senescence, the expression of the senescence-associated beta-galactosidase (SA-β-gal) enzyme seems to be the most reliable biomarker for distinguishing senescent cells from those arrested in the cell cycle. The presence of a stable DNA damage response (DDR) and the accumulation of senescence-associated secretory phenotype (SASP) mediators and ROS are the most representative hallmarks for senescence. Senotherapeutics based on natural compounds such as quercetin, naringenin, and apigenin have shown promising results regarding SASP reduction. These compounds seem to prevent the accumulation of senescent cells, most likely through the inhibition of pro-survival signaling pathways. Although studies are still required to verify their short- and long-term effects, these therapies may be an effective strategy for skin aging.
Collapse
|
35
|
Fernández-Pérez I, Jiménez-Balado J, Lazcano U, Giralt-Steinhauer E, Rey Álvarez L, Cuadrado-Godia E, Rodríguez-Campello A, Macias-Gómez A, Suárez-Pérez A, Revert-Barberá A, Estragués-Gázquez I, Soriano-Tarraga C, Roquer J, Ois A, Jiménez-Conde J. Machine Learning Approximations to Predict Epigenetic Age Acceleration in Stroke Patients. Int J Mol Sci 2023; 24:ijms24032759. [PMID: 36769083 PMCID: PMC9917369 DOI: 10.3390/ijms24032759] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 01/23/2023] [Accepted: 01/27/2023] [Indexed: 02/04/2023] Open
Abstract
Age acceleration (Age-A) is a useful tool that is able to predict a broad range of health outcomes. It is necessary to determine DNA methylation levels to estimate it, and it is known that Age-A is influenced by environmental, lifestyle, and vascular risk factors (VRF). The aim of this study is to estimate the contribution of these easily measurable factors to Age-A in patients with cerebrovascular disease (CVD), using different machine learning (ML) approximations, and try to find a more accessible model able to predict Age-A. We studied a CVD cohort of 952 patients with information about VRF, lifestyle habits, and target organ damage. We estimated Age-A using Hannum's epigenetic clock, and trained six different models to predict Age-A: a conventional linear regression model, four ML models (elastic net regression (EN), K-Nearest neighbors, random forest, and support vector machine models), and one deep learning approximation (multilayer perceptron (MLP) model). The best-performing models were EN and MLP; although, the predictive capability was modest (R2 0.358 and 0.378, respectively). In conclusion, our results support the influence of these factors on Age-A; although, they were not enough to explain most of its variability.
Collapse
Affiliation(s)
- Isabel Fernández-Pérez
- Neurovascular Research Group, Department of Neurology, IMIM-Hospital del Mar (Institut Hospital del Mar d’Investigacions Mèdiques), 08003 Barcelona, Spain
| | - Joan Jiménez-Balado
- Neurovascular Research Group, Department of Neurology, IMIM-Hospital del Mar (Institut Hospital del Mar d’Investigacions Mèdiques), 08003 Barcelona, Spain
- Correspondence: (J.J.-B.); (J.J.-C.)
| | - Uxue Lazcano
- Unidad de Investigación AP-OSIs Guipúzcoa, 20014 Donostia, Spain
| | - Eva Giralt-Steinhauer
- Neurovascular Research Group, Department of Neurology, IMIM-Hospital del Mar (Institut Hospital del Mar d’Investigacions Mèdiques), 08003 Barcelona, Spain
| | - Lucía Rey Álvarez
- Neurovascular Research Group, Department of Neurology, IMIM-Hospital del Mar (Institut Hospital del Mar d’Investigacions Mèdiques), 08003 Barcelona, Spain
| | - Elisa Cuadrado-Godia
- Neurovascular Research Group, Department of Neurology, IMIM-Hospital del Mar (Institut Hospital del Mar d’Investigacions Mèdiques), 08003 Barcelona, Spain
- Medicine Department, DCEXS-Universitat Pompeu Fabra (UPF), 08002 Barcelona, Spain
| | - Ana Rodríguez-Campello
- Neurovascular Research Group, Department of Neurology, IMIM-Hospital del Mar (Institut Hospital del Mar d’Investigacions Mèdiques), 08003 Barcelona, Spain
- Medicine Department, DCEXS-Universitat Pompeu Fabra (UPF), 08002 Barcelona, Spain
| | - Adrià Macias-Gómez
- Neurovascular Research Group, Department of Neurology, IMIM-Hospital del Mar (Institut Hospital del Mar d’Investigacions Mèdiques), 08003 Barcelona, Spain
| | - Antoni Suárez-Pérez
- Neurovascular Research Group, Department of Neurology, IMIM-Hospital del Mar (Institut Hospital del Mar d’Investigacions Mèdiques), 08003 Barcelona, Spain
| | - Anna Revert-Barberá
- Neurovascular Research Group, Department of Neurology, IMIM-Hospital del Mar (Institut Hospital del Mar d’Investigacions Mèdiques), 08003 Barcelona, Spain
| | - Isabel Estragués-Gázquez
- Neurovascular Research Group, Department of Neurology, IMIM-Hospital del Mar (Institut Hospital del Mar d’Investigacions Mèdiques), 08003 Barcelona, Spain
| | - Carolina Soriano-Tarraga
- Department of Psychiatry, NeuroGenomics and Informatics, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Jaume Roquer
- Neurovascular Research Group, Department of Neurology, IMIM-Hospital del Mar (Institut Hospital del Mar d’Investigacions Mèdiques), 08003 Barcelona, Spain
- Medicine Department, DCEXS-Universitat Pompeu Fabra (UPF), 08002 Barcelona, Spain
| | - Angel Ois
- Neurovascular Research Group, Department of Neurology, IMIM-Hospital del Mar (Institut Hospital del Mar d’Investigacions Mèdiques), 08003 Barcelona, Spain
- Medicine Department, DCEXS-Universitat Pompeu Fabra (UPF), 08002 Barcelona, Spain
| | - Jordi Jiménez-Conde
- Neurovascular Research Group, Department of Neurology, IMIM-Hospital del Mar (Institut Hospital del Mar d’Investigacions Mèdiques), 08003 Barcelona, Spain
- Medicine Department, DCEXS-Universitat Pompeu Fabra (UPF), 08002 Barcelona, Spain
- Correspondence: (J.J.-B.); (J.J.-C.)
| |
Collapse
|
36
|
Gao C, Amador C, Walker RM, Campbell A, Madden RA, Adams MJ, Bai X, Liu Y, Li M, Hayward C, Porteous DJ, Shen X, Evans KL, Haley CS, McIntosh AM, Navarro P, Zeng Y. Phenome-wide analysis identifies parent-of-origin effects on the human methylome associated with changes in the rate of aging. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.18.524653. [PMID: 36711749 PMCID: PMC9882261 DOI: 10.1101/2023.01.18.524653] [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] [Indexed: 05/18/2023]
Abstract
Variation in the rate at which humans age may be rooted in early life events acting through genomic regions that are influenced by such events and subsequently are related to health phenotypes in later life. The parent-of-origin-effect (POE)-regulated methylome includes regions either enriched for genetically controlled imprinting effects (the typical type of POE) or atypical POE introduced by environmental effects associated with parents. This part of the methylome is heavily influenced by early life events, making it a potential route connecting early environmental exposures, the epigenome and the rate of aging. Here, we aim to test the association of POE-influenced methylation of CpG dinucleotides (POE-CpG sites) with early and later environmental exposures and subsequently with health-related phenotypes and adult aging phenotypes. We do this by performing phenome-wide association analyses of the POE-influenced methylome using a large family-based population cohort (GS:SFHS, Ndiscovery=5,087, Nreplication=4,450). At the single CpG level, 92 associations of POE-CpGs with phenotypic variation were identified and replicated. Most of the associations were contributed by POE-CpGs belonging to the atypical class and the most strongly enriched associations were with aging (DNAmTL acceleration), intelligence and parental (maternal) smoking exposure phenotypes. We further found that a proportion of the atypical-POE-CpGs formed co-methylation networks (modules) which are associated with these phenotypes, with one of the aging-associated modules displaying increased internal module connectivity (strength of methylation correlation across constituent CpGs) with age. Atypical POE-CpGs also displayed high levels of methylation heterogeneity and epigenetic drift (i.e. information loss with age) and a strong correlation with CpGs contained within epigenetic clocks. These results identified associations between the atypical-POE-influenced methylome and aging and provided new evidence for the "early development of origin" hypothesis for aging in humans.
Collapse
Affiliation(s)
- Chenhao Gao
- Faculty of Forensic Medicine, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou 510080, China
| | - Carmen Amador
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
| | - Rosie M. Walker
- Centre for Clinical Brain Sciences, Chancellor’s Building, 49 Little France Crescent, Edinburgh BioQuarter, Edinburgh, UK
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
- School of Psychology, University of Exeter, Perry Road, Exeter, UK
| | - Archie Campbell
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
| | - Rebecca A Madden
- Division of Psychiatry, University of Edinburgh, Edinburgh, United Kingdom
| | - Mark J. Adams
- Division of Psychiatry, University of Edinburgh, Edinburgh, United Kingdom
| | - Xiaomeng Bai
- Faculty of Forensic Medicine, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou 510080, China
| | - Ying Liu
- Faculty of Forensic Medicine, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou 510080, China
| | - Miaoxin Li
- Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou 510080, China
| | - Caroline Hayward
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
| | - David J. Porteous
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
| | - Xueyi Shen
- Division of Psychiatry, University of Edinburgh, Edinburgh, United Kingdom
| | - Kathryn L. Evans
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
| | - Chris S. Haley
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
- Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, UK
| | - Andrew M. McIntosh
- Division of Psychiatry, University of Edinburgh, Edinburgh, United Kingdom
| | - Pau Navarro
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
| | - Yanni Zeng
- Faculty of Forensic Medicine, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou 510080, China
- Guangdong Province Translational Forensic Medicine Engineering Technology Research Center Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou 510080, China
- Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou 510080, China
| |
Collapse
|
37
|
Le Goallec A, Collin S, Jabri M, Diai S, Vincent T, Patel CJ. Machine learning approaches to predict age from accelerometer records of physical activity at biobank scale. PLOS DIGITAL HEALTH 2023; 2:e0000176. [PMID: 36812610 PMCID: PMC9931315 DOI: 10.1371/journal.pdig.0000176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Accepted: 12/02/2022] [Indexed: 01/25/2023]
Abstract
Physical activity improves quality of life and protects against age-related diseases. With age, physical activity tends to decrease, increasing vulnerability to disease in the elderly. In the following, we trained a neural network to predict age from 115,456 one week-long 100Hz wrist accelerometer recordings from the UK Biobank (mean absolute error = 3.7±0.2 years), using a variety of data structures to capture the complexity of real-world activity. We achieved this performance by preprocessing the raw frequency data as 2,271 scalar features, 113 time series, and four images. We defined accelerated aging for a participant as being predicted older than one's actual age and identified both genetic and environmental exposure factors associated with the new phenotype. We performed a genome wide association on the accelerated aging phenotypes to estimate its heritability (h_g2 = 12.3±0.9%) and identified ten single nucleotide polymorphisms in close proximity to genes in a histone and olfactory cluster on chromosome six (e.g HIST1H1C, OR5V1). Similarly, we identified biomarkers (e.g blood pressure), clinical phenotypes (e.g chest pain), diseases (e.g hypertension), environmental (e.g smoking), and socioeconomic (e.g income and education) variables associated with accelerated aging. Physical activity-derived biological age is a complex phenotype associated with both genetic and non-genetic factors.
Collapse
Affiliation(s)
- Alan Le Goallec
- Department of Biomedical Informatics, Harvard Medical School, Boston, Massachusetts, United States of America
- Department of Systems, Synthetic and Quantitative Biology, Harvard University, Cambridge, Massachusetts, United States of America
| | - Sasha Collin
- Department of Biomedical Informatics, Harvard Medical School, Boston, Massachusetts, United States of America
| | - M’Hamed Jabri
- Department of Biomedical Informatics, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Samuel Diai
- Department of Biomedical Informatics, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Théo Vincent
- Department of Biomedical Informatics, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Chirag J. Patel
- Department of Biomedical Informatics, Harvard Medical School, Boston, Massachusetts, United States of America
| |
Collapse
|
38
|
El Damaty S, Darcey VL, McQuaid GA, Picci G, Stoianova M, Mucciarone V, Chun Y, Laws ML, Campano V, Van Hecke K, Ryan M, Rose EJ, Fishbein DH, VanMeter AS. Introducing an adolescent cognitive maturity index. Front Psychol 2022; 13:1017317. [PMID: 36571021 PMCID: PMC9771453 DOI: 10.3389/fpsyg.2022.1017317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Accepted: 10/25/2022] [Indexed: 12/12/2022] Open
Abstract
Children show substantial variation in the rate of physical, cognitive, and social maturation as they traverse adolescence and enter adulthood. Differences in developmental paths are thought to underlie individual differences in later life outcomes, however, there remains a lack of consensus on the normative trajectory of cognitive maturation in adolescence. To address this problem, we derive a Cognitive Maturity Index (CMI), to estimate the difference between chronological and cognitive age predicted with latent factor estimates of inhibitory control, risky decision-making and emotional processing measured with standard neuropsychological instruments. One hundred and forty-one children from the Adolescent Development Study (ADS) were followed longitudinally across three time points from ages 11-14, 13-16, and 14-18. Age prediction with latent factor estimates of cognitive skills approximated age within ±10 months (r = 0.71). Males in advanced puberty displayed lower cognitive maturity relative to peers of the same age; manifesting as weaker inhibitory control, greater risk-taking, desensitization to negative affect, and poor recognition of positive affect.
Collapse
Affiliation(s)
- Shady El Damaty
- Interdisciplinary Program in Neuroscience, Georgetown University, Washington, DC, United States
- Center for Functional & Molecular Imaging, Georgetown University Medical Center, Department of Neurology, Washington, DC, United States
| | - Valerie L. Darcey
- Interdisciplinary Program in Neuroscience, Georgetown University, Washington, DC, United States
- Center for Functional & Molecular Imaging, Georgetown University Medical Center, Department of Neurology, Washington, DC, United States
| | - Goldie A. McQuaid
- Center for Functional & Molecular Imaging, Georgetown University Medical Center, Department of Neurology, Washington, DC, United States
| | - Giorgia Picci
- Institute for Human Neuroscience, Boys Town National Research Hospital, Boys Town, NE, United States
- Center for Pediatric Brain Health, Boys Town National Research Hospital, Boys Town, NE, United States
| | - Maria Stoianova
- Center for Functional & Molecular Imaging, Georgetown University Medical Center, Department of Neurology, Washington, DC, United States
| | - Veronica Mucciarone
- Center for Functional & Molecular Imaging, Georgetown University Medical Center, Department of Neurology, Washington, DC, United States
| | - Yewon Chun
- Center for Functional & Molecular Imaging, Georgetown University Medical Center, Department of Neurology, Washington, DC, United States
| | - Marissa L. Laws
- Interdisciplinary Program in Neuroscience, Georgetown University, Washington, DC, United States
| | - Victor Campano
- Center for Functional & Molecular Imaging, Georgetown University Medical Center, Department of Neurology, Washington, DC, United States
| | - Kinney Van Hecke
- Center for Functional & Molecular Imaging, Georgetown University Medical Center, Department of Neurology, Washington, DC, United States
| | - Mary Ryan
- Center for Functional & Molecular Imaging, Georgetown University Medical Center, Department of Neurology, Washington, DC, United States
| | - Emma Jane Rose
- Department of Psychology, The Pennsylvania State University, University Park, PA, United States
| | - Diana H. Fishbein
- Translational Neuro-Prevention Research, Frank Porter Graham Child Development Institute, University of Northern Carolina, Chapel Hill, NC, United States
| | - Ashley S. VanMeter
- Interdisciplinary Program in Neuroscience, Georgetown University, Washington, DC, United States
- Center for Functional & Molecular Imaging, Georgetown University Medical Center, Department of Neurology, Washington, DC, United States
| |
Collapse
|
39
|
Brown TJ, Spurgin LG, Dugdale HL, Komdeur J, Burke T, Richardson DS. Causes and consequences of telomere lengthening in a wild vertebrate population. Mol Ecol 2022; 31:5933-5945. [PMID: 34219315 DOI: 10.1111/mec.16059] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Revised: 05/24/2021] [Accepted: 06/21/2021] [Indexed: 01/31/2023]
Abstract
Telomeres have been advocated to be important markers of biological age in evolutionary and ecological studies. Telomeres usually shorten with age and shortening is frequently associated with environmental stressors and increased subsequent mortality. Telomere lengthening - an apparent increase in telomere length between repeated samples from the same individual - also occurs. However, the exact circumstances, and consequences, of telomere lengthening are poorly understood. Using longitudinal data from the Seychelles warbler (Acrocephalus sechellensis), we tested whether telomere lengthening - which occurs in adults of this species - is associated with specific stressors (reproductive effort, food availability, malarial infection and cooperative breeding) and predicts subsequent survival. In females, telomere shortening was observed under greater stress (i.e., low food availability, malaria infection), while telomere lengthening was observed in females experiencing lower stress (i.e., high food availability, assisted by helpers, without malaria). The telomere dynamics of males were not associated with the key stressors tested. These results indicate that, at least for females, telomere lengthening occurs in circumstances more conducive to self-maintenance. Importantly, both females and males with lengthened telomeres had improved subsequent survival relative to individuals that displayed unchanged, or shortened, telomeres - indicating that telomere lengthening is associated with individual fitness. These results demonstrate that telomere dynamics are bidirectionally responsive to the level of stress that an individual faces, but may poorly reflect the accumulation of stress over an individuals lifetime.
Collapse
Affiliation(s)
- Thomas J Brown
- School of Biological Sciences, University of East Anglia, Norwich, UK
| | - Lewis G Spurgin
- School of Biological Sciences, University of East Anglia, Norwich, UK
| | - Hannah L Dugdale
- Behavioural and Physiological Ecology, Groningen Institute for Evolutionary Life Sciences, University of Groningen, Groningen, The Netherlands
| | - Jan Komdeur
- Behavioural and Physiological Ecology, Groningen Institute for Evolutionary Life Sciences, University of Groningen, Groningen, The Netherlands
| | - Terry Burke
- Department of Animal and Plant Sciences, University of Sheffield, Sheffield, UK
| | - David S Richardson
- School of Biological Sciences, University of East Anglia, Norwich, UK.,Nature Seychelles, Victoria, Mahé, Seychelles
| |
Collapse
|
40
|
Abstract
Age is the key risk factor for diseases and disabilities of the elderly. Efforts to tackle age-related diseases and increase healthspan have suggested targeting the ageing process itself to 'rejuvenate' physiological functioning. However, achieving this aim requires measures of biological age and rates of ageing at the molecular level. Spurred by recent advances in high-throughput omics technologies, a new generation of tools to measure biological ageing now enables the quantitative characterization of ageing at molecular resolution. Epigenomic, transcriptomic, proteomic and metabolomic data can be harnessed with machine learning to build 'ageing clocks' with demonstrated capacity to identify new biomarkers of biological ageing.
Collapse
Affiliation(s)
- Jarod Rutledge
- Department of Genetics, Stanford University, Stanford, CA, USA
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA
- Paul F. Glenn Center for the Biology of Ageing, Stanford University School of Medicine, Stanford, CA, USA
| | - Hamilton Oh
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA
- Paul F. Glenn Center for the Biology of Ageing, Stanford University School of Medicine, Stanford, CA, USA
- Graduate Program in Stem Cell and Regenerative Medicine, Stanford University, Stanford, CA, USA
| | - Tony Wyss-Coray
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA.
- Paul F. Glenn Center for the Biology of Ageing, Stanford University School of Medicine, Stanford, CA, USA.
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA.
| |
Collapse
|
41
|
An evaluation of aging measures: from biomarkers to clocks. Biogerontology 2022; 24:303-328. [PMID: 36418661 DOI: 10.1007/s10522-022-09997-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Accepted: 10/21/2022] [Indexed: 11/25/2022]
Abstract
With the increasing number of aged population and growing burden of healthy aging demands, a rational standard for evaluation aging is in urgent need. The advancement of medical testing technology and the prospering of artificial intelligence make it possible to evaluate the biological status of aging from a more comprehensive view. In this review, we introduced common aging biomarkers and concluded several famous aging clocks. Aging biomarkers reflect changes in the organism at a molecular or cellular level over time while aging clocks tend to be more of a generalization of the overall state of the organism. We expect to construct a framework for aging evaluation measurement from both micro and macro perspectives. Especially, population-specific aging clocks and multi-omics aging clocks may better fit the demands to evaluate aging in a comprehensive and multidimensional manner and make a detailed classification to represent different aging rates at tissue/organ levels. This framework will promisingly provide a crucial basis for disease diagnosis and intervention assessment in geroscience.
Collapse
|
42
|
Familial aggregation of the aging process: biological age measured in young adult offspring as a predictor of parental mortality. GeroScience 2022; 45:901-913. [PMID: 36401109 PMCID: PMC9886744 DOI: 10.1007/s11357-022-00687-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Accepted: 11/06/2022] [Indexed: 11/20/2022] Open
Abstract
Measures of biological age (BA) integrate information across organ systems to quantify "biological aging," i.e., inter-individual differences in aging-related health decline. While longevity and lifespan aggregate in families, reflecting transmission of genes and environments across generations, little is known about intergenerational continuity of biological aging or the extent to which this continuity may be modified by environmental factors. Using data from the Jerusalem Perinatal Study (JPS), we tested if differences in offspring BA were related to mortality in their parents. We measured BA using biomarker data collected from 1473 offspring during clinical exams in 2007-2009, at age 32 ± 1.1. Parental mortality was obtained from population registry data for the years 2004-2016. We fitted parametric survival models to investigate the associations between offspring BA and parental all-cause and cause-specific mortality. We explored potential differences in these relationships by socioeconomic position (SEP) and offspring sex. Participants' BAs widely varied (SD = 6.95). Among those measured to be biologically older, parents had increased all-cause mortality (HR = 1.10, 95% CI: 1.08, 1.13), diabetes mortality (HR = 1.19, 95% CI: 1.08, 1.30), and cancer mortality (HR = 1.07, 95% CI: 1.02, 1.13). The association with all-cause mortality was stronger for families with low compared with high SEP (Pinteraction = 0.04) and for daughters as compared to sons (Pinteraction < 0.001). Using a clinical-biomarker-based BA estimate, observable by young adulthood prior to the onset of aging-related diseases, we demonstrate intergenerational continuity of the aging process. Furthermore, variation in this familial aggregation according to household socioeconomic position (SEP) at offspring birth and between families of sons and daughters proposes that the environment alters individuals' aging trajectory set by their parents.
Collapse
|
43
|
Coelho-Júnior HJ, Calvani R, Tosato M, Landi F, Picca A, Marzetti E. Protein intake and physical function in older adults: A systematic review and meta-analysis. Ageing Res Rev 2022; 81:101731. [PMID: 36087703 DOI: 10.1016/j.arr.2022.101731] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Revised: 08/24/2022] [Accepted: 09/05/2022] [Indexed: 02/07/2023]
Abstract
BACKGROUND The present study explored cross-sectional and longitudinal associations between protein intake and physical function in older adults. METHODS We conducted a systematic review and meta-analysis of cross-sectional and longitudinal studies that investigated the association between protein intake and measures of physical function in older adults. Cross-sectional, case-control, and longitudinal cohort studies that investigated the association between protein intake and physical function as a primary or secondary outcome in people aged 60 + years were included. Studies published in languages other than English, Italian, Portuguese, or Spanish were excluded. Studies were retrieved from MEDLINE, SCOPUS, EMBASE, CINAHL, AgeLine, and Food Science Source databases through January 31, 2022. A pooled effect size was calculated based on standard mean differences (SMD), MD, log odds ratio (OR) and Z-score.. RESULTS Twenty-two cross-sectional studies examined a total of 11,332 community-dwellers, hospitalized older adults, and elite senior athletes with a mean age of approximately 75 years. The pooled analysis indicated that a protein intake higher than the recommended dietary allowance (RDA) was significantly associated with higher Short Physical Performance Battery (SPPB) scores (SMD: 0.63, 95% CI: 0.27, 0.99, P-value: 0.0006), faster walking speed, greater lower-limb (SMD: 0.22, 95% CI: 0.04, 0.40, P-value: 0.02) and isometric handgrip strength (Z-score: 0.087, 95% CI: 0.046-0.128, P-value: 0.0001), and better balance (SMD: 0.33, 95% CI: 0.05, 0.62, P-value: 0.02). Nine longitudinal studies investigated 12,424 community-dwelling and native older adults with a mean age of approximately 85 years. A protein intake higher than the current RDA was not associated with lower decline in either isometric handgrip strength (logOR: 0.99, 95% CI: 0.97-1.02, P-value= 0.67) or walking speed (logOR: 0.92, 95% CI: 0.77-1.10, P-value= 0.35). CONCLUSIONS A protein intake higher than the RDA is cross-sectionally associated with better physical performance and greater muscle strength in older adults. However, a high consumption of proteins does not seem to prevent physical function decline over time.
Collapse
Affiliation(s)
- Hélio José Coelho-Júnior
- Department of Geriatrics and Orthopedics, Università Cattolica del Sacro Cuore, Rome, Italy; Fondazione Policlinico Universitario "Agostino Gemelli" IRCCS, Rome, Italy.
| | - Riccardo Calvani
- Fondazione Policlinico Universitario "Agostino Gemelli" IRCCS, Rome, Italy.
| | - Matteo Tosato
- Fondazione Policlinico Universitario "Agostino Gemelli" IRCCS, Rome, Italy
| | - Francesco Landi
- Department of Geriatrics and Orthopedics, Università Cattolica del Sacro Cuore, Rome, Italy; Fondazione Policlinico Universitario "Agostino Gemelli" IRCCS, Rome, Italy
| | - Anna Picca
- Fondazione Policlinico Universitario "Agostino Gemelli" IRCCS, Rome, Italy
| | - Emanuele Marzetti
- Department of Geriatrics and Orthopedics, Università Cattolica del Sacro Cuore, Rome, Italy; Fondazione Policlinico Universitario "Agostino Gemelli" IRCCS, Rome, Italy
| |
Collapse
|
44
|
Di Lena P, Sala C, Nardini C. Evaluation of different computational methods for DNA methylation-based biological age. Brief Bioinform 2022; 23:6632619. [PMID: 35794713 DOI: 10.1093/bib/bbac274] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 05/27/2022] [Accepted: 06/14/2022] [Indexed: 11/13/2022] Open
Abstract
In recent years there has been a widespread interest in researching biomarkers of aging that could predict physiological vulnerability better than chronological age. Aging, in fact, is one of the most relevant risk factors for a wide range of maladies, and molecular surrogates of this phenotype could enable better patients stratification. Among the most promising of such biomarkers is DNA methylation-based biological age. Given the potential and variety of computational implementations (epigenetic clocks), we here present a systematic review of such clocks. Furthermore, we provide a large-scale performance comparison across different tissues and diseases in terms of age prediction accuracy and age acceleration, a measure of deviance from physiology. Our analysis offers both a state-of-the-art overview of the computational techniques developed so far and a heterogeneous picture of performances, which can be helpful in orienting future research.
Collapse
Affiliation(s)
- Pietro Di Lena
- Department of Computer Science and Engineering, University of Bologna, Mura Anteo Zamboni 7, 40126 Bologna, Italy
| | - Claudia Sala
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Via Massarenti 9, 40138, Bologna, Italy
| | | |
Collapse
|
45
|
Shafqat S, Arana Chicas E, Shafqat A, Hashmi SK. The Achilles' heel of cancer survivors: fundamentals of accelerated cellular senescence. J Clin Invest 2022; 132:e158452. [PMID: 35775492 PMCID: PMC9246373 DOI: 10.1172/jci158452] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Recent improvements in cancer treatment have increased the lifespan of pediatric and adult cancer survivors. However, cancer treatments accelerate aging in survivors, which manifests clinically as the premature onset of chronic diseases, such as endocrinopathies, osteoporosis, cardiac dysfunction, subsequent cancers, and geriatric syndromes of frailty, among others. Therefore, cancer treatment-induced early aging accounts for significant morbidity, mortality, and health expenditures among cancer survivors. One major mechanism driving this accelerated aging is cellular senescence; cancer treatments induce cellular senescence in tumor cells and in normal, nontumor tissue, thereby helping mediate the onset of several chronic diseases. Studies on clinical monitoring and therapeutic targeting of cellular senescence have made considerable progress in recent years. Large-scale clinical trials are currently evaluating senotherapeutic drugs, which inhibit or eliminate senescent cells to ameliorate cancer treatment-related aging. In this article, we survey the recent literature on phenotypes and mechanisms of aging in cancer survivors and provide an up-to-date review of the major preclinical and translational evidence on cellular senescence as a mechanism of accelerated aging in cancer survivors, as well as insight into the potential of senotherapeutic drugs. However, only with time will the clinical effect of senotherapies on cancer survivors be visible.
Collapse
Affiliation(s)
| | - Evelyn Arana Chicas
- Department of Surgery, University of Rochester Medical Center, Rochester, New York, USA
| | - Areez Shafqat
- College of Medicine, Alfaisal University, Riyadh, Saudi Arabia
| | - Shahrukh K. Hashmi
- Department of Internal Medicine, Mayo Clinic, Rochester, Minnesota, USA
- Clinical Affairs, Khalifa University, Abu Dhabi, United Arab Emirates
- Department of Medicine, Sheikh Shakhbout Medical City, Abu Dhabi, United Arab Emirates
| |
Collapse
|
46
|
Husted KLS, Brink-Kjær A, Fogelstrøm M, Hulst P, Bleibach A, Henneberg KÅ, Sørensen HBD, Dela F, Jacobsen JCB, Helge JW. A Model for Estimating Biological Age From Physiological Biomarkers of Healthy Aging: Cross-sectional Study. JMIR Aging 2022; 5:e35696. [PMID: 35536617 PMCID: PMC9131142 DOI: 10.2196/35696] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 03/21/2022] [Accepted: 04/06/2022] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND Individual differences in the rate of aging and susceptibility to disease are not accounted for by chronological age alone. These individual differences are better explained by biological age, which may be estimated by biomarker prediction models. In the light of the aging demographics of the global population and the increase in lifestyle-related morbidities, it is interesting to invent a new biological age model to be used for health promotion. OBJECTIVE This study aims to develop a model that estimates biological age based on physiological biomarkers of healthy aging. METHODS Carefully selected physiological variables from a healthy study population of 100 women and men were used as biomarkers to establish an estimate of biological age. Principal component analysis was applied to the biomarkers and the first principal component was used to define the algorithm estimating biological age. RESULTS The first principal component accounted for 31% in women and 25% in men of the total variance in the biological age model combining mean arterial pressure, glycated hemoglobin, waist circumference, forced expiratory volume in 1 second, maximal oxygen consumption, adiponectin, high-density lipoprotein, total cholesterol, and soluble urokinase-type plasminogen activator receptor. The correlation between the corrected biological age and chronological age was r=0.86 (P<.001) and r=0.81 (P<.001) for women and men, respectively, and the agreement was high and unbiased. No difference was found between mean chronological age and mean biological age, and the slope of the regression line was near 1 for both sexes. CONCLUSIONS Estimating biological age from these 9 biomarkers of aging can be used to assess general health compared with the healthy aging trajectory. This may be useful to evaluate health interventions and as an aid to enhance awareness of individual health risks and behavior when deviating from this trajectory. TRIAL REGISTRATION ClinicalTrials.gov NCT03680768; https://clinicaltrials.gov/ct2/show/NCT03680768. INTERNATIONAL REGISTERED REPORT IDENTIFIER (IRRID) RR2-10.2196/19209.
Collapse
Affiliation(s)
- Karina Louise Skov Husted
- Xlab, Center for Healthy Aging, Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark.,Department of Physiotherapy and Occupational Therapy, University College Copenhagen, Copenhagen, Denmark
| | - Andreas Brink-Kjær
- Digital Health, Department of Health Technology, Technical University of Denmark, Lyngby, Denmark
| | - Mathilde Fogelstrøm
- Xlab, Center for Healthy Aging, Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Pernille Hulst
- Xlab, Center for Healthy Aging, Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Akita Bleibach
- Xlab, Center for Healthy Aging, Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Kaj-Åge Henneberg
- Biomedical Engineering, Department of Health Technology, Technical University of Denmark, Lyngby, Denmark
| | | | - Flemming Dela
- Xlab, Center for Healthy Aging, Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark.,Department of Geriatrics, Bispebjerg and Frederiksberg Hospital, Copenhagen, Denmark
| | - Jens Christian Brings Jacobsen
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jørn Wulff Helge
- Xlab, Center for Healthy Aging, Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
47
|
Vijayakumar KA, Cho GW. Pan-tissue methylation aging clock: Recalibrated and a method to analyze and interpret the selected features. Mech Ageing Dev 2022; 204:111676. [PMID: 35489615 DOI: 10.1016/j.mad.2022.111676] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 03/29/2022] [Accepted: 04/23/2022] [Indexed: 11/29/2022]
Abstract
The abundance of the biological data and the rapid evolution of the newer machine learning technologies have increased the epigenetics research in the last decade. This has enhanced the ability to measure the biological age of humans and different organisms via their omics data. DNA methylation array data are commonly used in the prediction of methylation age. Horvath clock has been adopted in various aging studies as a DNA methylation age predicting clock due to its higher accuracy and multi tissue prediction potential. In the current study, we have developed a pan tissue methylation-aging clock by using the publicly available illumina 450k and EPIC array methylation datasets. In doing that, we developed a highly accurate epigenetic clock, which predicts the age of multiple tissues with higher accuracy. We have also analyzed the selected probes for their biological relevance. Upon analyzing the selected features further, we found out evidences, which support the Antagonistic pleiotropy theory of aging.
Collapse
Affiliation(s)
- Karthikeyan A Vijayakumar
- Department of Biology, College of Natural Science, Chosun University, Gwangju 501-759, South Korea; BK21 FOUR Education Research Group for Age-Associated Disorder Control Technology, Department of Integrative Biological Science, Chosun University, Gwangju 501-759, South Korea
| | - Gwang-Won Cho
- Department of Biology, College of Natural Science, Chosun University, Gwangju 501-759, South Korea; BK21 FOUR Education Research Group for Age-Associated Disorder Control Technology, Department of Integrative Biological Science, Chosun University, Gwangju 501-759, South Korea.
| |
Collapse
|
48
|
Ghisla V, Chocano-Bedoya PO, Orav EJ, Abderhalden LA, Sadlon A, Egli A, Krützfeldt J, Kanis JA, Bischoff-Ferrari HA. Prospective Study of Ageing Trajectories in the European DO-HEALTH Study. Gerontology 2022; 69:57-64. [PMID: 35443250 PMCID: PMC9148895 DOI: 10.1159/000523923] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Accepted: 02/10/2022] [Indexed: 01/06/2023] Open
Abstract
INTRODUCTION Ageing trajectories range from delayed ageing with extended health to accelerated ageing, with an increased risk of frailty. We evaluated the prevalence and prospective change between health states among community-dwelling European older adults. METHODS This prospective study is a secondary analysis of DO-HEALTH, a randomized trial that included adults aged 70 years and older across 5 European countries. Healthy agers (HA) fulfilled the Nurses' Health Study healthy ageing criteria and accelerated agers were non-HA being at least pre-frail according to the Fried frailty criteria. We assessed the proportion of participants changing between health states over 4 assessments and evaluated the odds of changing to a more favourable category. To increase reliability and avoid regression to the mean, we averaged the first 2 years and compared them to the average of the last 2 years. RESULTS Of 2,157 participants, 12.4% were excluded for meeting both healthy ageing and pre-frailty criteria simultaneously. Among the remaining 1,889 participants (mean age 75.1 years, 60.9% female), 23.1% were initially HA, 44.4% were non-HA but not pre-frail, and 32.6% were pre-frail or frail. Subsequently, 65.3% remained in the same health state, 12.0% improved to a healthier state, and 22.8% progressed to a less advantageous state. After adjusting for sex, study centre, treatment, and body mass index, each year of age was associated with 6% lower odds of improving health states. Women had 35% higher odds than men of following a disadvantageous trajectory. CONCLUSION We observed dynamic trajectories of ageing where transitioning to a healthier state became less likely with advancing age and among women.
Collapse
Affiliation(s)
- Virginia Ghisla
- Center on Aging and Mobility, University Hospital Zurich, Zurich City Hospital-Waid and University of Zurich, Zurich, Switzerland,University Clinic for Aging Medicine, Zurich City Hospital-Waid, Zurich, Switzerland
| | - Patricia O. Chocano-Bedoya
- Center on Aging and Mobility, University Hospital Zurich, Zurich City Hospital-Waid and University of Zurich, Zurich, Switzerland,Institute of Primary Health Care (BIHAM), University of Bern, Bern, Switzerland,Population Health Lab, University of Fribourg, Fribourg, Switzerland
| | - Endel John Orav
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Lauren A. Abderhalden
- Center on Aging and Mobility, University Hospital Zurich, Zurich City Hospital-Waid and University of Zurich, Zurich, Switzerland
| | - Angélique Sadlon
- Department of Aging Medicine and Aging Research, University Hospital Zurich and University of Zurich, Zurich, Switzerland
| | - Andreas Egli
- Center on Aging and Mobility, University Hospital Zurich, Zurich City Hospital-Waid and University of Zurich, Zurich, Switzerland
| | - Jan Krützfeldt
- Department of Endocrinology, University Hospital Zurich, Zurich, Switzerland
| | - John A. Kanis
- Mary McKillop Institute for Health Research, Australian Catholic University, Melbourne, Victoria, Australia,Centre for Metabolic Bone Diseases, University of Sheffield, Sheffield, UK
| | - Heike A. Bischoff-Ferrari
- Center on Aging and Mobility, University Hospital Zurich, Zurich City Hospital-Waid and University of Zurich, Zurich, Switzerland,University Clinic for Aging Medicine, Zurich City Hospital-Waid, Zurich, Switzerland,Department of Aging Medicine and Aging Research, University Hospital Zurich and University of Zurich, Zurich, Switzerland,*Heike A. Bischoff-Ferrari,
| | | |
Collapse
|
49
|
Shalnova SA, Imaeva NA, Imaeva AE, Kapustina AV. Aging Challenges. Perceived Age – a New Predictor of Longevity? RATIONAL PHARMACOTHERAPY IN CARDIOLOGY 2022. [DOI: 10.20996/1819-6446-2022-02-06] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The ageing process is accompanied by the manifestation of many characteristics, so-called biomarkers, which can be quantified and used to assess a patient's health status. One of these signs is the progressive decline of a human's facial look, which is described by the concept of 'perceived age'. Facial aging is the most important parameter of perceived age. However, over the years, researchers have identified risk factors that affect the facial skin, including smoking, systematic consumption of alcoholic beverages, overweight or underweight, environmental conditions, and psychosocial determinants. The influence of psychological state on the appearance and life prognosis is shown. The authors presented data from the international literature on the study of perceived age. The frontiers of using perceived age as a biomarker of aging were Danish scientists who developed the main methodological approaches to determine this indicator. One such methodology used in population studies has been the clinical technique of assessing perceived age through photography. The review presents this methodology in detail, with its advantages and modifications. The authors conclude that the measurement of an individual's perceived age can serve not only as a prognostic indicator, but also over time can become a useful marker of the effectiveness of various treatments. Until now perceived age has hardly been studied in population studies, the authors presented data from the works of V.A. Labunskaya, G.V. Serikov, T.A. Shkurko who develop the direction related to psychology of perceived age and in their studies use social-psychological approaches of appearance assessment.
Collapse
Affiliation(s)
- S. A. Shalnova
- National Medical Research Center for Therapy and Preventive Medicine
| | | | - A. E. Imaeva
- National Medical Research Center for Therapy and Preventive Medicine
| | - A. V. Kapustina
- National Medical Research Center for Therapy and Preventive Medicine
| |
Collapse
|
50
|
Li S, Kim HE. Implications of Sphingolipids on Aging and Age-Related Diseases. FRONTIERS IN AGING 2022; 2:797320. [PMID: 35822041 PMCID: PMC9261390 DOI: 10.3389/fragi.2021.797320] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Accepted: 12/31/2021] [Indexed: 01/14/2023]
Abstract
Aging is a process leading to a progressive loss of physiological integrity and homeostasis, and a primary risk factor for many late-onset chronic diseases. The mechanisms underlying aging have long piqued the curiosity of scientists. However, the idea that aging is a biological process susceptible to genetic manipulation was not well established until the discovery that the inhibition of insulin/IGF-1 signaling extended the lifespan of C. elegans. Although aging is a complex multisystem process, López-Otín et al. described aging in reference to nine hallmarks of aging. These nine hallmarks include: genomic instability, telomere attrition, epigenetic alterations, loss of proteostasis, deregulated nutrient sensing, mitochondrial dysfunction, cellular senescence, stem cell exhaustion, and altered intercellular communication. Due to recent advances in lipidomic, investigation into the role of lipids in biological aging has intensified, particularly the role of sphingolipids (SL). SLs are a diverse group of lipids originating from the Endoplasmic Reticulum (ER) and can be modified to create a vastly diverse group of bioactive metabolites that regulate almost every major cellular process, including cell cycle regulation, senescence, proliferation, and apoptosis. Although SL biology reaches all nine hallmarks of aging, its contribution to each hallmark is disproportionate. In this review, we will discuss in detail the major contributions of SLs to the hallmarks of aging and age-related diseases while also summarizing the importance of their other minor but integral contributions.
Collapse
Affiliation(s)
- Shengxin Li
- Department of Integrative Biology and Pharmacology, McGovern Medical School, University of Texas Health Science Center at Houston, TX, United States
- Graduate School of Biomedical Sciences, University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Hyun-Eui Kim
- Department of Integrative Biology and Pharmacology, McGovern Medical School, University of Texas Health Science Center at Houston, TX, United States
- Graduate School of Biomedical Sciences, University of Texas MD Anderson Cancer Center, Houston, TX, United States
| |
Collapse
|