1
|
Mboning L, Costa EK, Chen J, Bouchard LS, Pellegrini M. BayesAge 2.0: a maximum likelihood algorithm to predict transcriptomic age. GeroScience 2025:10.1007/s11357-024-01499-0. [PMID: 39751713 DOI: 10.1007/s11357-024-01499-0] [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: 09/24/2024] [Accepted: 12/22/2024] [Indexed: 01/04/2025] Open
Abstract
Aging is a complex biological process influenced by various factors, including genetic and environmental influences. In this study, we present BayesAge 2.0, an upgraded version of our maximum likelihood algorithm designed for predicting transcriptomic age (tAge) from RNA-seq data. Building on the original BayesAge framework, which was developed for epigenetic age prediction, BayesAge 2.0 integrates a Poisson distribution to model count-based gene expression data and employs LOWESS smoothing to capture nonlinear gene-age relationships. BayesAge 2.0 provides significant improvements over traditional linear models, such as Elastic Net regression. Specifically, it addresses issues of age bias in predictions, with minimal age-associated bias observed in residuals. Its computational efficiency further distinguishes it from traditional models, as reference construction and cross-validation are completed more quickly compared to Elastic Net regression, which requires extensive hyperparameter tuning. Overall, BayesAge 2.0 represents a step forward in tAge prediction, offering a robust, accurate, and efficient tool for aging research and biomarker development.
Collapse
Affiliation(s)
- Lajoyce Mboning
- Department of Chemistry and Biochemistry, University of California Los Angeles, Los Angeles, CA, USA
| | - Emma K Costa
- Department of Neurology and Neurological Sciences, Stanford University, Palo Alto, CA, USA
- Neurosciences Interdepartmental Program, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Jingxun Chen
- Department of Human Genetics, Stanford University, Palo Alto, CA, USA
| | - Louis-S Bouchard
- Department of Chemistry and Biochemistry, University of California Los Angeles, Los Angeles, CA, USA
| | - Matteo Pellegrini
- Department of Molecular, Cell and Developmental Biology, University of California Los Angeles, Los Angeles, CA, USA.
| |
Collapse
|
2
|
Novelle MG, Naranjo-Martínez B, López-Cánovas JL, Díaz-Ruiz A. Fecal microbiota transplantation, a tool to transfer healthy longevity. Ageing Res Rev 2025; 103:102585. [PMID: 39586550 DOI: 10.1016/j.arr.2024.102585] [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: 06/14/2024] [Revised: 10/13/2024] [Accepted: 11/12/2024] [Indexed: 11/27/2024]
Abstract
The complex gut microbiome influences host aging and plays an important role in the manifestation of age-related diseases. Restoring a healthy gut microbiome via Fecal Microbiota Transplantation (FMT) is receiving extensive consideration to therapeutically transfer healthy longevity. Herein, we comprehensively review the benefits of gut microbial rejuvenation - via FMT - to promote healthy aging, with few studies documenting life length properties. This review explores how preconditioning donors via standard - lifestyle and pharmacological - antiaging interventions reshape gut microbiome, with the resulting benefits being also FMT-transferable. Finally, we expose the current clinical uses of FMT in the context of aging therapy and address FMT challenges - regulatory landscape, protocol standardization, and health risks - that require refinement to effectively utilize microbiome interventions in the elderly.
Collapse
Affiliation(s)
- Marta G Novelle
- Department of Genetics, Physiology and Microbiology (Unity of Animal Physiology), Faculty of Biology, Complutense University of Madrid (UCM), Madrid, Spain; CIBER Physiopathology of Obesity and Nutrition (CIBERobn), Spain
| | - Beatriz Naranjo-Martínez
- Laboratory of Cellular and Molecular Gerontology, Precision Nutrition and Aging, Madrid Institute for Advanced Studies - IMDEA Food, CEI UAM+CSIC, Madrid, Spain
| | - Juan L López-Cánovas
- Laboratory of Cellular and Molecular Gerontology, Precision Nutrition and Aging, Madrid Institute for Advanced Studies - IMDEA Food, CEI UAM+CSIC, Madrid, Spain
| | - Alberto Díaz-Ruiz
- Laboratory of Cellular and Molecular Gerontology, Precision Nutrition and Aging, Madrid Institute for Advanced Studies - IMDEA Food, CEI UAM+CSIC, Madrid, Spain; CIBER Physiopathology of Obesity and Nutrition (CIBERobn), Spain.
| |
Collapse
|
3
|
Wang Y, Cao X, Ma J, Liu S, Jin X, Liu B. Unveiling the Longevity Potential of Natural Phytochemicals: A Comprehensive Review of Active Ingredients in Dietary Plants and Herbs. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:24908-24927. [PMID: 39480905 PMCID: PMC11565747 DOI: 10.1021/acs.jafc.4c07756] [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: 08/22/2024] [Revised: 10/25/2024] [Accepted: 10/25/2024] [Indexed: 11/02/2024]
Abstract
Ancient humans used dietary plants and herbs to treat disease and to pursue eternal life. Today, phytochemicals in dietary plants and herbs have been shown to be the active ingredients, some of which have antiaging and longevity-promoting effects. Here, we summarize 210 antiaging phytochemicals in dietary plants and herbs, systematically classify them into 8 groups. We found that all groups of phytochemicals can be categorized into six areas that regulate organism longevity: ROS levels, nutrient sensing network, mitochondria, autophagy, gut microbiota, and lipid metabolism. We review the role of these processes in aging and the molecular mechanism of the health benefits through phytochemical-mediated regulation. Among these, how phytochemicals promote longevity through the gut microbiota and lipid metabolism is rarely highlighted in the field. Our understanding of the mechanisms of phytochemicals based on the above six aspects may provide a theoretical basis for the further development of antiaging drugs and new insights into the promotion of human longevity.
Collapse
Affiliation(s)
- Yu Wang
- State
Key Laboratory of Subtropical Silviculture, School of Forestry and
Biotechnology, Zhejiang A&F University, Hangzhou 311300, China
| | - Xiuling Cao
- State
Key Laboratory of Subtropical Silviculture, School of Forestry and
Biotechnology, Zhejiang A&F University, Hangzhou 311300, China
| | - Jin Ma
- State
Key Laboratory of Subtropical Silviculture, School of Forestry and
Biotechnology, Zhejiang A&F University, Hangzhou 311300, China
| | - Shenkui Liu
- State
Key Laboratory of Subtropical Silviculture, School of Forestry and
Biotechnology, Zhejiang A&F University, Hangzhou 311300, China
| | - Xuejiao Jin
- State
Key Laboratory of Subtropical Silviculture, School of Forestry and
Biotechnology, Zhejiang A&F University, Hangzhou 311300, China
| | - Beidong Liu
- State
Key Laboratory of Subtropical Silviculture, School of Forestry and
Biotechnology, Zhejiang A&F University, Hangzhou 311300, China
- Department
of Chemistry and Molecular Biology, University
of Gothenburg, Gothenburg 41390, Sweden
| |
Collapse
|
4
|
Badial K, Lacayo P, Murakami S. Biology of Healthy Aging: Biological Hallmarks of Stress Resistance Related and Unrelated to Longevity in Humans. Int J Mol Sci 2024; 25:10493. [PMID: 39408822 PMCID: PMC11477412 DOI: 10.3390/ijms251910493] [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: 07/25/2024] [Revised: 09/23/2024] [Accepted: 09/27/2024] [Indexed: 10/20/2024] Open
Abstract
Stress resistance is highly associated with longer and healthier lifespans in various model organisms, including nematodes, fruit flies, and mice. However, we lack a complete understanding of stress resistance in humans; therefore, we investigated how stress resistance and longevity are interlinked in humans. Using more than 180 databases, we identified 541 human genes associated with stress resistance. The curated gene set is highly enriched with genes involved in the cellular response to stress. The Reactome analysis identified 398 biological pathways, narrowed down to 172 pathways using a medium threshold (p-value < 1 × 10-4). We further summarized these pathways into 14 pathway categories, e.g., cellular response to stimuli/stress, DNA repair, gene expression, and immune system. There were overlapping categories between stress resistance and longevity, including gene expression, signal transduction, immune system, and cellular responses to stimuli/stress. The categories include the PIP3-AKT-FOXO and mTOR pathways, known to specify lifespans in the model systems. They also include the accelerated aging syndrome genes (WRN and HGPS/LMNA), while the genes were also involved in non-overlapped categories. Notably, nuclear pore proteins are enriched among the stress-resistance pathways and overlap with diverse metabolic pathways. This study fills the knowledge gap in humans, suggesting that stress resistance is closely linked to longevity pathways but not entirely identical. While most longevity categories intersect with stress-resistance categories, some do not, particularly those related to cell proliferation and beta-cell development. We also note inconsistencies in pathway terminologies with aging hallmarks reported previously, and propose them to be more unified and integral.
Collapse
Affiliation(s)
| | | | - Shin Murakami
- Department of Foundational Biomedical Sciences, College of Osteopathic Medicine, Touro University California, Vallejo, CA 94592, USA
| |
Collapse
|
5
|
Mboning L, Costa EK, Chen J, Bouchard LS, Pellegrini M. BayesAge 2.0: A Maximum Likelihood Algorithm to Predict Transcriptomic Age. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.16.613354. [PMID: 39345375 PMCID: PMC11429879 DOI: 10.1101/2024.09.16.613354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
Aging is a complex biological process influenced by various factors, including genetic and environmental influences. In this study, we present BayesAge 2.0, an improved version of our maximum likelihood algorithm designed for predicting transcriptomic age (tAge) from RNA-seq data. Building on the original BayesAge framework, which was developed for epigenetic age prediction, BayesAge 2.0 integrates a Poisson distribution to model count-based gene expression data and employs LOWESS smoothing to capture non-linear gene-age relationships. BayesAge 2.0 provides significant improvements over traditional linear models, such as Elastic Net regression. Specifically, it addresses issues of age bias in predictions, with minimal age-associated bias observed in residuals. Its computational efficiency further distinguishes it from traditional models, as reference construction and cross-validation are completed more quickly compared to Elastic Net regression, which requires extensive hyperparameter tuning. Overall, BayesAge 2.0 represents a notable advance in transcriptomic age prediction, offering a robust, accurate, and efficient tool for aging research and biomarker development.
Collapse
Affiliation(s)
- Lajoyce Mboning
- Department of Chemistry and Biochemistry, University of California, Los Angeles, Los Angeles, California, United States
| | - Emma K. Costa
- Department of Neurology and Neurological Sciences, Stanford University, Palo Alto, California, United States
- Neurosciences Interdepartmental Program, Stanford University School of Medicine, Palo Alto, California, United States
| | - Jingxun Chen
- Department of Human Genetics, Stanford University, Palo Alto, California, United States
| | - Louis-S Bouchard
- Department of Chemistry and Biochemistry, University of California, Los Angeles, Los Angeles, California, United States
| | - Matteo Pellegrini
- Department of Molecular, Cell and Developmental Biology, University of California Los Angeles, Los Angeles, California, United States
| |
Collapse
|
6
|
Liebich A, Zheng S, Schachner T, Mair J, Jovanova M, Müller-Riemenschneider F, Kowatsch T. Non-pharmaceutical interventions and epigenetic aging in adults: Protocol for a scoping review. PLoS One 2024; 19:e0301763. [PMID: 39159141 PMCID: PMC11332928 DOI: 10.1371/journal.pone.0301763] [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: 01/11/2024] [Accepted: 03/20/2024] [Indexed: 08/21/2024] Open
Abstract
INTRODUCTION Aging is the strongest risk factor for most chronic diseases. The rising burden of an aging population and non-communicable diseases (NCDs), contributes to escalating costs for society. Several non-pharmaceutical interventions can lower the risk of NCDs, including common mental disorders (CMDs), and may slow down biological aging, as evidenced by outcome markers such as epigenetic clocks. However, a comprehensive overview of whether and which non-pharmaceutical interventions may impact human epigenetic aging is missing. Synthesizing evidence of interventions on epigenetic aging that can be adopted by a wider population is key to guide healthy aging initiatives and to reduce the burden of NCDs and CMDs. This scoping review will identify and assess non-pharmaceutical interventions aimed to slow down epigenetic aging, including their intervention components, and the mode used for intervention delivery. METHODS AND ANALYSIS This protocol will include single- and multicomponent intervention studies that target individuals ≥ 18 years of age and use epigenetic clocks as primary or secondary outcomes. Five electronic databases will be searched for studies between July 2011 until December 2023. The final search will include the search terms adults, non-pharmaceutical interventions, epigenetic aging and their respective synonyms. We will include randomized controlled trials, non-randomized controlled studies, cohort studies, and case-control studies. Additionally, the reference list of other reviews will be screened for relevant articles. Study selection is carried out based on the defined eligibility criteria by two authors. Quality and risk of bias for the included studies will be assessed using the Critical Appraisal Skills Programme (CASP) checklist. Data extraction will include generic key information such as the research question and results, the intervention components, and specific epigenetic outcome measures used. Further data regarding the delivery mode of the treatment protocol will be collected. ETHICS AND DISSEMINATION This scoping review will summarize the characteristics of non-pharmaceutical intervention studies on epigenetic aging. This review will be the first step to formally identify key intervention components and delivery modes to guide future research on healthy aging interventions. The results will be disseminated through a peer-reviewed publication and presented at relevant conferences. This review will synthesize information from available publications and does not require further ethical approval. REGISTRATION DETAILS Open Science Framework https://doi.org/10.17605/OSF.IO/FEHNB.
Collapse
Affiliation(s)
- Alina Liebich
- School of Medicine, University of St. Gallen, St.Gallen, Switzerland
| | - Shenglin Zheng
- Saw Swee Hock School of Public Health, National University of Singapore and National University Health System, Singapore, Singapore
- Future Health Technologies, Singapore–ETH Centre, Campus for Research Excellence And Technological Enterprise (CREATE), Singapore, Singapore
| | - Theresa Schachner
- Department of Management, Technology, and Economics, ETH Zurich, Zurich, Switzerland
| | - Jacqueline Mair
- Saw Swee Hock School of Public Health, National University of Singapore and National University Health System, Singapore, Singapore
- Future Health Technologies, Singapore–ETH Centre, Campus for Research Excellence And Technological Enterprise (CREATE), Singapore, Singapore
| | - Mia Jovanova
- School of Medicine, University of St. Gallen, St.Gallen, Switzerland
| | - Falk Müller-Riemenschneider
- Saw Swee Hock School of Public Health, National University of Singapore and National University Health System, Singapore, Singapore
- Digital Health Center, Berlin Institute of Health, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Tobias Kowatsch
- School of Medicine, University of St. Gallen, St.Gallen, Switzerland
- Department of Management, Technology, and Economics, ETH Zurich, Zurich, Switzerland
- Institute for Implementation Science in Health Care, University of Zurich, Zurich, Switzerland
| |
Collapse
|
7
|
Mironov S, Borysova O, Morgunov I, Zhou Z, Moskalev A. A Framework for an Effective Healthy Longevity Clinic. Aging Dis 2024:AD.2024.0328-1. [PMID: 38607731 DOI: 10.14336/ad.2024.0328-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Accepted: 07/15/2024] [Indexed: 09/11/2024] Open
Abstract
In the context of an aging global population and the imperative for innovative healthcare solutions, the concept of longevity clinics emerges as a timely and vital area of exploration. Unlike traditional medical facilities, longevity clinics offer a unique approach to preclinical prevention, focusing on "prevention of prevention" through the utilization of aging clocks and biomarkers from healthy individuals. This article presents a comprehensive overview of longevity clinics, encompassing descriptions of existing models, the development of a proposed framework, and insights into biomarkers, wearable devices, and therapeutic interventions. Additionally, economic justifications for investing in longevity clinics are examined, highlighting the significant growth potential of the global biotechnology market and its alignment with the goals of achieving active longevity. Anchored by an Analytical Center, the proposed framework underscores the importance of data-driven decision-making and innovation in promoting prolonged and enhanced human life. At present, there is no universally accepted standard model for longevity clinics. This absence highlights the need for additional research and ongoing improvements in this field. Through a synthesis of scientific research and practical considerations, this article aims to stimulate further discussion and innovation in the field of longevity clinics, ultimately contributing to the advancement of healthcare practices aimed at extending and enhancing human life.
Collapse
Affiliation(s)
- Sergey Mironov
- Longaevus Technologies LTD, London, United Kingdom
- Human and health division, DEKRA Automobil GmbH, Chemnitz, Germany
| | | | | | - Zhongjun Zhou
- School of Biomedical Sciences, University of Hong Kong, Hong Kong
| | - Alexey Moskalev
- Longaevus Technologies LTD, London, United Kingdom
- Institute of biogerontology, National Research Lobachevsky State University of Nizhni Novgorod (Lobachevsky University), Nizhny Novgorod, Russia
- Gerontological Research and Clinical Center, Russian National Research Medical University, Moscow, Russia
| |
Collapse
|
8
|
Harinath G, Zalzala S, Nyquist A, Wouters M, Isman A, Moel M, Verdin E, Kaeberlein M, Kennedy B, Bischof E. The role of quality of life data as an endpoint for collecting real-world evidence within geroscience clinical trials. Ageing Res Rev 2024; 97:102293. [PMID: 38574864 DOI: 10.1016/j.arr.2024.102293] [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: 02/02/2024] [Revised: 03/21/2024] [Accepted: 04/01/2024] [Indexed: 04/06/2024]
Abstract
With geroscience research evolving at a fast pace, the need arises for human randomized controlled trials to assess the efficacy of geroprotective interventions to prevent age-related adverse outcomes, disease, and mortality in normative aging cohorts. However, to confirm efficacy requires a long-term and costly approach as time to the event of morbidity and mortality can be decades. While this could be circumvented using sensitive biomarkers of aging, current molecular, physiological, and digital endpoints require further validation. In this review, we discuss how collecting real-world evidence (RWE) by obtaining health data that is amenable for collection from large heterogeneous populations in a real-world setting can help speed up validation of geroprotective interventions. Further, we propose inclusion of quality of life (QoL) data as a biomarker of aging and candidate endpoint for geroscience clinical trials to aid in distinguishing healthy from unhealthy aging. We highlight how QoL assays can aid in accelerating data collection in studies gathering RWE on the geroprotective effects of repurposed drugs to support utilization within healthy longevity medicine. Finally, we summarize key metrics to consider when implementing QoL assays in studies, and present the short-form 36 (SF-36) as the most well-suited candidate endpoint.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Eric Verdin
- Buck Institute for Research on Aging, Novato, CA, USA
| | | | - Brian Kennedy
- Healthy Longevity Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Centre for Healthy Longevity, National University Health System, Singapore
| | - Evelyne Bischof
- Department of Medical Oncology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China; Shanghai University of Medicine and Health Sciences, Shanghai, China; Sheba Longevity Center, Sheba Medical Center, Tel Aviv, Israel.
| |
Collapse
|
9
|
Bueno Yáñez O, Calvo Aguirre JJ, Uranga Zaldua J, Alustiza Navarro J, Ugartemendia Yerobi M. [Alusti test as a premonitory variable of adverse health events in a nursing home. Two-years follow-up]. Rev Esp Geriatr Gerontol 2024; 59:101476. [PMID: 38417197 DOI: 10.1016/j.regg.2024.101476] [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: 07/31/2023] [Revised: 01/25/2024] [Accepted: 01/29/2024] [Indexed: 03/01/2024]
Abstract
INTRODUCTION Functional capacity is a good indicator of health, quality of life, and a good predictor of morbimortality. It is a priority to functionally assess the geriatric population through objective, precise, and simple instruments. The Alusti Test in its two versions, complete (TA) and abbreviated (TAA), is a scale that meets these criteria. OBJECTIVE To determine the usefulness of the Alusti Test as a predictor of adverse health events: falls, hospitalizations, cognitive deterioration, and mortality in the elderly institutionalized population, with a two-year follow-up. MATERIAL AND METHODS This observational study's sample included 176 persons admitted to a nursing home for 32months, with a mean age of 85.5years. The TA was performed on 138 and the TAA on 38. RESULTS The ratio of falls is much higher in residents with mild dependence than in those with total dependence (P<.001). Hospitalizations increase as the results of the Alusti Test are more favorable. The risk of hospitalization in dependent patients is 50% lower (P<.001) than in those with preserved mobility. Cognitive impairment is similar in all the populations with some mild-moderate level of functional dependence and decreases in the population with preserved mobility. Categorization as total and mild/severe dependence is related to a 3-4times higher mortality at six months follow-up. CONCLUSIONS A higher mild-moderate level of dependence on the AT correlates with a lower risk of falls, a lower rate of hospitalization, and a higher risk of mortality at six months.
Collapse
Affiliation(s)
- Olga Bueno Yáñez
- Osakidetza, Centro de Salud Beraun, Errenteria, Guipúzcoa, España
| | | | | | | | - Maider Ugartemendia Yerobi
- Departamento de Enfermería, Facultad de Medicina y Enfermería, Universidad del País Vasco UPV/EHU, San Sebastián, España.
| |
Collapse
|
10
|
Wang S, Song L, Fan R, Chen Q, You M, Cai M, Wu Y, Li Y, Xu M. Targeting Aging and Longevity with Exogenous Nucleotides (TALENTs): Rationale, Design, and Baseline Characteristics from a Randomized Controlled Trial in Older Adults. Nutrients 2024; 16:1343. [PMID: 38732590 PMCID: PMC11085046 DOI: 10.3390/nu16091343] [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: 03/26/2024] [Revised: 04/25/2024] [Accepted: 04/28/2024] [Indexed: 05/13/2024] Open
Abstract
Nucleotides (NTs), important biomolecules involved in numerous cellular processes, have been proposed as potential candidates for anti-aging interventions. However, whether nucleotides can act as an anti-aging supplement in older adults remains unclear. TALENTs is a randomized, double-blinded, placebo-controlled trial that evaluates the efficacy and safety of NTs as an anti-aging supplement in older adults by exploring the effects of NTs on multiple dimensions of aging in a rigorous scientific setting. Eligible community-dwelling adults aged 60-70 years were randomly assigned equally to two groups: nucleotides intervention group and placebo control group. Comprehensive geriatric health assessments were performed at baseline, 2-months, and 4-months of the intervention. Biological specimens were collected and stored for age-related biomarker testing and multi-omics sequencing. The primary outcome was the change from baseline to 4 months on leukocyte telomere length and DNA methylation age. The secondary aims were the changes in possible mechanisms underlying aging processes (immunity, inflammatory profile, oxidative stress, gene stability, endocrine, metabolism, and cardiovascular function). Other outcomes were changes in physical function, body composition and geriatric health assessment (including sleep quality, cognitive function, fatigue, frailty, and psychology). In the RCT, 301 participants were assessed for eligibility and 122 were enrolled. Participants averaged 65.65 years of age, and were predominately female (67.21%). All baseline characteristics were well-balanced between groups, as expected due to randomization. The majority of participants were pre-frailty and had at least one chronic condition. The mean scores for physical activity, psychological, fatigue and quality of life were within the normal range. However, nearly half of the participants still had room for improvement in cognitive level and sleep quality. This TALENTs trial will represent one of the most comprehensive experimental clinical trials in which supplements are administered to elderly participants. The findings of this study will contribute to our understanding of the anti-aging effects of NTs and provide insights into their potential applications in geriatric healthcare.
Collapse
Affiliation(s)
- Shuyue Wang
- Department of Nutrition and Food Hygiene, School of Public Health, Peking University, Beijing 100191, China; (S.W.); (L.S.); (R.F.); (Q.C.); (M.Y.); (M.C.); (Y.W.); (Y.L.)
- Beijing Key Laboratory of Toxicological Research and Risk Assessment for Food Safety, Peking University, Beijing 100191, China
| | - Lixia Song
- Department of Nutrition and Food Hygiene, School of Public Health, Peking University, Beijing 100191, China; (S.W.); (L.S.); (R.F.); (Q.C.); (M.Y.); (M.C.); (Y.W.); (Y.L.)
- Beijing Key Laboratory of Toxicological Research and Risk Assessment for Food Safety, Peking University, Beijing 100191, China
| | - Rui Fan
- Department of Nutrition and Food Hygiene, School of Public Health, Peking University, Beijing 100191, China; (S.W.); (L.S.); (R.F.); (Q.C.); (M.Y.); (M.C.); (Y.W.); (Y.L.)
- Beijing Key Laboratory of Toxicological Research and Risk Assessment for Food Safety, Peking University, Beijing 100191, China
| | - Qianqian Chen
- Department of Nutrition and Food Hygiene, School of Public Health, Peking University, Beijing 100191, China; (S.W.); (L.S.); (R.F.); (Q.C.); (M.Y.); (M.C.); (Y.W.); (Y.L.)
- Beijing Key Laboratory of Toxicological Research and Risk Assessment for Food Safety, Peking University, Beijing 100191, China
| | - Mei You
- Department of Nutrition and Food Hygiene, School of Public Health, Peking University, Beijing 100191, China; (S.W.); (L.S.); (R.F.); (Q.C.); (M.Y.); (M.C.); (Y.W.); (Y.L.)
- Beijing Key Laboratory of Toxicological Research and Risk Assessment for Food Safety, Peking University, Beijing 100191, China
| | - Meng Cai
- Department of Nutrition and Food Hygiene, School of Public Health, Peking University, Beijing 100191, China; (S.W.); (L.S.); (R.F.); (Q.C.); (M.Y.); (M.C.); (Y.W.); (Y.L.)
- Beijing Key Laboratory of Toxicological Research and Risk Assessment for Food Safety, Peking University, Beijing 100191, China
| | - Yuxiao Wu
- Department of Nutrition and Food Hygiene, School of Public Health, Peking University, Beijing 100191, China; (S.W.); (L.S.); (R.F.); (Q.C.); (M.Y.); (M.C.); (Y.W.); (Y.L.)
- Beijing Key Laboratory of Toxicological Research and Risk Assessment for Food Safety, Peking University, Beijing 100191, China
| | - Yong Li
- Department of Nutrition and Food Hygiene, School of Public Health, Peking University, Beijing 100191, China; (S.W.); (L.S.); (R.F.); (Q.C.); (M.Y.); (M.C.); (Y.W.); (Y.L.)
- Beijing Key Laboratory of Toxicological Research and Risk Assessment for Food Safety, Peking University, Beijing 100191, China
| | - Meihong Xu
- Department of Nutrition and Food Hygiene, School of Public Health, Peking University, Beijing 100191, China; (S.W.); (L.S.); (R.F.); (Q.C.); (M.Y.); (M.C.); (Y.W.); (Y.L.)
- Beijing Key Laboratory of Toxicological Research and Risk Assessment for Food Safety, Peking University, Beijing 100191, China
| |
Collapse
|
11
|
Ganau A, Orrù M, Floris M, Saba PS, Loi F, Sanna GD, Marongiu M, Balaci L, Curreli N, Ferreli LAP, Loi F, Masala M, Parodi G, Delitala AP, Schlessinger D, Lakatta E, Fiorillo E, Cucca F. Echocardiographic heart ageing patterns predict cardiovascular and non-cardiovascular events and reflect biological age: the SardiNIA study. Eur J Prev Cardiol 2024; 31:677-685. [PMID: 37527539 PMCID: PMC11025036 DOI: 10.1093/eurjpc/zwad254] [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: 05/10/2023] [Revised: 07/04/2023] [Accepted: 07/28/2023] [Indexed: 08/03/2023]
Abstract
AIMS Age is a crucial risk factor for cardiovascular (CV) and non-CV diseases. As people age at different rates, the concept of biological age has been introduced as a personalized measure of functional deterioration. Associations of age with echocardiographic quantitative traits were analysed to assess different heart ageing rates and their ability to predict outcomes and reflect biological age. METHODS AND RESULTS Associations of age with left ventricular mass, geometry, diastolic function, left atrial volume, and aortic root size were measured in 2614 healthy subjects. Based on the 95% two-sided tolerance intervals of each correlation, three discrete ageing trajectories were identified and categorized as 'slow', 'normal', and 'accelerated' heart ageing patterns. The primary endpoint included fatal and non-fatal CV events, and the secondary endpoint was a composite of CV and non-CV events and all-cause death. The phenotypic age of the heart (HeartPhAge) was estimated as a proxy of biological age. The slow ageing pattern was found in 8.7% of healthy participants, the normal pattern in 76.9%, and the accelerated pattern in 14.4%. Kaplan-Meier curves of the heart ageing patterns diverged significantly (P = 0.0001) for both primary and secondary endpoints, with the event rate being lowest in the slow, intermediate in the normal, and highest in the accelerated pattern. In the Cox proportional hazards model, heart ageing patterns predicted both primary (P = 0.01) and secondary (P = 0.03 to <0.0001) endpoints, independent of chronological age and risk factors. Compared with chronological age, HeartPhAge was 9 years younger in slow, 4 years older in accelerated (both P < 0.0001), and overlapping in normal ageing patterns. CONCLUSION Standard Doppler echocardiography detects slow, normal, and accelerated heart ageing patterns. They predict CV and non-CV events, reflect biological age, and provide a new tool to calibrate prevention timing and intensity.
Collapse
Affiliation(s)
- Antonello Ganau
- Department of Medicine, Surgery, and Pharmacy, University of Sassari, Via Istria12, 07100 Sassari, Italy, Italy
| | - Marco Orrù
- Armando Businco Hospital, Azienda Ospedaliera Brotzu, Cagliari 09047, Italy
| | - Matteo Floris
- Department of Biomedical Sciences, University of Sassari, Sassari 07100, Italy
- Institute of Genetics and Biomedical Research, National Research Council, Monserrato, Cagliari 09042, Italy
| | - Pier Sergio Saba
- Cardiac Thoracic Vascular Department, Azienda Ospedaliero Universitaria, Sassari 07100, Italy
| | - Federica Loi
- Department of Cardiac Thoracic Vascular Sciences and Public Health, University of Padova, Padova 35128, Italy
| | - Giuseppe D Sanna
- Cardiac Thoracic Vascular Department, Azienda Ospedaliero Universitaria, Sassari 07100, Italy
| | - Michele Marongiu
- Institute of Genetics and Biomedical Research, National Research Council, Monserrato, Cagliari 09042, Italy
| | - Lenuta Balaci
- Institute of Genetics and Biomedical Research, National Research Council, Monserrato, Cagliari 09042, Italy
| | - Niccolò Curreli
- Institute of Genetics and Biomedical Research, National Research Council, Monserrato, Cagliari 09042, Italy
| | - Liana A P Ferreli
- Institute of Genetics and Biomedical Research, National Research Council, Monserrato, Cagliari 09042, Italy
| | - Francesco Loi
- Institute of Genetics and Biomedical Research, National Research Council, Monserrato, Cagliari 09042, Italy
| | - Marco Masala
- Institute of Genetics and Biomedical Research, National Research Council, Monserrato, Cagliari 09042, Italy
| | - Guido Parodi
- Department of Medicine, Surgery, and Pharmacy, University of Sassari, Via Istria12, 07100 Sassari, Italy, Italy
| | - Alessandro P Delitala
- Department of Medicine, Surgery, and Pharmacy, University of Sassari, Via Istria12, 07100 Sassari, Italy, Italy
| | - David Schlessinger
- Laboratory of Genetics & Genomics, NIH/National Institute of Ageing, Bethesda, MD, USA
| | - Edward Lakatta
- Laboratory of Cardiovascular Science, NIH/National Institute of Ageing, Bethesda, MD, USA
| | - Edoardo Fiorillo
- Institute of Genetics and Biomedical Research, National Research Council, Monserrato, Cagliari 09042, Italy
| | - Francesco Cucca
- Department of Biomedical Sciences, University of Sassari, Sassari 07100, Italy
- Institute of Genetics and Biomedical Research, National Research Council, Monserrato, Cagliari 09042, Italy
| |
Collapse
|
12
|
Schellnegger M, Hofmann E, Carnieletto M, Kamolz LP. Unlocking longevity: the role of telomeres and its targeting interventions. FRONTIERS IN AGING 2024; 5:1339317. [PMID: 38333665 PMCID: PMC10850353 DOI: 10.3389/fragi.2024.1339317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Accepted: 01/11/2024] [Indexed: 02/10/2024]
Abstract
Average life expectancy has been steadily increasing in developed countries worldwide. These demographic changes are associated with an ever-growing social and economic strain to healthcare systems as well as society. The aging process typically manifests as a decline in physiological and cognitive functions, accompanied by a rise in chronic diseases. Consequently, strategies that both mitigate age-related diseases and promote healthy aging are urgently needed. Telomere attrition, characterized by the shortening of telomeres with each cell division, paradoxically serves as both a protective mechanism and a contributor to tissue degeneration and age-related ailments. Based on the essential role of telomere biology in aging, research efforts aim to develop approaches designed to counteract telomere attrition, aiming to delay or reduce age-related diseases. In this review, telomere biology and its role in aging and age-related diseases is summarized along with recent approaches to interfere with telomere shortening aiming at well- and healthy-aging as well as longevity. As aging research enters a new era, this review emphasizes telomere-targeting therapeutics, including telomerase activators and tankyrase inhibitors, while also exploring the effects of antioxidative and anti-inflammatory agents, along with indirectly related approaches like statins.
Collapse
Affiliation(s)
- Marlies Schellnegger
- Division of Plastic, Aesthetic and Reconstructive Surgery, Department of Surgery, Medical University of Graz, Graz, Austria
- COREMED–Centre for Regenerative and Precision Medicine, JOANNEUM RESEARCH Forschungsgesellschaft mbH, Graz, Austria
| | - Elisabeth Hofmann
- Division of Plastic, Aesthetic and Reconstructive Surgery, Department of Surgery, Medical University of Graz, Graz, Austria
- COREMED–Centre for Regenerative and Precision Medicine, JOANNEUM RESEARCH Forschungsgesellschaft mbH, Graz, Austria
- Research Unit for Tissue Regeneration, Repair and Reconstruction, Division of Plastic, Aesthetic and Reconstructive Surgery, Department of Surgery, Medical University of Graz, Graz, Austria
| | - Martina Carnieletto
- Division of Plastic, Aesthetic and Reconstructive Surgery, Department of Surgery, Medical University of Graz, Graz, Austria
- COREMED–Centre for Regenerative and Precision Medicine, JOANNEUM RESEARCH Forschungsgesellschaft mbH, Graz, Austria
| | - Lars-Peter Kamolz
- Division of Plastic, Aesthetic and Reconstructive Surgery, Department of Surgery, Medical University of Graz, Graz, Austria
- COREMED–Centre for Regenerative and Precision Medicine, JOANNEUM RESEARCH Forschungsgesellschaft mbH, Graz, Austria
| |
Collapse
|
13
|
Venn-Watson S, Schork NJ. Pentadecanoic Acid (C15:0), an Essential Fatty Acid, Shares Clinically Relevant Cell-Based Activities with Leading Longevity-Enhancing Compounds. Nutrients 2023; 15:4607. [PMID: 37960259 PMCID: PMC10649853 DOI: 10.3390/nu15214607] [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: 10/08/2023] [Revised: 10/24/2023] [Accepted: 10/27/2023] [Indexed: 11/15/2023] Open
Abstract
Pentadecanoic acid (C15:0) is an essential odd-chain saturated fatty acid with broad activities relevant to protecting cardiometabolic, immune, and liver health. C15:0 activates AMPK and inhibits mTOR, both of which are core components of the human longevity pathway. To assess the potential for C15:0 to enhance processes associated with longevity and healthspan, we used human cell-based molecular phenotyping assays to compare C15:0 with three longevity-enhancing candidates: acarbose, metformin, and rapamycin. C15:0 (n = 36 activities in 10 of 12 cell systems) and rapamycin (n = 32 activities in 12 of 12 systems) had the most clinically relevant, dose-dependent activities. At their optimal doses, C15:0 (17 µM) and rapamycin (9 µM) shared 24 activities across 10 cell systems, including anti-inflammatory (e.g., lowered MCP-1, TNFα, IL-10, IL-17A/F), antifibrotic, and anticancer activities, which are further supported by previously published in vitro and in vivo studies. Paired with prior demonstrated abilities for C15:0 to target longevity pathways, hallmarks of aging, aging rate biomarkers, and core components of type 2 diabetes, heart disease, cancer, and nonalcoholic fatty liver disease, our results support C15:0 as an essential nutrient with activities equivalent to, or surpassing, leading longevity-enhancing candidate compounds.
Collapse
Affiliation(s)
- Stephanie Venn-Watson
- Epitracker Inc., San Diego, CA 92106, USA
- Seraphina Therapeutics, Inc., San Diego, CA 92106, USA;
| | - Nicholas J. Schork
- Seraphina Therapeutics, Inc., San Diego, CA 92106, USA;
- Translational Genomics Research Institute (TGen), City of Hope, Phoenix, AZ 85004, USA
| |
Collapse
|
14
|
Takasugi M, Yoshida Y, Nonaka Y, Ohtani N. Gene expressions associated with longer lifespan and aging exhibit similarity in mammals. Nucleic Acids Res 2023; 51:7205-7219. [PMID: 37351606 PMCID: PMC10415134 DOI: 10.1093/nar/gkad544] [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: 03/09/2023] [Revised: 05/08/2023] [Accepted: 06/13/2023] [Indexed: 06/24/2023] Open
Abstract
Although molecular features underlying aging and species maximum lifespan (MLS) have been comprehensively studied by transcriptome analyses, the actual impact of transcriptome on aging and MLS remains elusive. Here, we found that transcriptional signatures that are associated with mammalian MLS exhibited significant similarity to those of aging. Moreover, transcriptional signatures of longer MLS and aging both exhibited significant similarity to that of longer-lived mouse strains, suggesting that gene expression patterns associated with species MLS contribute to extended lifespan even within a species and that aging-related gene expression changes overall represent adaptations that extend lifespan rather than deterioration. Finally, we found evidence of co-evolution of MLS and promoter sequences of MLS-associated genes, highlighting the evolutionary contribution of specific transcription factor binding motifs such as that of E2F1 in shaping MLS-associated gene expression signature. Our results highlight the importance of focusing on adaptive aspects of aging transcriptome and demonstrate that cross-species genomics can be a powerful approach for understanding adaptive aging transcriptome.
Collapse
Affiliation(s)
- Masaki Takasugi
- Department of Pathophysiology, Osaka Metropolitan University, Graduate School of Medicine, Osaka, Japan
| | - Yuya Yoshida
- Department of Pathophysiology, Osaka Metropolitan University, Graduate School of Medicine, Osaka, Japan
| | - Yoshiki Nonaka
- Department of Pathophysiology, Osaka Metropolitan University, Graduate School of Medicine, Osaka, Japan
| | - Naoko Ohtani
- Department of Pathophysiology, Osaka Metropolitan University, Graduate School of Medicine, Osaka, Japan
| |
Collapse
|
15
|
Lu Y, Pike JR, Kucharska-Newton AM, Palta P, Whitsel EA, Bey GS, Zannas AS, Windham BG, Walker KA, Griswold M, Heiss G. Aging-Related Multisystem Dysregulation Over the Adult Life Span and Physical Function in Later Life: The Atherosclerosis Risk in Communities (ARIC) Study. J Gerontol A Biol Sci Med Sci 2023; 78:1497-1503. [PMID: 36453688 PMCID: PMC10395554 DOI: 10.1093/gerona/glac236] [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: 07/21/2022] [Indexed: 08/03/2023] Open
Abstract
BACKGROUND Multisystem dysregulation (Dm) shows promise as a metric of aging and predicts mortality. However, Dm needs to be studied with less severe endpoints indicating modifiable aging stages. Physical function, reflecting healthy longevity rather than just longevity, is more relevant to the goals of geroscience but has not been well investigated. METHODS We tested the association of midlife Dm and its change over ~20 years with physical function in later life in 5 583 the Atherosclerosis Risk in Communities Study cohort participants (baseline mean age 54.7). Dm quantifies the multivariate statistical deviation of 17 physiologically motivated biomarkers relative to their distribution in a young healthy sample at baseline. Physical function was assessed from grip strength and the Short Physical Performance Battery (SPPB). Associations were quantified using linear regression and ordinal logistic regression adjusting for age, sex, race, and education. RESULTS Each unit increment in midlife Dm was associated with 1.71 times the odds of having a lower SPPB score. Compared to the first quartile of midlife Dm, the odds ratios of having a lower SPPB score were 1.25, 1.56, and 2.45, respectively, for the second-fourth quartiles. Similar graded association patterns were observed for each SPPB component test and grip strength. An inverse monotonic relationship also was observed between the annual growth rate of Dm and physical function. CONCLUSION Greater Dm and progression in midlife were associated with lower physical function in later life. Future studies on the factors that lead to the progression of Dm may highlight opportunities to preserve physical function.
Collapse
Affiliation(s)
- Yifei Lu
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - James R Pike
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Anna M Kucharska-Newton
- Department of Epidemiology, College of Public Health, University of Kentucky, Lexington, Kentucky, USA
| | - Priya Palta
- Departments of Medicine and Epidemiology, Columbia University Irving Medical Center, New York, New York, USA
- Department of Neurology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Eric A Whitsel
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Ganga S Bey
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Anthony S Zannas
- Department of Psychiatry, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - B Gwen Windham
- School of Medicine, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Keenan A Walker
- Laboratory of Behavioral Neuroscience, National Institute on Aging, Baltimore, Maryland, USA
| | - Michael Griswold
- School of Medicine, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Gerardo Heiss
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| |
Collapse
|
16
|
Hepowit NL, Blalock E, Lee S, Bretland KM, MacGurn JA, Dickson RC. Reduced sphingolipid biosynthesis modulates proteostasis networks to enhance longevity. Aging (Albany NY) 2023; 15:472-491. [PMID: 36640272 PMCID: PMC9925692 DOI: 10.18632/aging.204485] [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: 08/25/2022] [Accepted: 12/29/2022] [Indexed: 01/15/2023]
Abstract
As the elderly population increases, chronic, age-associated diseases are challenging healthcare systems around the world. Nutrient limitation is well known to slow the aging process and improve health. Regrettably, practicing nutrient restriction to improve health is unachievable for most people. Alternatively, pharmacological strategies are being pursued including myriocin which increases lifespan in budding yeast. Myriocin impairs sphingolipid synthesis, resulting in lowered amino acid pools which promote entry into a quiescent, long-lived state. Here we present transcriptomic data during the first 6 hours of drug treatment that improves our mechanistic understanding of the cellular response to myriocin and reveals a new role for ubiquitin in longevity. Previously we found that the methionine transporter Mup1 traffics to the plasma membrane normally in myriocin-treated cells but is not active and undergoes endocytic clearance. We now show that UBI4, a gene encoding stressed-induced ubiquitin, is vital for myriocin-enhanced lifespan. Furthermore, we show that Mup1 fused to a deubiquitinase domain impairs myriocin-enhanced longevity. Broader effects of myriocin treatment on ubiquitination are indicated by our finding of a significant increase in K63-linked ubiquitin polymers following myriocin treatment. Although proteostasis is broadly accepted as a pillar of aging, our finding that ubiquitination of an amino acid transporter promotes longevity in myriocin-treated cells is novel. Addressing the role of ubiquitination/deubiquitination in longevity has the potential to reveal new strategies and targets for promoting healthy aging.
Collapse
Affiliation(s)
- Nathaniel L. Hepowit
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN 37240, USA
| | - Eric Blalock
- Department of Pharmacology and Nutritional Science, University of Kentucky, Lexington, KY 40536, USA
| | - Sangderk Lee
- College of Pharmacy, University of Kentucky, Lexington, KY 40536, USA
| | - Kimberly M. Bretland
- Department of Pharmacology and Nutritional Science, University of Kentucky, Lexington, KY 40536, USA
| | - Jason A. MacGurn
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN 37240, USA
| | - Robert C. Dickson
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, KY 40536, USA
| |
Collapse
|
17
|
Brinkley TE, Justice JN, Basu S, Bauer SR, Loh KP, Mukli P, Ng TKS, Turney IC, Ferrucci L, Cummings SR, Kritchevsky SB. Research priorities for measuring biologic age: summary and future directions from the Research Centers Collaborative Network Workshop. GeroScience 2022; 44:2573-2583. [PMID: 36242692 PMCID: PMC9768050 DOI: 10.1007/s11357-022-00661-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Accepted: 09/09/2022] [Indexed: 01/07/2023] Open
Abstract
Biologic aging reflects the genetic, molecular, and cellular changes underlying the development of morbidity and mortality with advancing chronological age. As several potential mechanisms have been identified, there is a growing interest in developing robust measures of biologic age that can better reflect the underlying biology of aging and predict age-related outcomes. To support this endeavor, the Research Centers Collaborative Network (RCCN) conducted a workshop in January 2022 to discuss emerging concepts in the field and identify opportunities to move the science forward. This paper presents workshop proceedings and summarizes the identified research needs, priorities, and recommendations for measuring biologic age. The highest priorities identified were the need for more robust measures, longitudinal studies, multidisciplinary collaborations, and translational approaches.
Collapse
Affiliation(s)
- Tina E Brinkley
- Department of Internal Medicine, Section on Gerontology and Geriatric Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, USA.
| | - Jamie N Justice
- Department of Internal Medicine, Section on Gerontology and Geriatric Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Shubhashrita Basu
- Center for Demography of Health and Aging, University of Wisconsin, Madison, WI, USA
| | - Scott R Bauer
- Departments of Medicine and Urology, University of California and San Francisco VA Medical Center, San Francisco, CA, USA
| | - Kah Poh Loh
- Division of Hematology/Oncology, Department of Medicine, University of Rochester Medical Center, James P. Wilmot Cancer Institute, Rochester, NY, USA
| | - Peter Mukli
- Department of Biochemistry/Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Ted Kheng Siang Ng
- Edson College of Nursing and Health Innovation, Arizona State University, Tempe, AZ, USA
| | - Indira C Turney
- Department of Neurology, Columbia University Medical Center, New York City, NY, USA
| | - Luigi Ferrucci
- Intramural Research Program of the National Institute On Aging, NIH, Baltimore, MD, USA
| | - Steven R Cummings
- San Francisco Coordinating Center, California Pacific Medical Center Research Institute and the Department of Epidemiology and Biostatistics, University of California, San Francisco, CA, USA
| | - Stephen B Kritchevsky
- Department of Internal Medicine, Section on Gerontology and Geriatric Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| |
Collapse
|
18
|
Farrell S, Kane AE, Bisset E, Howlett SE, Rutenberg AD. Measurements of damage and repair of binary health attributes in aging mice and humans reveal that robustness and resilience decrease with age, operate over broad timescales, and are affected differently by interventions. eLife 2022; 11:e77632. [PMID: 36409200 PMCID: PMC9725749 DOI: 10.7554/elife.77632] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2022] [Accepted: 11/20/2022] [Indexed: 11/23/2022] Open
Abstract
As an organism ages, its health-state is determined by a balance between the processes of damage and repair. Measuring these processes requires longitudinal data. We extract damage and repair transition rates from repeated observations of binary health attributes in mice and humans to explore robustness and resilience, which respectively represent resisting or recovering from damage. We assess differences in robustness and resilience using changes in damage rates and repair rates of binary health attributes. We find a conserved decline with age in robustness and resilience in mice and humans, implying that both contribute to worsening aging health - as assessed by the frailty index (FI). A decline in robustness, however, has a greater effect than a decline in resilience on the accelerated increase of the FI with age, and a greater association with reduced survival. We also find that deficits are damaged and repaired over a wide range of timescales ranging from the shortest measurement scales toward organismal lifetime timescales. We explore the effect of systemic interventions that have been shown to improve health, including the angiotensin-converting enzyme inhibitor enalapril and voluntary exercise for mice. We have also explored the correlations with household wealth for humans. We find that these interventions and factors affect both damage and repair rates, and hence robustness and resilience, in age and sex-dependent manners.
Collapse
Affiliation(s)
| | - Alice E Kane
- Blavatnik Institute, Department of Genetics, Paul F. Glenn Center for Biology of Aging Research at Harvard Medical SchoolBostonUnited States
| | - Elise Bisset
- Department of Pharmacology, Dalhousie UniversityHalifaxCanada
| | - Susan E Howlett
- Department of Pharmacology, Dalhousie UniversityHalifaxCanada
- Department of Medicine (GeriatricMedicine), Dalhousie UniversityHalifaxCanada
| | - Andrew D Rutenberg
- Department of Physics and Atmospheric Science, Dalhousie UniversityHalifaxCanada
| |
Collapse
|
19
|
Effects of lifespan-extending interventions on cognitive healthspan. Expert Rev Mol Med 2022; 25:e2. [PMID: 36377361 DOI: 10.1017/erm.2022.36] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Ageing is known to be the primary risk factor for most neurodegenerative diseases, including Alzheimer's disease, Parkinson's disease and Huntington's disease. They are currently incurable and worsen over time, which has broad implications in the context of lifespan and healthspan extension. Adding years to life and even to physical health is suboptimal or even insufficient, if cognitive ageing is not adequately improved. In this review, we will examine how interventions that have the potential to extend lifespan in animals affect the brain, and if they would be able to thwart or delay the development of cognitive dysfunction and/or neurodegeneration. These interventions range from lifestyle (caloric restriction, physical exercise and environmental enrichment) through pharmacological (nicotinamide adenine dinucleotide precursors, resveratrol, rapamycin, metformin, spermidine and senolytics) to epigenetic reprogramming. We argue that while many of these interventions have clear potential to improve cognitive health and resilience, large-scale and long-term randomised controlled trials are needed, along with studies utilising washout periods to determine the effects of supplementation cessation, particularly in aged individuals.
Collapse
|
20
|
Birkisdóttir MB, van Galen I, Brandt RMC, Barnhoorn S, van Vliet N, van Dijk C, Nagarajah B, Imholz S, van Oostrom CT, Reiling E, Gyenis Á, Mastroberardino PG, Jaarsma D, van Steeg H, Hoeijmakers JHJ, Dollé MET, Vermeij WP. The use of progeroid DNA repair-deficient mice for assessing anti-aging compounds, illustrating the benefits of nicotinamide riboside. FRONTIERS IN AGING 2022; 3:1005322. [PMID: 36313181 PMCID: PMC9596940 DOI: 10.3389/fragi.2022.1005322] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Accepted: 09/14/2022] [Indexed: 11/06/2022]
Abstract
Despite efficient repair, DNA damage inevitably accumulates with time affecting proper cell function and viability, thereby driving systemic aging. Interventions that either prevent DNA damage or enhance DNA repair are thus likely to extend health- and lifespan across species. However, effective genome-protecting compounds are largely lacking. Here, we use Ercc1 Δ/- and Xpg -/- DNA repair-deficient mutants as two bona fide accelerated aging mouse models to test propitious anti-aging pharmaceutical interventions. Ercc1 Δ/- and Xpg -/- mice show shortened lifespan with accelerated aging across numerous organs and tissues. Previously, we demonstrated that a well-established anti-aging intervention, dietary restriction, reduced DNA damage, and dramatically improved healthspan, strongly extended lifespan, and delayed all aging pathology investigated. Here, we further utilize the short lifespan and early onset of signs of neurological degeneration in Ercc1 Δ/- and Xpg -/- mice to test compounds that influence nutrient sensing (metformin, acarbose, resveratrol), inflammation (aspirin, ibuprofen), mitochondrial processes (idebenone, sodium nitrate, dichloroacetate), glucose homeostasis (trehalose, GlcNAc) and nicotinamide adenine dinucleotide (NAD+) metabolism. While some of the compounds have shown anti-aging features in WT animals, most of them failed to significantly alter lifespan or features of neurodegeneration of our mice. The two NAD+ precursors; nicotinamide riboside (NR) and nicotinic acid (NA), did however induce benefits, consistent with the role of NAD+ in facilitating DNA damage repair. Together, our results illustrate the applicability of short-lived repair mutants for systematic screening of anti-aging interventions capable of reducing DNA damage accumulation.
Collapse
Affiliation(s)
- María B. Birkisdóttir
- Princess Máxima Center for Pediatric Oncology, Utrecht, Netherlands,Oncode Institute, Utrecht, Netherlands
| | - Ivar van Galen
- Princess Máxima Center for Pediatric Oncology, Utrecht, Netherlands,Oncode Institute, Utrecht, Netherlands
| | - Renata M. C. Brandt
- Department of Molecular Genetics, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Sander Barnhoorn
- Department of Molecular Genetics, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Nicole van Vliet
- Department of Molecular Genetics, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Claire van Dijk
- Department of Molecular Genetics, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, Netherlands,Department of Hematology, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Bhawani Nagarajah
- Centre for Health Protection, National Institute for Public Health and the Environment, (RIVM), Bilthoven, Netherlands
| | - Sandra Imholz
- Centre for Health Protection, National Institute for Public Health and the Environment, (RIVM), Bilthoven, Netherlands
| | - Conny T. van Oostrom
- Centre for Health Protection, National Institute for Public Health and the Environment, (RIVM), Bilthoven, Netherlands
| | - Erwin Reiling
- Centre for Health Protection, National Institute for Public Health and the Environment, (RIVM), Bilthoven, Netherlands
| | - Ákos Gyenis
- Faculty of Medicine, CECAD, Institute for Genome Stability in Aging and Disease, University of Cologne, Cologne, Germany
| | - Pier G. Mastroberardino
- Department of Molecular Genetics, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, Netherlands,IFOM-The FIRC Institute of Molecular Oncology, Milan, Italy,Department of Life, Health, and Environmental Sciences, University of L'Aquila, L'Aquila, Italy
| | - Dick Jaarsma
- Department of Neuroscience, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Harry van Steeg
- Centre for Health Protection, National Institute for Public Health and the Environment, (RIVM), Bilthoven, Netherlands
| | - Jan H. J. Hoeijmakers
- Princess Máxima Center for Pediatric Oncology, Utrecht, Netherlands,Oncode Institute, Utrecht, Netherlands,Department of Molecular Genetics, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, Netherlands,Faculty of Medicine, CECAD, Institute for Genome Stability in Aging and Disease, University of Cologne, Cologne, Germany
| | - Martijn E. T. Dollé
- Centre for Health Protection, National Institute for Public Health and the Environment, (RIVM), Bilthoven, Netherlands,*Correspondence: Wilbert P. Vermeij, ; Martijn E. T. Dollé,
| | - Wilbert P. Vermeij
- Princess Máxima Center for Pediatric Oncology, Utrecht, Netherlands,Oncode Institute, Utrecht, Netherlands,*Correspondence: Wilbert P. Vermeij, ; Martijn E. T. Dollé,
| |
Collapse
|
21
|
Sugden K, Caspi A, Elliott ML, Bourassa KJ, Chamarti K, Corcoran DL, Hariri AR, Houts RM, Kothari M, Kritchevsky S, Kuchel GA, Mill JS, Williams BS, Belsky DW, Moffitt TE. Association of Pace of Aging Measured by Blood-Based DNA Methylation With Age-Related Cognitive Impairment and Dementia. Neurology 2022; 99:e1402-e1413. [PMID: 35794023 PMCID: PMC9576288 DOI: 10.1212/wnl.0000000000200898] [Citation(s) in RCA: 56] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 05/13/2022] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND AND OBJECTIVES DNA methylation algorithms are increasingly used to estimate biological aging; however, how these proposed measures of whole-organism biological aging relate to aging in the brain is not known. We used data from the Alzheimer's Disease Neuroimaging Initiative (ADNI) and the Framingham Heart Study (FHS) Offspring Cohort to test the association between blood-based DNA methylation measures of biological aging and cognitive impairment and dementia in older adults. METHODS We tested 3 "generations" of DNA methylation age algorithms (first generation: Horvath and Hannum clocks; second generation: PhenoAge and GrimAge; and third generation: DunedinPACE, Dunedin Pace of Aging Calculated from the Epigenome) against the following measures of cognitive impairment in ADNI: clinical diagnosis of dementia and mild cognitive impairment, scores on Alzheimer disease (AD) / Alzheimer disease and related dementias (ADRD) screening tests (Alzheimer's Disease Assessment Scale, Mini-Mental State Examination, and Montreal Cognitive Assessment), and scores on cognitive tests (Rey Auditory Verbal Learning Test, Logical Memory test, and Trail Making Test). In an independent replication in the FHS Offspring Cohort, we further tested the longitudinal association between the DNA methylation algorithms and the risk of developing dementia. RESULTS In ADNI (N = 649 individuals), the first-generation (Horvath and Hannum DNA methylation age clocks) and the second-generation (PhenoAge and GrimAge) DNA methylation measures of aging were not consistently associated with measures of cognitive impairment in older adults. By contrast, a third-generation measure of biological aging, DunedinPACE, was associated with clinical diagnosis of Alzheimer disease (beta [95% CI] = 0.28 [0.08-0.47]), poorer scores on Alzheimer disease/ADRD screening tests (beta [Robust SE] = -0.10 [0.04] to 0.08[0.04]), and cognitive tests (beta [Robust SE] = -0.12 [0.04] to 0.10 [0.03]). The association between faster pace of aging, as measured by DunedinPACE, and risk of developing dementia was confirmed in a longitudinal analysis of the FHS Offspring Cohort (N = 2,264 individuals, hazard ratio [95% CI] = 1.27 [1.07-1.49]). DISCUSSION Third-generation blood-based DNA methylation measures of aging could prove valuable for measuring differences between individuals in the rate at which they age and in their risk for cognitive decline, and for evaluating interventions to slow aging.
Collapse
Affiliation(s)
- Karen Sugden
- From the Department of Psychology and Neuroscience (K.S., A.C., M.L.E., K.C., A.R.H., R.M.H., B.S.W., T.E.M.), and Center for Genomic and Computational Biology (K.S., A.C., B.S.W., T.E.M.), Duke University, Durham, NC; Department of Psychiatry and Behavioral Sciences (A.C., T.E.M.), Duke University School of Medicine, Durham, NC; Social, Genetic, and Developmental Psychiatry Centre (A.C, T.E.M.), Institute of Psychiatry, Psychology, and Neuroscience, King's College London, UK. Center for the Study of Aging and Human Development (K.J.B.), Duke University, Durham, NC; Department of Genetics (D.L.C.), University of North Carolina School of Medicine, Chapel Hill; Butler Columbia Aging Center (M.K., D.W.B.), Columbia University, New York, New York; Sticht Center for Healthy Aging and Alzheimer's Prevention (S.K.), Wake Forest School of Medicine, Winston-Salem, NC; UConn Center on Aging (G.A.K.), University of Connecticut, Farmington, Connecticut, USA; College of Medicine and Health (J.S.M.), University of Exeter Medical School, Devon, UK; and Department of Epidemiology (D.W.B.), Columbia University Mailman School of Public Health, New York, New York.
| | - Avshalom Caspi
- From the Department of Psychology and Neuroscience (K.S., A.C., M.L.E., K.C., A.R.H., R.M.H., B.S.W., T.E.M.), and Center for Genomic and Computational Biology (K.S., A.C., B.S.W., T.E.M.), Duke University, Durham, NC; Department of Psychiatry and Behavioral Sciences (A.C., T.E.M.), Duke University School of Medicine, Durham, NC; Social, Genetic, and Developmental Psychiatry Centre (A.C, T.E.M.), Institute of Psychiatry, Psychology, and Neuroscience, King's College London, UK. Center for the Study of Aging and Human Development (K.J.B.), Duke University, Durham, NC; Department of Genetics (D.L.C.), University of North Carolina School of Medicine, Chapel Hill; Butler Columbia Aging Center (M.K., D.W.B.), Columbia University, New York, New York; Sticht Center for Healthy Aging and Alzheimer's Prevention (S.K.), Wake Forest School of Medicine, Winston-Salem, NC; UConn Center on Aging (G.A.K.), University of Connecticut, Farmington, Connecticut, USA; College of Medicine and Health (J.S.M.), University of Exeter Medical School, Devon, UK; and Department of Epidemiology (D.W.B.), Columbia University Mailman School of Public Health, New York, New York
| | - Maxwell L Elliott
- From the Department of Psychology and Neuroscience (K.S., A.C., M.L.E., K.C., A.R.H., R.M.H., B.S.W., T.E.M.), and Center for Genomic and Computational Biology (K.S., A.C., B.S.W., T.E.M.), Duke University, Durham, NC; Department of Psychiatry and Behavioral Sciences (A.C., T.E.M.), Duke University School of Medicine, Durham, NC; Social, Genetic, and Developmental Psychiatry Centre (A.C, T.E.M.), Institute of Psychiatry, Psychology, and Neuroscience, King's College London, UK. Center for the Study of Aging and Human Development (K.J.B.), Duke University, Durham, NC; Department of Genetics (D.L.C.), University of North Carolina School of Medicine, Chapel Hill; Butler Columbia Aging Center (M.K., D.W.B.), Columbia University, New York, New York; Sticht Center for Healthy Aging and Alzheimer's Prevention (S.K.), Wake Forest School of Medicine, Winston-Salem, NC; UConn Center on Aging (G.A.K.), University of Connecticut, Farmington, Connecticut, USA; College of Medicine and Health (J.S.M.), University of Exeter Medical School, Devon, UK; and Department of Epidemiology (D.W.B.), Columbia University Mailman School of Public Health, New York, New York
| | - Kyle J Bourassa
- From the Department of Psychology and Neuroscience (K.S., A.C., M.L.E., K.C., A.R.H., R.M.H., B.S.W., T.E.M.), and Center for Genomic and Computational Biology (K.S., A.C., B.S.W., T.E.M.), Duke University, Durham, NC; Department of Psychiatry and Behavioral Sciences (A.C., T.E.M.), Duke University School of Medicine, Durham, NC; Social, Genetic, and Developmental Psychiatry Centre (A.C, T.E.M.), Institute of Psychiatry, Psychology, and Neuroscience, King's College London, UK. Center for the Study of Aging and Human Development (K.J.B.), Duke University, Durham, NC; Department of Genetics (D.L.C.), University of North Carolina School of Medicine, Chapel Hill; Butler Columbia Aging Center (M.K., D.W.B.), Columbia University, New York, New York; Sticht Center for Healthy Aging and Alzheimer's Prevention (S.K.), Wake Forest School of Medicine, Winston-Salem, NC; UConn Center on Aging (G.A.K.), University of Connecticut, Farmington, Connecticut, USA; College of Medicine and Health (J.S.M.), University of Exeter Medical School, Devon, UK; and Department of Epidemiology (D.W.B.), Columbia University Mailman School of Public Health, New York, New York
| | - Kartik Chamarti
- From the Department of Psychology and Neuroscience (K.S., A.C., M.L.E., K.C., A.R.H., R.M.H., B.S.W., T.E.M.), and Center for Genomic and Computational Biology (K.S., A.C., B.S.W., T.E.M.), Duke University, Durham, NC; Department of Psychiatry and Behavioral Sciences (A.C., T.E.M.), Duke University School of Medicine, Durham, NC; Social, Genetic, and Developmental Psychiatry Centre (A.C, T.E.M.), Institute of Psychiatry, Psychology, and Neuroscience, King's College London, UK. Center for the Study of Aging and Human Development (K.J.B.), Duke University, Durham, NC; Department of Genetics (D.L.C.), University of North Carolina School of Medicine, Chapel Hill; Butler Columbia Aging Center (M.K., D.W.B.), Columbia University, New York, New York; Sticht Center for Healthy Aging and Alzheimer's Prevention (S.K.), Wake Forest School of Medicine, Winston-Salem, NC; UConn Center on Aging (G.A.K.), University of Connecticut, Farmington, Connecticut, USA; College of Medicine and Health (J.S.M.), University of Exeter Medical School, Devon, UK; and Department of Epidemiology (D.W.B.), Columbia University Mailman School of Public Health, New York, New York
| | - David L Corcoran
- From the Department of Psychology and Neuroscience (K.S., A.C., M.L.E., K.C., A.R.H., R.M.H., B.S.W., T.E.M.), and Center for Genomic and Computational Biology (K.S., A.C., B.S.W., T.E.M.), Duke University, Durham, NC; Department of Psychiatry and Behavioral Sciences (A.C., T.E.M.), Duke University School of Medicine, Durham, NC; Social, Genetic, and Developmental Psychiatry Centre (A.C, T.E.M.), Institute of Psychiatry, Psychology, and Neuroscience, King's College London, UK. Center for the Study of Aging and Human Development (K.J.B.), Duke University, Durham, NC; Department of Genetics (D.L.C.), University of North Carolina School of Medicine, Chapel Hill; Butler Columbia Aging Center (M.K., D.W.B.), Columbia University, New York, New York; Sticht Center for Healthy Aging and Alzheimer's Prevention (S.K.), Wake Forest School of Medicine, Winston-Salem, NC; UConn Center on Aging (G.A.K.), University of Connecticut, Farmington, Connecticut, USA; College of Medicine and Health (J.S.M.), University of Exeter Medical School, Devon, UK; and Department of Epidemiology (D.W.B.), Columbia University Mailman School of Public Health, New York, New York
| | - Ahmad R Hariri
- From the Department of Psychology and Neuroscience (K.S., A.C., M.L.E., K.C., A.R.H., R.M.H., B.S.W., T.E.M.), and Center for Genomic and Computational Biology (K.S., A.C., B.S.W., T.E.M.), Duke University, Durham, NC; Department of Psychiatry and Behavioral Sciences (A.C., T.E.M.), Duke University School of Medicine, Durham, NC; Social, Genetic, and Developmental Psychiatry Centre (A.C, T.E.M.), Institute of Psychiatry, Psychology, and Neuroscience, King's College London, UK. Center for the Study of Aging and Human Development (K.J.B.), Duke University, Durham, NC; Department of Genetics (D.L.C.), University of North Carolina School of Medicine, Chapel Hill; Butler Columbia Aging Center (M.K., D.W.B.), Columbia University, New York, New York; Sticht Center for Healthy Aging and Alzheimer's Prevention (S.K.), Wake Forest School of Medicine, Winston-Salem, NC; UConn Center on Aging (G.A.K.), University of Connecticut, Farmington, Connecticut, USA; College of Medicine and Health (J.S.M.), University of Exeter Medical School, Devon, UK; and Department of Epidemiology (D.W.B.), Columbia University Mailman School of Public Health, New York, New York
| | - Renate M Houts
- From the Department of Psychology and Neuroscience (K.S., A.C., M.L.E., K.C., A.R.H., R.M.H., B.S.W., T.E.M.), and Center for Genomic and Computational Biology (K.S., A.C., B.S.W., T.E.M.), Duke University, Durham, NC; Department of Psychiatry and Behavioral Sciences (A.C., T.E.M.), Duke University School of Medicine, Durham, NC; Social, Genetic, and Developmental Psychiatry Centre (A.C, T.E.M.), Institute of Psychiatry, Psychology, and Neuroscience, King's College London, UK. Center for the Study of Aging and Human Development (K.J.B.), Duke University, Durham, NC; Department of Genetics (D.L.C.), University of North Carolina School of Medicine, Chapel Hill; Butler Columbia Aging Center (M.K., D.W.B.), Columbia University, New York, New York; Sticht Center for Healthy Aging and Alzheimer's Prevention (S.K.), Wake Forest School of Medicine, Winston-Salem, NC; UConn Center on Aging (G.A.K.), University of Connecticut, Farmington, Connecticut, USA; College of Medicine and Health (J.S.M.), University of Exeter Medical School, Devon, UK; and Department of Epidemiology (D.W.B.), Columbia University Mailman School of Public Health, New York, New York
| | - Meeraj Kothari
- From the Department of Psychology and Neuroscience (K.S., A.C., M.L.E., K.C., A.R.H., R.M.H., B.S.W., T.E.M.), and Center for Genomic and Computational Biology (K.S., A.C., B.S.W., T.E.M.), Duke University, Durham, NC; Department of Psychiatry and Behavioral Sciences (A.C., T.E.M.), Duke University School of Medicine, Durham, NC; Social, Genetic, and Developmental Psychiatry Centre (A.C, T.E.M.), Institute of Psychiatry, Psychology, and Neuroscience, King's College London, UK. Center for the Study of Aging and Human Development (K.J.B.), Duke University, Durham, NC; Department of Genetics (D.L.C.), University of North Carolina School of Medicine, Chapel Hill; Butler Columbia Aging Center (M.K., D.W.B.), Columbia University, New York, New York; Sticht Center for Healthy Aging and Alzheimer's Prevention (S.K.), Wake Forest School of Medicine, Winston-Salem, NC; UConn Center on Aging (G.A.K.), University of Connecticut, Farmington, Connecticut, USA; College of Medicine and Health (J.S.M.), University of Exeter Medical School, Devon, UK; and Department of Epidemiology (D.W.B.), Columbia University Mailman School of Public Health, New York, New York
| | - Stephen Kritchevsky
- From the Department of Psychology and Neuroscience (K.S., A.C., M.L.E., K.C., A.R.H., R.M.H., B.S.W., T.E.M.), and Center for Genomic and Computational Biology (K.S., A.C., B.S.W., T.E.M.), Duke University, Durham, NC; Department of Psychiatry and Behavioral Sciences (A.C., T.E.M.), Duke University School of Medicine, Durham, NC; Social, Genetic, and Developmental Psychiatry Centre (A.C, T.E.M.), Institute of Psychiatry, Psychology, and Neuroscience, King's College London, UK. Center for the Study of Aging and Human Development (K.J.B.), Duke University, Durham, NC; Department of Genetics (D.L.C.), University of North Carolina School of Medicine, Chapel Hill; Butler Columbia Aging Center (M.K., D.W.B.), Columbia University, New York, New York; Sticht Center for Healthy Aging and Alzheimer's Prevention (S.K.), Wake Forest School of Medicine, Winston-Salem, NC; UConn Center on Aging (G.A.K.), University of Connecticut, Farmington, Connecticut, USA; College of Medicine and Health (J.S.M.), University of Exeter Medical School, Devon, UK; and Department of Epidemiology (D.W.B.), Columbia University Mailman School of Public Health, New York, New York
| | - George A Kuchel
- From the Department of Psychology and Neuroscience (K.S., A.C., M.L.E., K.C., A.R.H., R.M.H., B.S.W., T.E.M.), and Center for Genomic and Computational Biology (K.S., A.C., B.S.W., T.E.M.), Duke University, Durham, NC; Department of Psychiatry and Behavioral Sciences (A.C., T.E.M.), Duke University School of Medicine, Durham, NC; Social, Genetic, and Developmental Psychiatry Centre (A.C, T.E.M.), Institute of Psychiatry, Psychology, and Neuroscience, King's College London, UK. Center for the Study of Aging and Human Development (K.J.B.), Duke University, Durham, NC; Department of Genetics (D.L.C.), University of North Carolina School of Medicine, Chapel Hill; Butler Columbia Aging Center (M.K., D.W.B.), Columbia University, New York, New York; Sticht Center for Healthy Aging and Alzheimer's Prevention (S.K.), Wake Forest School of Medicine, Winston-Salem, NC; UConn Center on Aging (G.A.K.), University of Connecticut, Farmington, Connecticut, USA; College of Medicine and Health (J.S.M.), University of Exeter Medical School, Devon, UK; and Department of Epidemiology (D.W.B.), Columbia University Mailman School of Public Health, New York, New York
| | - Jonathan S Mill
- From the Department of Psychology and Neuroscience (K.S., A.C., M.L.E., K.C., A.R.H., R.M.H., B.S.W., T.E.M.), and Center for Genomic and Computational Biology (K.S., A.C., B.S.W., T.E.M.), Duke University, Durham, NC; Department of Psychiatry and Behavioral Sciences (A.C., T.E.M.), Duke University School of Medicine, Durham, NC; Social, Genetic, and Developmental Psychiatry Centre (A.C, T.E.M.), Institute of Psychiatry, Psychology, and Neuroscience, King's College London, UK. Center for the Study of Aging and Human Development (K.J.B.), Duke University, Durham, NC; Department of Genetics (D.L.C.), University of North Carolina School of Medicine, Chapel Hill; Butler Columbia Aging Center (M.K., D.W.B.), Columbia University, New York, New York; Sticht Center for Healthy Aging and Alzheimer's Prevention (S.K.), Wake Forest School of Medicine, Winston-Salem, NC; UConn Center on Aging (G.A.K.), University of Connecticut, Farmington, Connecticut, USA; College of Medicine and Health (J.S.M.), University of Exeter Medical School, Devon, UK; and Department of Epidemiology (D.W.B.), Columbia University Mailman School of Public Health, New York, New York
| | - Benjamin S Williams
- From the Department of Psychology and Neuroscience (K.S., A.C., M.L.E., K.C., A.R.H., R.M.H., B.S.W., T.E.M.), and Center for Genomic and Computational Biology (K.S., A.C., B.S.W., T.E.M.), Duke University, Durham, NC; Department of Psychiatry and Behavioral Sciences (A.C., T.E.M.), Duke University School of Medicine, Durham, NC; Social, Genetic, and Developmental Psychiatry Centre (A.C, T.E.M.), Institute of Psychiatry, Psychology, and Neuroscience, King's College London, UK. Center for the Study of Aging and Human Development (K.J.B.), Duke University, Durham, NC; Department of Genetics (D.L.C.), University of North Carolina School of Medicine, Chapel Hill; Butler Columbia Aging Center (M.K., D.W.B.), Columbia University, New York, New York; Sticht Center for Healthy Aging and Alzheimer's Prevention (S.K.), Wake Forest School of Medicine, Winston-Salem, NC; UConn Center on Aging (G.A.K.), University of Connecticut, Farmington, Connecticut, USA; College of Medicine and Health (J.S.M.), University of Exeter Medical School, Devon, UK; and Department of Epidemiology (D.W.B.), Columbia University Mailman School of Public Health, New York, New York
| | - Daniel W Belsky
- From the Department of Psychology and Neuroscience (K.S., A.C., M.L.E., K.C., A.R.H., R.M.H., B.S.W., T.E.M.), and Center for Genomic and Computational Biology (K.S., A.C., B.S.W., T.E.M.), Duke University, Durham, NC; Department of Psychiatry and Behavioral Sciences (A.C., T.E.M.), Duke University School of Medicine, Durham, NC; Social, Genetic, and Developmental Psychiatry Centre (A.C, T.E.M.), Institute of Psychiatry, Psychology, and Neuroscience, King's College London, UK. Center for the Study of Aging and Human Development (K.J.B.), Duke University, Durham, NC; Department of Genetics (D.L.C.), University of North Carolina School of Medicine, Chapel Hill; Butler Columbia Aging Center (M.K., D.W.B.), Columbia University, New York, New York; Sticht Center for Healthy Aging and Alzheimer's Prevention (S.K.), Wake Forest School of Medicine, Winston-Salem, NC; UConn Center on Aging (G.A.K.), University of Connecticut, Farmington, Connecticut, USA; College of Medicine and Health (J.S.M.), University of Exeter Medical School, Devon, UK; and Department of Epidemiology (D.W.B.), Columbia University Mailman School of Public Health, New York, New York
| | - Terrie E Moffitt
- From the Department of Psychology and Neuroscience (K.S., A.C., M.L.E., K.C., A.R.H., R.M.H., B.S.W., T.E.M.), and Center for Genomic and Computational Biology (K.S., A.C., B.S.W., T.E.M.), Duke University, Durham, NC; Department of Psychiatry and Behavioral Sciences (A.C., T.E.M.), Duke University School of Medicine, Durham, NC; Social, Genetic, and Developmental Psychiatry Centre (A.C, T.E.M.), Institute of Psychiatry, Psychology, and Neuroscience, King's College London, UK. Center for the Study of Aging and Human Development (K.J.B.), Duke University, Durham, NC; Department of Genetics (D.L.C.), University of North Carolina School of Medicine, Chapel Hill; Butler Columbia Aging Center (M.K., D.W.B.), Columbia University, New York, New York; Sticht Center for Healthy Aging and Alzheimer's Prevention (S.K.), Wake Forest School of Medicine, Winston-Salem, NC; UConn Center on Aging (G.A.K.), University of Connecticut, Farmington, Connecticut, USA; College of Medicine and Health (J.S.M.), University of Exeter Medical School, Devon, UK; and Department of Epidemiology (D.W.B.), Columbia University Mailman School of Public Health, New York, New York
| |
Collapse
|
22
|
Hu Y, Zhou J, Cao Y, Zhang J, Zou L. Anti-aging effects of polysaccharides from quinoa (Chenopodium quinoa Willd.) in improving memory and cognitive function. J Funct Foods 2022. [DOI: 10.1016/j.jff.2022.105097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
|
23
|
Longitudinal phenotypic aging metrics in the Baltimore Longitudinal Study of Aging. NATURE AGING 2022; 2:635-643. [PMID: 36910594 PMCID: PMC9997119 DOI: 10.1038/s43587-022-00243-7] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
To define metrics of phenotypic aging, it is essential to identify biological and environmental factors that influence the pace of aging. Previous attempts to develop aging metrics were hampered by cross-sectional designs and/or focused on younger populations. In the Baltimore Longitudinal Study of Aging (BLSA), we collected longitudinally across the adult age range a comprehensive list of phenotypes within four domains (body composition, energetics, homeostatic mechanisms and neurodegeneration/neuroplasticity) and functional outcomes. We integrated individual deviations from population trajectories into a global longitudinal phenotypic metric of aging and demonstrate that accelerated longitudinal phenotypic aging is associated with faster physical and cognitive decline, faster accumulation of multimorbidity and shorter survival. These associations are more robust compared with the use of phenotypic and epigenetic measurements at a single time point. Estimation of these metrics required repeated measures of multiple phenotypes over time but may uniquely facilitate the identification of mechanisms driving phenotypic aging and subsequent age-related functional decline.
Collapse
|
24
|
Jayarathne HSM, Debarba LK, Jaboro JJ, Ginsburg BC, Miller RA, Sadagurski M. Neuroprotective effects of Canagliflozin: Lessons from aged genetically diverse UM-HET3 mice. Aging Cell 2022; 21:e13653. [PMID: 35707855 PMCID: PMC9282842 DOI: 10.1111/acel.13653] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 05/13/2022] [Accepted: 05/25/2022] [Indexed: 01/24/2023] Open
Abstract
The aging brain is characterized by progressive increases in neuroinflammation and central insulin resistance, which contribute to neurodegenerative diseases and cognitive impairment. Recently, the Interventions Testing Program demonstrated that the anti-diabetes drug, Canagliflozin (Cana), a sodium-glucose transporter 2 inhibitor, led to lower fasting glucose and improved glucose tolerance in both sexes, but extended median lifespan by 14% in male mice only. Here, we show that Cana treatment significantly improved central insulin sensitivity in the hypothalamus and the hippocampus of 30-month-old male mice. Aged males produce more robust neuroimmune responses than aged females. Remarkably, Cana-treated male and female mice showed significant reductions in age-associated hypothalamic gliosis with a decrease in inflammatory cytokine production by microglia. However, in the hippocampus, Cana reduced microgliosis and astrogliosis in males, but not in female mice. The decrease in microgliosis was partially correlated with reduced phosphorylation of S6 kinase in microglia of Cana-treated aged male, but not female mice. Thus, Cana treatment improved insulin responsiveness in aged male mice. Furthermore, Cana treatment improved exploratory and locomotor activity of 30-month-old male but not female mice. Taken together, we demonstrate the sex-specific neuroprotective effects of Cana treatment, suggesting its application for the potential treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- Hashan S. M. Jayarathne
- Department of Biological Sciences, IBio (Integrative Biosciences Center)Wayne State UniversityDetroitMichiganUSA
| | - Lucas K. Debarba
- Department of Biological Sciences, IBio (Integrative Biosciences Center)Wayne State UniversityDetroitMichiganUSA
| | - Jacob J. Jaboro
- Department of Biological Sciences, IBio (Integrative Biosciences Center)Wayne State UniversityDetroitMichiganUSA
| | - Brett C. Ginsburg
- Department of Psychiatry and Behavioral SciencesUniversity of Texas Health Science CenterSan AntonioTexasUSA
| | - Richard A. Miller
- Department of Pathology and Geriatrics CenterUniversity of MichiganAnn ArborMichiganUSA
| | - Marianna Sadagurski
- Department of Biological Sciences, IBio (Integrative Biosciences Center)Wayne State UniversityDetroitMichiganUSA
| |
Collapse
|
25
|
Norman P, Exeter D, Shelton N, Head J, Murray E. (Un-) healthy ageing: Geographic inequalities in disability-free life expectancy in England and Wales. Health Place 2022; 76:102820. [PMID: 35690019 DOI: 10.1016/j.healthplace.2022.102820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 04/08/2022] [Accepted: 05/03/2022] [Indexed: 11/04/2022]
Abstract
Health expectancies are an indicator of healthy ageing that reflect quantity and quality of life. Using limiting long term illness and mortality prevalence, we calculate disability-free life expectancy for small areas in England and Wales between 1991 and 2011 for males and females aged 50-74, the life stage when people may be changing their occupation from main career to retirement or alternative work activities. We find that inequalities in disability-free life expectancy are deeply entrenched, including former coalfield and ex-industrial areas and that areas of persistent (dis-) advantage, worsening or improving deprivation have health change in line with deprivation change. A mixed health picture for rural and coastal areas requires further investigation as do the demographic processes which underpin these area level health differences.
Collapse
Affiliation(s)
- Paul Norman
- School of Geography, University of Leeds, UK.
| | - Dan Exeter
- School of Population Health, University of Auckland, New Zealand
| | - Nicola Shelton
- Research Department of Epidemiology and Public Health, University College London, UK
| | - Jenny Head
- Research Department of Epidemiology and Public Health, University College London, UK
| | - Emily Murray
- Research Department of Epidemiology and Public Health, University College London, UK
| |
Collapse
|
26
|
Acosta-Rodríguez V, Rijo-Ferreira F, Izumo M, Xu P, Wight-Carter M, Green CB, Takahashi JS. Circadian alignment of early onset caloric restriction promotes longevity in male C57BL/6J mice. Science 2022; 376:1192-1202. [PMID: 35511946 PMCID: PMC9262309 DOI: 10.1126/science.abk0297] [Citation(s) in RCA: 184] [Impact Index Per Article: 61.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Caloric restriction (CR) prolongs lifespan, yet the mechanisms by which it does so remain poorly understood. Under CR, mice self-impose chronic cycles of 2-hour-feeding and 22-hour-fasting, raising the question whether calories, fasting, or time of day are causal. We show that 30%-CR is sufficient to extend lifespan 10%; however, a daily fasting interval and circadian-alignment of feeding act together to extend lifespan 35% in male C57BL/6J mice. These effects are independent of body weight. Aging induces widespread increases in gene expression associated with inflammation and decreases in expression of genes encoding components of metabolic pathways in liver from ad lib fed mice. CR at night ameliorates these aging-related changes. Thus, circadian interventions promote longevity and provide a perspective to further explore mechanisms of aging. Timed caloric restriction at night enhances longevity.
Collapse
Affiliation(s)
- Victoria Acosta-Rodríguez
- Department of Neuroscience, Peter O'Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Filipa Rijo-Ferreira
- Department of Neuroscience, Peter O'Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.,Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Mariko Izumo
- Department of Neuroscience, Peter O'Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Pin Xu
- Department of Neuroscience, Peter O'Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Mary Wight-Carter
- Animal Resources Center, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Carla B Green
- Department of Neuroscience, Peter O'Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Joseph S Takahashi
- Department of Neuroscience, Peter O'Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.,Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| |
Collapse
|
27
|
Yanai H, Dunn C, Park B, Coletta C, McDevitt RA, McNeely T, Leone M, Wersto RP, Perdue KA, Beerman I. Male rat leukocyte population dynamics predict a window for intervention in aging. eLife 2022; 11:76808. [PMID: 35507394 PMCID: PMC9150891 DOI: 10.7554/elife.76808] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Accepted: 05/03/2022] [Indexed: 12/03/2022] Open
Abstract
Many age-associated changes in the human hematopoietic system have been reproduced in murine models; however, such changes have not been as robustly explored in rats despite the fact these larger rodents are more physiologically similar to humans. We examined peripheral blood of male F344 rats ranging from 3 to 27 months of age and found significant age-associated changes with distinct leukocyte population shifts. We report CD25+ CD4+ population frequency is a strong predictor of healthy aging, generate a model using blood parameters, and find rats with blood profiles that diverge from chronologic age indicate debility; thus, assessments of blood composition may be useful for non-lethal disease profiling or as a surrogate measure for efficacy of aging interventions. Importantly, blood parameters and DNA methylation alterations, defined distinct juncture points during aging, supporting a non-linear aging process. Our results suggest these inflection points are important considerations for aging interventions. Overall, we present rat blood aging metrics that can serve as a resource to evaluate health and the effects of interventions in a model system physiologically more reflective of humans. Our blood contains many types of white blood cells, which play important roles in defending the body against infections and other threats to our health. The number of these cells changes with age, and this in turn contributes to many other alterations that happen in the body as we get older. For example, the immune system generally gets weaker at fighting infections and preventing other cells from developing into cancer. On top of that, the white blood cells themselves can become cancerous, resulting in several types of blood cancer that are more likely to happen in older people. Many previous studies have examined how the number of white blood cells changes with age in humans and mice. However, our understanding of this process in rats is still poor, despite the fact that the way the human body works has more in common with the rat body than the mouse body. Here, Yanai, Dunn et al. have studied samples of blood from rats between three to 27 months old. The experiments found that it is possible to accurately predict the age of healthy rats by measuring the frequency of populations of white blood cells, especially a certain type known as CD25+ CD4+ cells. If the animals had any form of illness, their predicted age deviated from their actual age. Furthermore, while some changes in the blood were gradual and continuous, others displayed distinct shifts when the rats reached specific ages. In the future, these findings may be used as a tool to help researchers diagnose illnesses in rats before the animals develop symptoms, or to more easily establish if a treatment is having a positive effect on the rats’ health. The work of Yanai, Dunn et al. also provides new insights into aging that could potentially aid the design of new screening methods to predict cancer and intervene using a model system that is more similar to humans.
Collapse
Affiliation(s)
- Hagai Yanai
- Flow Cytometry Core, National Institute on Aging, Baltimore, United States
| | - Christopher Dunn
- Flow Cytometry Core, National Institute on Aging, Baltimore, United States
| | - Bongsoo Park
- Epigenetics and Stem Cell Unit, National Institute on Aging, Baltimore, United States
| | - Christopher Coletta
- Computational Biology and Genomics Core, National Institute on Aging, Baltimore, United States
| | - Ross A McDevitt
- Comparative Medicine Section, National Institute on Aging, Baltimore, United States
| | - Taylor McNeely
- Epigenetics and Stem Cell Unit, National Institute on Aging, Baltimore, United States
| | - Michael Leone
- Epigenetics and Stem Cell Unit, National Institute on Aging, Baltimore, United States
| | - Robert P Wersto
- Flow Cytometry Core, National Institute on Aging, Baltimore, United States
| | - Kathy A Perdue
- Comparative Medicine Section, National Institute on Aging, Baltimore, United States
| | - Isabel Beerman
- Epigenetics and Stem Cell Unit, National Institute on Aging, Baltimore, United States
| |
Collapse
|
28
|
McKenzie BA, Chen F, LaCroix-Fralish ML. The phenotype of aging in the dog: how aging impacts the health and well-being of dogs and their caregivers. J Am Vet Med Assoc 2022; 260:963-970. [DOI: 10.2460/javma.22.02.0088] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Aging is the single most important cause of disease, disability, and death in adult dogs. Contrary to the common view of aging as a mysterious and inevitable natural event, it is more usefully understood as a set of complex but comprehensible biological processes that are highly conserved across species. Although the phenotypic expression of these processes is variable, there are consistent patterns both within and between species.
The purpose of this feature is to describe the patterns currently recognized in the physical and behavioral manifestations of aging in the dog and how these impact the health and welfare of companion dogs and their human caregivers. Important gaps in our knowledge of the canine aging phenotype will be identified, and current research efforts to better characterize aging in the dog will be discussed. This will help set the context for future efforts to develop clinical assessments and treatments to mitigate the negative impact of aging on dogs and humans.
Collapse
Affiliation(s)
| | - Frances Chen
- Cellular Longevity Inc dba Loyal, San Francisco, CA
| | | |
Collapse
|
29
|
Moturi S, Ghosh-Choudhary SK, Finkel T. Cardiovascular disease and the biology of aging. J Mol Cell Cardiol 2022; 167:109-117. [PMID: 35421400 DOI: 10.1016/j.yjmcc.2022.04.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 04/02/2022] [Accepted: 04/07/2022] [Indexed: 10/18/2022]
Abstract
The incidence and prevalence of a wide range of cardiovascular diseases increases as a function of age. This well-established epidemiological relationship suggests that chronological aging might contribute or increase susceptibility to varied conditions such as atherosclerosis, vascular stiffening or heart failure. Here, we explore the mechanistic links that connect both rare and common cardiovascular conditions to the basic biology of aging. These links provide a rational basis to begin to develop a new set of therapeutics targeting the fundamental mechanisms underlying the aging process and suggest that in the near future, age itself might become a modifiable cardiovascular risk factor.
Collapse
Affiliation(s)
- Shria Moturi
- Aging Institute, University of Pittsburgh School of Medicine/UPMC, Pittsburgh, PA 15219, United States of America
| | - Shohini K Ghosh-Choudhary
- Aging Institute, University of Pittsburgh School of Medicine/UPMC, Pittsburgh, PA 15219, United States of America
| | - Toren Finkel
- Aging Institute, University of Pittsburgh School of Medicine/UPMC, Pittsburgh, PA 15219, United States of America.
| |
Collapse
|
30
|
Amorim JA, Coppotelli G, Rolo AP, Palmeira CM, Ross JM, Sinclair DA. Mitochondrial and metabolic dysfunction in ageing and age-related diseases. Nat Rev Endocrinol 2022; 18:243-258. [PMID: 35145250 PMCID: PMC9059418 DOI: 10.1038/s41574-021-00626-7] [Citation(s) in RCA: 334] [Impact Index Per Article: 111.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/17/2021] [Indexed: 12/11/2022]
Abstract
Organismal ageing is accompanied by progressive loss of cellular function and systemic deterioration of multiple tissues, leading to impaired function and increased vulnerability to death. Mitochondria have become recognized not merely as being energy suppliers but also as having an essential role in the development of diseases associated with ageing, such as neurodegenerative and cardiovascular diseases. A growing body of evidence suggests that ageing and age-related diseases are tightly related to an energy supply and demand imbalance, which might be alleviated by a variety of interventions, including physical activity and calorie restriction, as well as naturally occurring molecules targeting conserved longevity pathways. Here, we review key historical advances and progress from the past few years in our understanding of the role of mitochondria in ageing and age-related metabolic diseases. We also highlight emerging scientific innovations using mitochondria-targeted therapeutic approaches.
Collapse
Affiliation(s)
- João A Amorim
- Department of Genetics, Blavatnik Institute, Paul F. Glenn Center for the Biology of Aging, Harvard Medical School, Boston, MA, USA
- Center for Neurosciences and Cell Biology of the University of Coimbra, Coimbra, Portugal
- IIIUC, Institute of Interdisciplinary Research, University of Coimbra, Coimbra, Portugal
| | - Giuseppe Coppotelli
- Department of Genetics, Blavatnik Institute, Paul F. Glenn Center for the Biology of Aging, Harvard Medical School, Boston, MA, USA
- George and Anne Ryan Institute for Neuroscience, College of Pharmacy, Department of Biomedical and Pharmaceutical Sciences, University of Rhode Island, Kingston, RI, USA
| | - Anabela P Rolo
- Center for Neurosciences and Cell Biology of the University of Coimbra, Coimbra, Portugal
- Department of Life Sciences of the University of Coimbra, Coimbra, Portugal
| | - Carlos M Palmeira
- Center for Neurosciences and Cell Biology of the University of Coimbra, Coimbra, Portugal
- Department of Life Sciences of the University of Coimbra, Coimbra, Portugal
| | - Jaime M Ross
- Department of Genetics, Blavatnik Institute, Paul F. Glenn Center for the Biology of Aging, Harvard Medical School, Boston, MA, USA
- George and Anne Ryan Institute for Neuroscience, College of Pharmacy, Department of Biomedical and Pharmaceutical Sciences, University of Rhode Island, Kingston, RI, USA
| | - David A Sinclair
- Department of Genetics, Blavatnik Institute, Paul F. Glenn Center for the Biology of Aging, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
31
|
Wang TH, Tseng WC, Leu YL, Chen CY, Lee WC, Chi YC, Cheng SF, Lai CY, Kuo CH, Yang SL, Yang SH, Shen JJ, Feng CH, Wu CC, Hwang TL, Wang CJ, Wang SH, Chen CC. The flavonoid corylin exhibits lifespan extension properties in mouse. Nat Commun 2022; 13:1238. [PMID: 35264584 PMCID: PMC8907184 DOI: 10.1038/s41467-022-28908-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Accepted: 02/16/2022] [Indexed: 01/23/2023] Open
Abstract
In the long history of traditional Chinese medicine, single herbs and complex formulas have been suggested to increase lifespan. However, the identification of single molecules responsible for lifespan extension has been challenging. Here, we collected a list of traditional Chinese medicines with potential longevity properties from pharmacopeias. By utilizing the mother enrichment program, we systematically screened these traditional Chinese medicines and identified a single herb, Psoralea corylifolia, that increases lifespan in Saccharomyces cerevisiae. Next, twenty-two pure compounds were isolated from Psoralea corylifolia. One of the compounds, corylin, was found to extend the replicative lifespan in yeast by targeting the Gtr1 protein. In human umbilical vein endothelial cells, RNA sequencing data showed that corylin ameliorates cellular senescence. We also examined an in vivo mammalian model, and found that corylin extends lifespan in mice fed a high-fat diet. Taken together, these findings suggest that corylin may promote longevity.
Collapse
Affiliation(s)
- Tong-Hong Wang
- Tissue Bank, Chang Gung Memorial Hospital, Taoyuan, Taiwan.,Graduate Institute of Health Industry Technology, Research Center for Food and Cosmetic Safety, Research Center for Chinese Herbal Medicine, College of Human Ecology, Chang Gung University of Science and Technology, Taoyuan, Taiwan
| | - Wei-Che Tseng
- Graduate Institute of Natural Products, Chang Gung University, Taoyuan, Taiwan
| | - Yann-Lii Leu
- Tissue Bank, Chang Gung Memorial Hospital, Taoyuan, Taiwan.,Graduate Institute of Natural Products, Chang Gung University, Taoyuan, Taiwan
| | - Chi-Yuan Chen
- Tissue Bank, Chang Gung Memorial Hospital, Taoyuan, Taiwan.,Graduate Institute of Health Industry Technology, Research Center for Food and Cosmetic Safety, Research Center for Chinese Herbal Medicine, College of Human Ecology, Chang Gung University of Science and Technology, Taoyuan, Taiwan
| | - Wen-Chih Lee
- Office of Research and Development, Tzu Chi University, Hualien, Taiwan
| | - Ying-Chih Chi
- Cryo-EM Center, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, USA
| | - Shu-Fang Cheng
- Graduate Institute of Natural Products, Chang Gung University, Taoyuan, Taiwan
| | - Chun-Yu Lai
- Graduate Institute of Natural Products, Chang Gung University, Taoyuan, Taiwan
| | - Chen-Hsin Kuo
- Graduate Institute of Natural Products, Chang Gung University, Taoyuan, Taiwan
| | - Shu-Ling Yang
- School of Traditional Chinese Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Sien-Hung Yang
- School of Traditional Chinese Medicine, Chang Gung University, Taoyuan, Taiwan.,Division of Chinese Internal Medicine, Center for Traditional Chinese Medicine, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Jiann-Jong Shen
- School of Traditional Chinese Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Chun-Hao Feng
- Graduate Institute of Natural Products, Chang Gung University, Taoyuan, Taiwan
| | - Chih-Ching Wu
- Department of Medical Biotechnology and Laboratory Science, Chang Gung University, Taoyuan, Taiwan.,Department of Otolaryngology-Head & Neck Surgery, Chang Gung Memorial Hospital, Taoyuan, Taiwan.,Molecular Medicine Research Center, Chang Gung University, Taoyuan, Taiwan.,Research Center for Emerging Viral Infections, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Tsong-Long Hwang
- Graduate Institute of Health Industry Technology, Research Center for Food and Cosmetic Safety, Research Center for Chinese Herbal Medicine, College of Human Ecology, Chang Gung University of Science and Technology, Taoyuan, Taiwan.,Graduate Institute of Natural Products, Chang Gung University, Taoyuan, Taiwan.,Department of Anesthesiology, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Chia-Jen Wang
- Cell Therapy Core Laboratory, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Shu-Huei Wang
- Department of Anatomy and Cell Biology, College of Medicine, National Taiwan University, Taipei, Taiwan.
| | - Chin-Chuan Chen
- Tissue Bank, Chang Gung Memorial Hospital, Taoyuan, Taiwan. .,Graduate Institute of Natural Products, Chang Gung University, Taoyuan, Taiwan.
| |
Collapse
|
32
|
Carroll JE, Bower JE, Ganz PA. Cancer-related accelerated ageing and biobehavioural modifiers: a framework for research and clinical care. Nat Rev Clin Oncol 2022; 19:173-187. [PMID: 34873313 PMCID: PMC9974153 DOI: 10.1038/s41571-021-00580-3] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/03/2021] [Indexed: 12/15/2022]
Abstract
A growing body of evidence indicates that patients with cancer who receive cytotoxic treatments (such as chemotherapy or radiotherapy) have an increased risk of accelerated physical and cognitive ageing. Furthermore, accelerated biological ageing is a suspected driving force behind many of these observed effects. In this Review, we describe the mechanisms of biological ageing and how they apply to patients with cancer. We highlight the important role of specific behavioural factors, namely stress, sleep and lifestyle-related factors such as physical activity, weight management, diet and substance use, in the accelerated ageing of patients with cancer and cancer survivors. We also present a framework of how modifiable behaviours could operate to either increase the risk of accelerated ageing, provide protection, or promote resilience at both the biological level and in terms of patient-reported outcomes.
Collapse
Affiliation(s)
- Judith E Carroll
- Norman Cousins Center for Psychoneuroimmunology, Jane and Terry Semel Institute for Neuroscience and Human Behaviour, University of California, Los Angeles, CA, USA.
- Department of Psychiatry & Biobehavioral Sciences, David Geffen School of Medicine, University of California, Los Angeles, CA, USA.
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, CA, USA.
| | - Julienne E Bower
- Norman Cousins Center for Psychoneuroimmunology, Jane and Terry Semel Institute for Neuroscience and Human Behaviour, University of California, Los Angeles, CA, USA
- Department of Psychiatry & Biobehavioral Sciences, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, CA, USA
- Department of Psychology, University of California, Los Angeles, CA, USA
| | - Patricia A Ganz
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, CA, USA
- Department of Health Policy & Management, Fielding School of Public Health, University of California, Los Angeles, CA, USA
- Department of Medicine (Hematology-Oncology), David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| |
Collapse
|
33
|
Using Microbiome-Based Approaches to Deprogram Chronic Disorders and Extend the Healthspan following Adverse Childhood Experiences. Microorganisms 2022; 10:microorganisms10020229. [PMID: 35208684 PMCID: PMC8879770 DOI: 10.3390/microorganisms10020229] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 01/04/2022] [Accepted: 01/19/2022] [Indexed: 12/01/2022] Open
Abstract
Adverse childhood experiences (ACEs), which can include child trafficking, are known to program children for disrupted biological cycles, premature aging, microbiome dysbiosis, immune-inflammatory misregulation, and chronic disease multimorbidity. To date, the microbiome has not been a major focus of deprogramming efforts despite its emerging role in every aspect of ACE-related dysbiosis and dysfunction. This article examines: (1) the utility of incorporating microorganism-based, anti-aging approaches to combat ACE-programmed chronic diseases (also known as noncommunicable diseases and conditions, NCDs) and (2) microbiome regulation of core systems biology cycles that affect NCD comorbid risk. In this review, microbiota influence over three key cyclic rhythms (circadian cycles, the sleep cycle, and the lifespan/longevity cycle) as well as tissue inflammation and oxidative stress are discussed as an opportunity to deprogram ACE-driven chronic disorders. Microbiota, particularly those in the gut, have been shown to affect host–microbe interactions regulating the circadian clock, sleep quality, as well as immune function/senescence, and regulation of tissue inflammation. The microimmunosome is one of several systems biology targets of gut microbiota regulation. Furthermore, correcting misregulated inflammation and increased oxidative stress is key to protecting telomere length and lifespan/longevity and extending what has become known as the healthspan. This review article concludes that to reverse the tragedy of ACE-programmed NCDs and premature aging, managing the human holobiont microbiome should become a routine part of healthcare and preventative medicine across the life course.
Collapse
|
34
|
Pandey M, Bansal S, Chawla G. Evaluation of lifespan promoting effects of biofortified wheat in Drosophila melanogaster. Exp Gerontol 2022; 160:111697. [PMID: 35016996 PMCID: PMC7613042 DOI: 10.1016/j.exger.2022.111697] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2021] [Revised: 12/15/2021] [Accepted: 01/05/2022] [Indexed: 11/04/2022]
Abstract
Evaluation of nutritionally enhanced biofortified dietary interventions that increase lifespan may uncover cost-effective and sustainable approaches for treatment of age-related morbidities and increasing healthy life expectancy. In this study, we report that anthocyanin rich, high yielding crossbred blue wheat prolongs lifespan of Drosophila melanogaster in different dietary contexts. In addition to functioning as an antioxidant rich intervention, the biofortified blue wheat also works through modulating expression of DR pathway genes including AMPK alpha, SREBP, PEPCK and Cry. Supplementation with blue- or purple-colored wheat provided better protection against paraquat-induced oxidative stress than control diet and increased survivability of flies in which superoxide dismutase 2 was knocked down conditionally in adults. Lastly, our findings indicate that supplementing biofortified blue wheat formulated diet prevented the decrease in lifespan and cardiac structural pathologies associated with intake of high fat diet. Overall, our findings indicate that plant-based diets formulated with biofortified cereal crops promote healthy ageing and delay progression of diseases that are exacerbated by accumulation of oxidative damage.
Collapse
Affiliation(s)
- Manish Pandey
- RNA Biology Laboratory, Regional Centre for Biotechnology, NCR Biotech Science Cluster, Faridabad 121001, Haryana, India
| | - Sakshi Bansal
- RNA Biology Laboratory, Regional Centre for Biotechnology, NCR Biotech Science Cluster, Faridabad 121001, Haryana, India
| | - Geetanjali Chawla
- RNA Biology Laboratory, Regional Centre for Biotechnology, NCR Biotech Science Cluster, Faridabad 121001, Haryana, India.
| |
Collapse
|
35
|
Yang Y, Du Z, Liu Y, Lao J, Sun X, Tang F. Disability and the risk of subsequent mortality in elderly: a 12-year longitudinal population-based study. BMC Geriatr 2021; 21:662. [PMID: 34814844 PMCID: PMC8609873 DOI: 10.1186/s12877-021-02611-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Accepted: 11/09/2021] [Indexed: 11/21/2022] Open
Abstract
Background Assessment the impact of disability on mortality among the elderly is vital to healthy ageing. The present study aimed to assess the long-term influence of disability on death in the elderly based on a longitudinal study. Method This study used the Chinese Longitudinal Healthy Longevity Study (CLHLS) data from 2002 to 2014, including 13,666 participants aged 65 years and older in analyses. The Katz ADL index was used to assess disability status and levels. Cumulative mortality rates were estimated by the Kaplan-Meier method. Cox proportional hazards models were conducted to estimate associations between disability and all-cause mortality for overall participants, two age groups as well as specific chronic disease groups. All reported results were adjusted by survey weights to account for the complex survey design. Results During the 12-year follow-up, the death density was 6.01 per 100 person-years. The 3-years’ cumulative mortality rate of nondisabled elderly was 11.9% (95%CI: 10.9, 12.9%). As the level of disability increased, the cumulative mortality rate was from 28.1% (95%CI: 23.0, 33.1%) to 77.6% (95%CI: 63.8, 91.4%). Compared with non-disabled elderly, the multiple-adjusted hazard ratio of death due to disability was 1.68 (95% CI: 1.48, 1.90). The hazard ratios varied from 1.44 (95%CI: 1.23, 1.67) to 4.45 (95%CI: 2.69, 7.38) after classifying the disability levels. The hazard ratios of death in the young-old group (65–79 years) were higher than the old-old group (80 years and over) in both level B (HR = 1.58, 95%CI: 1.25, 2.00 vs. HR = 1.22, 95%CI: 1.06, 1.39, P = 0.029) and level G (HR = 24.09, 95%CI: 10.83, 53.60 vs. HR = 2.56, 95%CI: 1.75, 3.74, P < 0.001). For patients with hypertension, diabetes, heart disease, cerebrovascular disease as well as dementia, disability increases their relative risk of mortality by 1.64 (95%CI: 1.40, 1.93), 2.85 (95%CI: 1.46, 5.58), 1.45 (95%CI: 1.02, 2.05), 2.13 (95%CI: 1.54, 2.93) and 3.56 (95%CI: 1.22, 10.38) times, respectively. Conclusions Disability increases the risk of all-cause death in the elderly, especially those with chronic diseases and the young-old group. Further studies are needed to better understand how to effectively prevent disability in the older population. Supplementary Information The online version contains supplementary material available at 10.1186/s12877-021-02611-1.
Collapse
Affiliation(s)
- Yang Yang
- Department of Gerontology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China.,Center for Big Data Research in Health and Medicine, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China
| | - Zhaohui Du
- Fred Hutchinson Cancer Research Center, Seattle, WA, 98109, USA
| | - Yafei Liu
- Center for Big Data Research in Health and Medicine, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China
| | - Jiahui Lao
- Center for Big Data Research in Health and Medicine, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China
| | - Xiaoru Sun
- Department of Biostatistics, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Fang Tang
- Department of Gerontology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China. .,Center for Big Data Research in Health and Medicine, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China. .,Shandong Provincial Qianfoshan Hospital, Cheeloo College of Medicine, Shandong University, No. 16766 Jingshi Road, Jinan, 250014, Shandong, China.
| |
Collapse
|
36
|
Hilmer SN. Bridging geriatric medicine, clinical pharmacology and ageing biology to understand and improve outcomes of medicines in old age and frailty. Ageing Res Rev 2021; 71:101457. [PMID: 34481922 DOI: 10.1016/j.arr.2021.101457] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 08/17/2021] [Accepted: 08/23/2021] [Indexed: 12/11/2022]
Affiliation(s)
- Sarah N Hilmer
- Kolling Institute, University of Sydney and Royal North Shore Hospital, St Leonards, NSW 2065, Australia.
| |
Collapse
|
37
|
Torrens-Mas M, Perelló-Reus C, Navas-Enamorado C, Ibargüen-González L, Sanchez-Polo A, Segura-Sampedro JJ, Masmiquel L, Barcelo C, Gonzalez-Freire M. Organoids: An Emerging Tool to Study Aging Signature across Human Tissues. Modeling Aging with Patient-Derived Organoids. Int J Mol Sci 2021; 22:10547. [PMID: 34638891 PMCID: PMC8508868 DOI: 10.3390/ijms221910547] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 09/24/2021] [Accepted: 09/25/2021] [Indexed: 12/12/2022] Open
Abstract
The biology of aging is focused on the identification of novel pathways that regulate the underlying processes of aging to develop interventions aimed at delaying the onset and progression of chronic diseases to extend lifespan. However, the research on the aging field has been conducted mainly in animal models, yeast, Caenorhabditis elegans, and cell cultures. Thus, it is unclear to what extent this knowledge is transferable to humans since they might not reflect the complexity of aging in people. An organoid culture is an in vitro 3D cell-culture technology that reproduces the physiological and cellular composition of the tissues and/or organs. This technology is being used in the cancer field to predict the response of a patient-derived tumor to a certain drug or treatment serving as patient stratification and drug-guidance approaches. Modeling aging with patient-derived organoids has a tremendous potential as a preclinical model tool to discover new biomarkers of aging, to predict adverse outcomes during aging, and to design personalized approaches for the prevention and treatment of aging-related diseases and geriatric syndromes. This could represent a novel approach to study chronological and/or biological aging, paving the way to personalized interventions targeting the biology of aging.
Collapse
Affiliation(s)
- Margalida Torrens-Mas
- Vascular and Metabolic Pathologies Group, Health Research Institute of the Balearic Islands (IdISBa), 07120 Palma de Mallorca, Spain; (M.T.-M.); (C.N.-E.); (A.S.-P.); (L.M.)
- Translational Research in Aging and Longevity (TRIAL) Group, Health Research Institute of the Balearic Islands (IdISBa), 07120 Palma de Mallorca, Spain
| | - Catalina Perelló-Reus
- Translational Pancreatic Cancer Oncogenesis Group, Health Research Institute of the Balearic Islands (IdISBa), 07120 Palma de Mallorca, Spain; (C.P.-R.); (L.I.-G.)
| | - Cayetano Navas-Enamorado
- Vascular and Metabolic Pathologies Group, Health Research Institute of the Balearic Islands (IdISBa), 07120 Palma de Mallorca, Spain; (M.T.-M.); (C.N.-E.); (A.S.-P.); (L.M.)
- Translational Research in Aging and Longevity (TRIAL) Group, Health Research Institute of the Balearic Islands (IdISBa), 07120 Palma de Mallorca, Spain
| | - Lesly Ibargüen-González
- Translational Pancreatic Cancer Oncogenesis Group, Health Research Institute of the Balearic Islands (IdISBa), 07120 Palma de Mallorca, Spain; (C.P.-R.); (L.I.-G.)
| | - Andres Sanchez-Polo
- Vascular and Metabolic Pathologies Group, Health Research Institute of the Balearic Islands (IdISBa), 07120 Palma de Mallorca, Spain; (M.T.-M.); (C.N.-E.); (A.S.-P.); (L.M.)
- Translational Research in Aging and Longevity (TRIAL) Group, Health Research Institute of the Balearic Islands (IdISBa), 07120 Palma de Mallorca, Spain
| | - Juan Jose Segura-Sampedro
- Translational Research in Aging and Longevity (TRIAL) Group, Health Research Institute of the Balearic Islands (IdISBa), 07120 Palma de Mallorca, Spain
- General & Digestive Surgery Department, Hospital Universitario Son Espases, 07120 Palma de Mallorca, Spain;
- School of Medicine, University of the Balearic Islands, 07120 Palma de Mallorca, Spain
| | - Luis Masmiquel
- Vascular and Metabolic Pathologies Group, Health Research Institute of the Balearic Islands (IdISBa), 07120 Palma de Mallorca, Spain; (M.T.-M.); (C.N.-E.); (A.S.-P.); (L.M.)
| | - Carles Barcelo
- Translational Pancreatic Cancer Oncogenesis Group, Health Research Institute of the Balearic Islands (IdISBa), 07120 Palma de Mallorca, Spain; (C.P.-R.); (L.I.-G.)
| | - Marta Gonzalez-Freire
- Vascular and Metabolic Pathologies Group, Health Research Institute of the Balearic Islands (IdISBa), 07120 Palma de Mallorca, Spain; (M.T.-M.); (C.N.-E.); (A.S.-P.); (L.M.)
- Translational Research in Aging and Longevity (TRIAL) Group, Health Research Institute of the Balearic Islands (IdISBa), 07120 Palma de Mallorca, Spain
| |
Collapse
|
38
|
Borgoni S, Kudryashova KS, Burka K, de Magalhães JP. Targeting immune dysfunction in aging. Ageing Res Rev 2021; 70:101410. [PMID: 34280555 DOI: 10.1016/j.arr.2021.101410] [Citation(s) in RCA: 97] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 06/28/2021] [Accepted: 07/14/2021] [Indexed: 12/23/2022]
Abstract
Human aging is a multifactorial phenomenon that affects numerous organ systems and cellular processes, with the immune system being one of the most dysregulated. Immunosenescence, the gradual deterioration of the immune system, and inflammaging, a chronic inflammatory state that persists in the elderly, are among the plethora of immune changes that occur during aging. Almost all populations of immune cells change with age in terms of numbers and/or activity. These alterations are in general highly detrimental, resulting in an increased susceptibility to infections, reduced healing abilities, and altered homeostasis that promote the emergence of age-associated diseases such as cancer, diabetes, and other diseases associated with inflammation. Thanks to recent developments, several strategies have been proposed to target central immunological processes or specific immune subpopulations affected by aging. These therapeutic approaches could soon be applied in the clinic to slow down or even reverse specific age-induced immune changes in order to rejuvenate the immune system and prevent or reduce the impact of various diseases. Due to its systemic nature and interconnection with all the other systems in the body, the immune system is an attractive target for aging intervention because relatively targeted modifications to a small set of cells have the potential to improve the health of multiple organ systems. Therefore, anti-aging immune targeting therapies could represent a potent approach for improving healthspan. Here, we review aging changes in the major components of the immune system, we summarize the current immune-targeting therapeutic approaches in the context of aging and discuss the future directions in the field of immune rejuvenation.
Collapse
|
39
|
Palliyaguru DL, Vieira Ligo Teixeira C, Duregon E, di Germanio C, Alfaras I, Mitchell SJ, Navas-Enamorado I, Shiroma EJ, Studenski S, Bernier M, Camandola S, Price NL, Ferrucci L, de Cabo R. Study of Longitudinal Aging in Mice: Presentation of Experimental Techniques. J Gerontol A Biol Sci Med Sci 2021; 76:552-560. [PMID: 33211821 DOI: 10.1093/gerona/glaa285] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Indexed: 12/11/2022] Open
Abstract
Aging is associated with functional and metabolic decline and is a risk factor for all noncommunicable diseases. Even though mice are routinely used for modeling human aging and aging-related conditions, no comprehensive assessment to date has been conducted on normative mouse aging. To address this gap, the Study of Longitudinal Aging in Mice (SLAM) was designed and implemented by the National Institute on Aging (NIA/NIH) as the mouse counterpart to the Baltimore Longitudinal Study of Aging (BLSA). In this manuscript, we describe the premise, study design, methodologies, and technologies currently employed in SLAM. We also discuss current and future study directions. In this large population mouse study, inbred C57BL/6J and outbred UM-HET3 mice of both sexes are longitudinally evaluated for functional, phenotypic, and biological health, and collection of biospecimens is conducted throughout their life span. Within the longitudinal cohorts, a cross-sectional arm of the study has also been implemented for the well-controlled collection of tissues to generate a biorepository. SLAM and studies stemming from SLAM seek to identify and characterize phenotypic and biological predictors of mouse aging and age-associated conditions, examine the degrees of functional and biomolecular variability that occur within inbred and genetically heterogeneous mouse populations with age, and assess whether these changes are consistent with alterations observed in human aging in BLSA. The findings from these studies will be critical for evaluating the utility of mouse models for studying different aspects of aging, both in terms of interpreting prior findings and designing and implementing future studies.
Collapse
Affiliation(s)
- Dushani L Palliyaguru
- Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, Maryland, USA
| | - Camila Vieira Ligo Teixeira
- Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, Maryland, USA
| | - Eleonora Duregon
- Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, Maryland, USA
| | - Clara di Germanio
- Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, Maryland, USA.,Vitalant Research Institute, San Francisco, California, USA
| | - Irene Alfaras
- Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, Maryland, USA.,Aging Institute of UPMC and the University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Sarah J Mitchell
- Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, Maryland, USA.,Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Ignacio Navas-Enamorado
- Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, Maryland, USA.,Boston University School of Medicine, Massachusetts, USA
| | - Eric J Shiroma
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, National Institutes of Health, Baltimore, Maryland, USA
| | - Stephanie Studenski
- Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, Maryland, USA
| | - Michel Bernier
- Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, Maryland, USA
| | - Simonetta Camandola
- Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, Maryland, USA
| | - Nathan L Price
- Department of Comparative Medicine, Yale University, New Haven, Connecticut, USA
| | - Luigi Ferrucci
- Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, Maryland, USA
| | - Rafael de Cabo
- Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, Maryland, USA
| |
Collapse
|
40
|
Ungvari Z, Adany R. The future of healthy aging: translation of geroscience discoveries to public health practice. Eur J Public Health 2021; 31:455-456. [PMID: 33594423 DOI: 10.1093/eurpub/ckaa212] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Affiliation(s)
- Zoltan Ungvari
- Healthy Aging Program, Department of Public Health, Semmelweis University, Budapest, Hungary
- Vascular Cognitive Impairment and Neurodegeneration Program, Center for Geroscience and Healthy Brain Aging/Reynolds Oklahoma Center on Aging, Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Department of Health Promotion Sciences, The Hudson College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Roza Adany
- Healthy Aging Program, Department of Public Health, Semmelweis University, Budapest, Hungary
- Department of Public Health and Epidemiology, MTA-DE Public Health Research Group, University of Debrecen, Debrecen, Hungary
| |
Collapse
|
41
|
Dai X, Guo X. Decoding and rejuvenating human ageing genomes: Lessons from mosaic chromosomal alterations. Ageing Res Rev 2021; 68:101342. [PMID: 33866012 DOI: 10.1016/j.arr.2021.101342] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Revised: 04/05/2021] [Accepted: 04/07/2021] [Indexed: 01/10/2023]
Abstract
One of the most curious findings emerged from genome-wide studies over the last decade was that genetic mosaicism is a dominant feature of human ageing genomes. The clonal dominance of genetic mosaicism occurs preceding the physiological and physical ageing and associates with propensity for diseases including cancer, Alzheimer's disease, cardiovascular disease and diabetes. These findings are revolutionizing the ways biologists thinking about health and disease pathogenesis. Among all mosaic mutations in ageing genomes, mosaic chromosomal alterations (mCAs) have the most significant functional consequences because they can produce intercellular genomic variations simultaneously involving dozens to hundreds or even thousands genes, and therefore have most profound effects in human ageing and disease etiology. Here, we provide a comprehensive picture of the landscapes, causes, consequences and rejuvenation of mCAs at multiple scales, from cell to human population, by reviewing data from cytogenetic, genetic and genomic studies in cells, animal models (fly and mouse) and, more frequently, large-cohort populations. A detailed decoding of ageing genomes with a focus on mCAs may yield important insights into the genomic architecture of human ageing, accelerate the risk stratification of age-related diseases (particularly cancers) and development of novel targets and strategies for delaying or rejuvenating human (genome) ageing.
Collapse
Affiliation(s)
- Xueqin Dai
- School of Life Sciences, Yunnan Normal University, Kunming, Yunnan, 650500, China
| | - Xihan Guo
- School of Life Sciences, Yunnan Normal University, Kunming, Yunnan, 650500, China; The Engineering Research Center of Sustainable Development and Utilization of Biomass Energy, Ministry of Education, Kunming, Yunnan, 650500, China; Yunnan Environmental Mutagen Society, Kunming, Yunnan, 650500, China.
| |
Collapse
|
42
|
Erlandson KM, Piggott DA. Frailty and HIV: Moving from Characterization to Intervention. Curr HIV/AIDS Rep 2021; 18:157-175. [PMID: 33817767 PMCID: PMC8193917 DOI: 10.1007/s11904-021-00554-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/22/2021] [Indexed: 12/12/2022]
Abstract
PURPOSE OF REVIEW While the characteristics associated with frailty in people with HIV (PWH) have been well described, little is known regarding interventions to slow or reverse frailty. Here we review interventions to prevent or treat frailty in the general population and in people with HIV (PWH). RECENT FINDINGS Frailty interventions have primarily relied on nonpharmacologic interventions (e.g., exercise and nutrition). Although few have addressed frailty, many of these therapies have shown benefit on components of frailty including gait speed, strength, and low activity among PWH. When nonpharmacologic interventions are insufficient, pharmacologic interventions may be necessary. Many interventions have been tested in preclinical models, but few have been tested or shown benefit among older adults with or without HIV. Ultimately, pharmacologic and nonpharmacologic interventions have the potential to improve vulnerability that underlies frailty in PWH, though clinical data is currently sparse.
Collapse
Affiliation(s)
- Kristine M Erlandson
- Department of Medicine, Division of Infectious Diseases, University of Colorado-Anschutz Medical Campus, 12700 E. 19th Avenue, Mail Stop B168, Aurora, CO, 80045, USA.
- Department of Epidemiology, Colorado School of Public Health, Anschutz Medical Campus, Aurora, CO, USA.
| | - Damani A Piggott
- Department of Medicine, Division of Infectious Diseases, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Epidemiology, Johns Hopkins University School of Public Health, Baltimore, MD, USA
| |
Collapse
|
43
|
Wongchum N, Dechakhamphu A. Xanthohumol prolongs lifespan and decreases stress-induced mortality in Drosophila melanogaster. Comp Biochem Physiol C Toxicol Pharmacol 2021; 244:108994. [PMID: 33549830 DOI: 10.1016/j.cbpc.2021.108994] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Revised: 01/12/2021] [Accepted: 01/24/2021] [Indexed: 11/23/2022]
Abstract
Aging is a significant risk factor that links to the genesis of human diseases. The capacity to scavenge free radicals and adapt to various stresses is essential for expanding living organisms' lifespan. The evidences on the promotion of longevity by dietary supplementation are growing. Drosophila or fruit fly is one of the most effective models for the evaluation of anti-aging compounds. Xanthohumol (XN) is a potential bioactive substance for the prevention and treatment of many diseases. The previous studies have reported its potent activities as antioxidant, anticancer, anti-inflammatory, antiviral, antibacterial antiplasmodial, and antiobesity. In this study, the effect of XN supplementation on the lifespan extension was investigated in Drosophila melanogaster. The effects of XN on the improvement of the recovery from cold and heat shock, the resistance to starvation stress, and free radical-induced oxidative stress in XN-treated flies were also evaluated. Results showed that supplementation with XN at 0.5 mg/mL diet extended the mean lifespan by 14.89%. This was consistent with a significant improvement of locomotor activity of the Drosophila fed with an XN-mixed diet compared with those fed with a control diet. XN supplementation significantly increased the antioxidant enzyme activities at both 25 and 40 days. Drosophila treated with XN exhibited increased survival after exposure to hydrogen peroxide and paraquat. Finally, XN supplementation improved the recovery from cold and heat shock, the resistance to starvation stress, and acetic acid-induced stress. The present study shows that dietary supplementation with XN revealed the longevity effect and ameliorated stress-induced mortality in Drosophila.
Collapse
Affiliation(s)
- Nattapong Wongchum
- Biology Program, Faculty of Science, Ubon Ratchathani Rajabhat University, Ubonratchathani 34000, Thailand
| | - Ananya Dechakhamphu
- Thai Traditional Medicine Program, Faculty of Thai Traditional and Alternative Medicine, Ubon Ratchathani Rajabhat University, Ubonratchathani 34000, Thailand.
| |
Collapse
|
44
|
Abstract
One of the best strategies for healthy brain aging is regular aerobic exercise. Commonly studied "anti-aging" compounds may mimic some effects of exercise on the brain, but novel approaches that target energy-sensing pathways similar to exercise probably will be more effective in this context. We review evidence in support of this hypothesis by focusing on biological hallmarks of brain aging.
Collapse
|
45
|
Trajano GS, Blazevich AJ. Static Stretching Reduces Motoneuron Excitability: The Potential Role of Neuromodulation. Exerc Sport Sci Rev 2021; 49:126-132. [PMID: 33720914 PMCID: PMC7967995 DOI: 10.1249/jes.0000000000000243] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/06/2022]
Abstract
Prolonged static muscle stretching transiently reduces maximal muscle force, and this force loss has a strong neural component. In this review, we discuss the evidence suggesting that stretching reduces the motoneuron's ability to amplify excitatory drive. We propose a hypothetical model in which stretching causes physiological relaxation, reducing the brainstem-derived neuromodulatory drive necessary to maximize motoneuron discharge rates.
Collapse
Affiliation(s)
- Gabriel S Trajano
- School of Exercise and Nutrition Sciences, Faculty of Health, Queensland University of Technology, Brisbane
| | - Anthony J Blazevich
- Centre for Exercise and Sports Science Research, School of Medical and Health Sciences, Edith Cowan University, Joondalup, Australia
| |
Collapse
|
46
|
Hepowit NL, Macedo JKA, Young LEA, Liu K, Sun RC, MacGurn JA, Dickson RC. Enhancing lifespan of budding yeast by pharmacological lowering of amino acid pools. Aging (Albany NY) 2021; 13:7846-7871. [PMID: 33744865 PMCID: PMC8034917 DOI: 10.18632/aging.202849] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Accepted: 01/21/2021] [Indexed: 04/20/2023]
Abstract
The increasing prevalence of age-related diseases and resulting healthcare insecurity and emotional burden require novel treatment approaches. Several promising strategies seek to limit nutrients and promote healthy aging. Unfortunately, the human desire to consume food means this strategy is not practical for most people but pharmacological approaches might be a viable alternative. We previously showed that myriocin, which impairs sphingolipid synthesis, increases lifespan in Saccharomyces cerevisiae by modulating signaling pathways including the target of rapamycin complex 1 (TORC1). Since TORC1 senses cellular amino acids, we analyzed amino acid pools and identified 17 that are lowered by myriocin treatment. Studying the methionine transporter, Mup1, we found that newly synthesized Mup1 traffics to the plasma membrane and is stable for several hours but is inactive in drug-treated cells. Activity can be restored by adding phytosphingosine to culture medium thereby bypassing drug inhibition, thus confirming a sphingolipid requirement for Mup1 activity. Importantly, genetic analysis of myriocin-induced longevity revealed a requirement for the Gtr1/2 (mammalian Rags) and Vps34-Pib2 amino acid sensing pathways upstream of TORC1, consistent with a mechanism of action involving decreased amino acid availability. These studies demonstrate the feasibility of pharmacologically inducing a state resembling amino acid restriction to promote healthy aging.
Collapse
Affiliation(s)
- Nathaniel L. Hepowit
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN 37240, USA
| | - Jessica K. A. Macedo
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, KY 40536, USA
- Markey Cancer Center, University of Kentucky, Lexington, KY 40536, USA
| | - Lyndsay E. A. Young
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, KY 40536, USA
| | - Ke Liu
- Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, Sichuan University, Chengdu 610000, Sichuan, P. R. China
| | - Ramon C. Sun
- Markey Cancer Center, University of Kentucky, Lexington, KY 40536, USA
- Department of Neuroscience, University of Kentucky, Lexington, KY 40536, USA
| | - Jason A. MacGurn
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN 37240, USA
| | - Robert C. Dickson
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, KY 40536, USA
| |
Collapse
|
47
|
Abstract
Aging has largely been defined by analog measures of organ and organismal dysfunction. This has led to the characterization of aging processes at the molecular and cellular levels that underlie these gradual changes. However, current knowledge does not fully explain the growing body of data emerging from large epidemiological, systems biology, and single cell studies of entire organisms pointing to varied rates of aging between individuals (different functionality and lifespan), across lifespan (asynchronous aging), and within an organism at the tissue and organ levels (aging mosaicism). Here we consider these inhomogeneities in the broader context of the rate of aging and from the perspective of underlying cellular changes. These changes reflect genetic, environmental, and stochastic factors that cells integrate to adopt new homeostatic, albeit less functional, states, such as cellular senescence. In this sense, cellular aging can be viewed, at least in part, as a quantal process we refer to as "digital aging". Nevertheless, analog declines of tissue dysfunction and organ failure with age could be the sum of underlying digital events. Importantly, cellular aging, digital or otherwise, is not uniform across time or space within the organism or between organisms of the same species. Certain tissues may exhibit earliest signs of cellular aging, acting as drivers for organismal aging as signals from those driver cells within those tissues may accelerate the aging of other cells locally or even systemically. Advanced methodologies at the systems level and at the single cell level are likely to continue to refine our understanding to the processes of how cells and tissues age and how the integration of those processes leads to the complexities of individual, organismal aging.
Collapse
|
48
|
Proshkina EN, Solovev IA, Shaposhnikov MV, Moskalev AA. Key Molecular Mechanisms of Aging, Biomarkers, and Potential Interventions. Mol Biol 2021. [DOI: 10.1134/s0026893320060096] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
|
49
|
Duregon E, Vieira Ligo Teixeira C, Palliyaguru DL, Rudderow AL, Hoffmann V, Bernier M, Price NL, Camandola S, de Cabo R. Spontaneous chordoma: a case report on a female UM-HET3 mouse from the SLAM study. ACTA ACUST UNITED AC 2020; 2:219-222. [PMID: 34355215 DOI: 10.31491/apt.2020.12.043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
A female UM-HET3 mouse from the Study of Longitudinal Aging in Mice (SLAM) was euthanized at 164 weeks of age due to hind limb weakness. Necropsy and histological analysis revealed that the most probable cause of the clinical finding was the compression of the thoracolumbar segment of the spinal cord by herniated intervertebral disks. In addition, a spontaneous chordoma was incidentally found in the coccygeal bones. Given the rarity of this type of tumor, bio-clinical annotations acquired throughout lifespan, detailed histopathological assessment, and comparative clinical-pathological correlations for this mouse are presented and discussed.
Collapse
Affiliation(s)
- Eleonora Duregon
- Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA
| | - Camila Vieira Ligo Teixeira
- Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA
| | - Dushani L Palliyaguru
- Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA
| | - Annamaria L Rudderow
- Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA
| | - Victoria Hoffmann
- Division of Veterinary Resources, National Institutes of Health, Bethesda, MD 20892, USA
| | - Michel Bernier
- Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA
| | - Nathan L Price
- Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA
| | - Simonetta Camandola
- Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA
| | - Rafael de Cabo
- Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA
| |
Collapse
|
50
|
Thelen M, Brown-Borg HM. Does Diet Have a Role in the Treatment of Alzheimer's Disease? Front Aging Neurosci 2020; 12:617071. [PMID: 33424583 PMCID: PMC7785773 DOI: 10.3389/fnagi.2020.617071] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Accepted: 11/30/2020] [Indexed: 12/23/2022] Open
Abstract
The aging process causes many changes to the brain and is a major risk factor for the development of neurodegenerative diseases such as Alzheimer's Disease (AD). Despite an already vast amount of research on AD, a greater understanding of the disease's pathology and therapeutic options are desperately needed. One important distinction that is also in need of further study is the ability to distinguish changes to the brain observed in early stages of AD vs. changes that occur with normal aging. Current FDA-approved therapeutic options for AD patients have proven to be ineffective and indicate the need for alternative therapies. Aging interventions including alterations in diet (such as caloric restriction, fasting, or methionine restriction) have been shown to be effective in mediating increased health and lifespan in mice and other model organisms. Because aging is the greatest risk factor for the development of neurodegenerative diseases, certain dietary interventions should be explored as they have the potential to act as a future treatment option for AD patients.
Collapse
Affiliation(s)
- Mitchell Thelen
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND, United States
| | - Holly M Brown-Borg
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND, United States
| |
Collapse
|