1
|
Singh M, Jhajharia A, Pruthi R, Carmichael OT. 31P-MRS-Measured Phosphocreatine Recovery Kinetics in Human Muscles in Health and Disease-A Systematic Review and Meta-Analysis. NMR IN BIOMEDICINE 2025; 38:e70023. [PMID: 40189235 DOI: 10.1002/nbm.70023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 03/06/2025] [Accepted: 03/22/2025] [Indexed: 05/17/2025]
Abstract
The noninvasive, in vivo measurement of postexercise phosphocreatine (PCr) recovery kinetics using 31-phosphorus magnetic resonance spectroscopy (31P-MRS) is a highly prevalent method for assessing skeletal muscle energetics. However, 31P-MRS methodology is notoriously laboratory-specific, leading to uncertainty about the normal range of PCr recovery kinetics among healthy individuals, as well as relationships with disease and demographic factors. This systematic review and meta-analysis characterized the normal range of PCr recovery kinetics from 31P-MRS in human skeletal muscles across the lifespan, differences between healthy and those with muscle-related diseases, and relationships between intermuscular PCr recovery measurements and demographic factors. PubMed, Web of Science, Cochrane, and Google Scholar databases were searched for PCr recovery studies, which resulted in a final set of 128 studies eligible for meta-analysis. Studies were categorized into three muscle groups (forearm, upper leg, and lower leg) and further subdivided into three groups: diseased, control (the comparator group in studies of disease), and healthy (those recruited into studies that lacked a disease group). Only English-language studies were included. All statistical analysis was performed using Stata 17 software. Forest plots showed significant heterogeneity across PCr recovery time estimates and outlier study removal significantly reduced this heterogeneity. Greater age was associated with longer PCr recovery in upper leg muscles among both healthy (ρ = 0.387, p < 0.05) and diseased (ρ = 0.733, p < 0.05) individuals. Additionally, longer PCr recovery time was correlated with more acidic end-of-exercise pH in all three muscle groups among healthy individuals. In conclusion, skeletal muscle energetics as indexed by 31P-MRS-based PCr recovery time is similar across three different skeletal muscle groups among healthy people. Common diseases significantly prolong PCr recovery times. Methodological heterogeneity has a significant impact on PCr recovery time measurements in this literature. Greater age and more acidic pH increase PCr recovery time among healthy people.
Collapse
Affiliation(s)
- Maninder Singh
- Pennington Biomedical Research Center, Baton Rouge, Louisiana, USA
| | - Aditya Jhajharia
- School of Medicine, University of Maryland, Baltimore, Maryland, USA
| | - Rajat Pruthi
- School of Plant Environmental and Soil Sciences, Louisiana State University, Baton Rouge, Louisiana, USA
| | | |
Collapse
|
2
|
Donega S, Banskota N, Gupta E, Gonzalez-Freire M, Moore AZ, Ubaida-Mohien C, Munk R, Zukley L, Piao Y, Bergeron C, Bergeron J, Bektas A, Zampino M, Stagg C, Indig F, Hartnell LM, Kaileh M, Fishbein K, Spencer RG, Gorospe M, De S, Egan JM, Sen R, Ferrucci L. Skeletal Muscle mRNA Splicing Variants Association With Four Different Fitness and Energetic Measures in the GESTALT Study. J Cachexia Sarcopenia Muscle 2025; 16:e13603. [PMID: 39621510 DOI: 10.1002/jcsm.13603] [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: 07/03/2023] [Revised: 07/03/2024] [Accepted: 08/12/2024] [Indexed: 01/04/2025] Open
Abstract
BACKGROUND Physical activity is essential for maintaining muscle mitochondrial function and aerobic capacity. The molecular mechanisms underlying such protective effects are incompletely understood, in part because it is difficult to separate the effects of disease status and physical activity. We explored the association of human skeletal muscle transcriptomic with four measures of energetics and mitochondria oxidative capacity in healthy individuals. METHODS Using RNA sequencing of vastus lateralis muscle biopsies from 82 GESTALT participants (52 males, aged 22-89 years), we explored gene and splicing variant expression profiles associated with self-reported physical activity, peak oxygen consumption (VO2 peak), muscle oxidative capacity (kPCr) and mitochondrial respiration (Mit-O2 flux). The effect of aging on gene expression was examined in participants with low and high VO2 peak. RESULTS The four measures of energetics were negative correlated with age and generally intercorrelated. We identified protein-coding genes associated with four energetic measures adjusting for age, muscle fiber-ratio, sex and batch effect. Mitochondrial pathways were overrepresented across all energetic variables, albeit with little overlap at the gene level. Alternative spliced transcript isoforms associated with energetics were primarily enriched for cytoplasmic ribonucleoprotein granules. The splicing pathway was up-regulated with aging in low but not in high fitness participants, and transcript isoforms detected in the low fitness group pertain to processes such as cell cycle regulation, RNA/protein localization, nuclear transport and catabolism. CONCLUSIONS A consistent mitochondrial signature emerged across all energetic measures. Alternative splicing was enhanced in older, low fitness participants supporting the energy-splicing axis hypothesis. The identified splicing variants were enriched in pathways involving the accumulation of ribonucleoproteins in cytoplasmic granules, whose function remains unclear. Further research is needed to understand the function of these proteoforms in promoting adaptation to low energy availability.
Collapse
Affiliation(s)
- Stefano Donega
- Longitudinal Studies Section (LSS), National Institute on Aging (NIA), National Institutes of Health (NIH), Baltimore, Maryland, USA
| | - Nirad Banskota
- Laboratory of Genetics and Genomics (LGG), National Institute on Aging (NIA), National Institutes of Health (NIH), Baltimore, Maryland, USA
| | - Esha Gupta
- Laboratory of Genetics and Genomics (LGG), National Institute on Aging (NIA), National Institutes of Health (NIH), Baltimore, Maryland, USA
| | - Marta Gonzalez-Freire
- Translational Research in Aging and Longevity Group (TRIAL group), Fundació Institut d'Investigació Sanitària Illes Balears (IdISBa), Palma de Mallorca, Spain
- Faculty of Experimental Sciences, Universidad Francisco de Vitoria (UFV), Madrid, Spain
| | - Ann Zenobia Moore
- Longitudinal Studies Section (LSS), National Institute on Aging (NIA), National Institutes of Health (NIH), Baltimore, Maryland, USA
| | - Ceereena Ubaida-Mohien
- Longitudinal Studies Section (LSS), National Institute on Aging (NIA), National Institutes of Health (NIH), Baltimore, Maryland, USA
| | - Rachel Munk
- Laboratory of Genetics and Genomics (LGG), National Institute on Aging (NIA), National Institutes of Health (NIH), Baltimore, Maryland, USA
| | - Linda Zukley
- Clinical Research Core (CRC), National Institute on Aging (NIA), National Institutes of Health (NIH), Baltimore, Maryland, USA
| | - Yulan Piao
- Laboratory of Genetics and Genomics (LGG), National Institute on Aging (NIA), National Institutes of Health (NIH), Baltimore, Maryland, USA
- Clinical Research Core (CRC), National Institute on Aging (NIA), National Institutes of Health (NIH), Baltimore, Maryland, USA
| | - Chris Bergeron
- Clinical Research Core (CRC), National Institute on Aging (NIA), National Institutes of Health (NIH), Baltimore, Maryland, USA
| | - Jan Bergeron
- Clinical Research Core (CRC), National Institute on Aging (NIA), National Institutes of Health (NIH), Baltimore, Maryland, USA
| | - Arsun Bektas
- Clinical Research Core (CRC), National Institute on Aging (NIA), National Institutes of Health (NIH), Baltimore, Maryland, USA
| | - Marta Zampino
- Longitudinal Studies Section (LSS), National Institute on Aging (NIA), National Institutes of Health (NIH), Baltimore, Maryland, USA
| | - Carole Stagg
- Confocal Imaging Facility, National Institute on Aging (NIA), National Institutes of Health (NIH), Baltimore, Maryland, USA
| | - Fred Indig
- Confocal Imaging Facility, National Institute on Aging (NIA), National Institutes of Health (NIH), Baltimore, Maryland, USA
| | - Lisa M Hartnell
- Longitudinal Studies Section (LSS), National Institute on Aging (NIA), National Institutes of Health (NIH), Baltimore, Maryland, USA
| | - Mary Kaileh
- Clinical Research Core (CRC), National Institute on Aging (NIA), National Institutes of Health (NIH), Baltimore, Maryland, USA
| | - Kenneth Fishbein
- Clinical Research Core (CRC), National Institute on Aging (NIA), National Institutes of Health (NIH), Baltimore, Maryland, USA
| | - Richard G Spencer
- Laboratory of Clinical Investigation, National Institute on Aging (NIA), National Institutes of Health (NIH), Baltimore, Maryland, USA
| | - Myriam Gorospe
- Laboratory of Genetics and Genomics (LGG), National Institute on Aging (NIA), National Institutes of Health (NIH), Baltimore, Maryland, USA
| | - Supriyo De
- Laboratory of Genetics and Genomics (LGG), National Institute on Aging (NIA), National Institutes of Health (NIH), Baltimore, Maryland, USA
| | - Josephine M Egan
- Clinical Research Core (CRC), National Institute on Aging (NIA), National Institutes of Health (NIH), Baltimore, Maryland, USA
- Laboratory of Clinical Investigation, National Institute on Aging (NIA), National Institutes of Health (NIH), Baltimore, Maryland, USA
| | - Ranjan Sen
- Laboratory of Molecular Biology and Immunology (LMBI), National Institute on Aging (NIA), National Institutes of Health (NIH), Baltimore, Maryland, USA
| | - Luigi Ferrucci
- Longitudinal Studies Section (LSS), National Institute on Aging (NIA), National Institutes of Health (NIH), Baltimore, Maryland, USA
| |
Collapse
|
3
|
Kamarulzaman NT, Makpol S. The link between Mitochondria and Sarcopenia. J Physiol Biochem 2025; 81:1-20. [PMID: 39969761 PMCID: PMC11958477 DOI: 10.1007/s13105-024-01062-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Accepted: 11/12/2024] [Indexed: 02/20/2025]
Abstract
Sarcopenia, a widespread condition, is characterized by a variety of factors influencing its development. The causes of sarcopenia differ depending on the age of the individual. It is defined as the combination of decreased muscle mass and impaired muscle function, primarily observed in association with ageing. As people age from 20 to 80 years old, there is an approximate 30% reduction in muscle mass and a 20% decline in cross-sectional area. This decline is attributed to a decrease in the size and number of muscle fibres. The regression of muscle mass and strength increases the risk of fractures, frailty, reduced quality of life, and loss of independence. Muscle cells, fibres, and tissues shrink, resulting in diminished muscle power, volume, and strength in major muscle groups. One prominent theory of cellular ageing posits a strong positive relationship between age and oxidative damage. Heightened oxidative stress leads to early-onset sarcopenia, characterized by neuromuscular innervation breakdown, muscle atrophy, and dysfunctional mitochondrial muscles. Ageing muscles generate more reactive oxygen species (ROS), and experience decreased oxygen consumption and ATP synthesis compared to younger muscles. Additionally, changes in mitochondrial protein interactions, cristae structure, and networks may contribute to ADP insensitivity, which ultimately leads to sarcopenia. Within this framework, this review provides a comprehensive summary of our current understanding of the role of mitochondria in sarcopenia and other muscle degenerative diseases, highlighting the crucial need for further research in these areas.
Collapse
Affiliation(s)
- Nurul Tihani Kamarulzaman
- Department of Biochemistry, Faculty of Medicine, Universiti Kebangsaan Malaysia, Jalan Yaacob Latif, Bandar Tun Razak, Cheras, Kuala Lumpur, 56000, Malaysia
| | - Suzana Makpol
- Department of Biochemistry, Faculty of Medicine, Universiti Kebangsaan Malaysia, Jalan Yaacob Latif, Bandar Tun Razak, Cheras, Kuala Lumpur, 56000, Malaysia.
| |
Collapse
|
4
|
Tian Q, Greig EE, Davatzikos C, Landman BA, Resnick SM, Ferrucci L. Higher skeletal muscle mitochondrial oxidative capacity is associated with preserved brain structure up to over a decade. Nat Commun 2024; 15:10786. [PMID: 39737971 PMCID: PMC11686348 DOI: 10.1038/s41467-024-55009-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Accepted: 11/25/2024] [Indexed: 01/01/2025] Open
Abstract
Impaired muscle mitochondrial oxidative capacity is associated with future cognitive impairment, and higher levels of PET and blood biomarkers of Alzheimer's disease and neurodegeneration. Here, we examine its associations with up to over a decade-long changes in brain atrophy and microstructure. Higher in vivo skeletal muscle oxidative capacity via MR spectroscopy (post-exercise recovery rate, kPCr) is associated with less ventricular enlargement and brain aging progression, and less atrophy in specific regions, notably primary sensorimotor cortex, temporal white and gray matter, thalamus, occipital areas, cingulate cortex, and cerebellum white matter. Higher kPCr is also associated with less microstructural integrity decline in white matter around cingulate, including superior longitudinal fasciculus, corpus callosum, and cingulum. Higher in vivo muscle oxidative capacity is associated with preserved brain structure up to over a decade, particularly in areas important for cognition, motor function, and sensorimotor integration.
Collapse
Affiliation(s)
- Qu Tian
- Longitudinal Studies Section, Translational Gerontology Branch, National Institute on Aging, Baltimore, MD, USA.
| | - Erin E Greig
- Longitudinal Studies Section, Translational Gerontology Branch, National Institute on Aging, Baltimore, MD, USA
| | - Christos Davatzikos
- Radiology Department, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Bennett A Landman
- Department of Computer Science, Vanderbilt University, Nashville, TN, USA
- Department of Electrical and Computer Engineering, Vanderbilt University, Nashville, TN, USA
| | - Susan M Resnick
- Brain Aging and Behavior Section, Laboratory of Behavioral Neuroscience, National Institute on Aging, Baltimore, MD, USA
| | - Luigi Ferrucci
- Longitudinal Studies Section, Translational Gerontology Branch, National Institute on Aging, Baltimore, MD, USA
| |
Collapse
|
5
|
Ferrucci L, Guerra F, Bucci C, Marzetti E, Picca A. Mitochondria break free: Mitochondria-derived vesicles in aging and associated conditions. Ageing Res Rev 2024; 102:102549. [PMID: 39427885 DOI: 10.1016/j.arr.2024.102549] [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: 07/04/2024] [Revised: 09/27/2024] [Accepted: 10/11/2024] [Indexed: 10/22/2024]
Abstract
Mitophagy is the intracellular recycling system that disposes damaged/inefficient mitochondria and allows biogenesis of new organelles to ensure mitochondrial quality is optimized. Dysfunctional mitophagy has been implicated in human aging and diseases. Multiple evolutionarily selected, redundant mechanisms of mitophagy have been identified, but their specific roles in human health and their potential exploitation as therapeutic targets are unclear. Recently, the characterization of the endosomal-lysosomal system has revealed additional mechanisms of mitophagy and mitochondrial quality control that operate via the production of mitochondria-derived vesicles (MDVs). Circulating MDVs can be isolated and characterized to provide an unprecedented opportunity to study this type of mitochondrial recycling in vivo and to relate it to human physiology and pathology. Defining the role of MDVs in human physiology, pathology, and aging is hampered by the lack of standardized methods to isolate, validate, and characterize these vesicles. Hence, some basic questions about MDVs remain unanswered. While MDVs are generated directly through the extrusion of mitochondrial membranes within the cell, a set of circulating extracellular vesicles leaking from the endosomal-lysosomal system and containing mitochondrial portions have also been identified and warrant investigation. Preliminary research indicates that MDV generation serves multiple biological roles and contributes to restoring cell homeostasis. However, studies have shown that MDVs may also be involved in pathological conditions. Therefore, further research is warranted to establish when/whether MDVs are supporting disease progression and/or are extracting damaged mitochondrial components to alleviate cellular oxidative burden and restore redox homeoastasis. This information will be relevant for exploiting these vesicles for therapeutic purpose. Herein, we provide an overview of preclinical and clinical studies on MDVs in aging and associated conditions and discuss the interplay between MDVs and some of the hallmarks of aging (mitophagy, inflammation, and proteostasis). We also outline open questions on MDV research that should be prioritized by future investigations.
Collapse
Affiliation(s)
- Luigi Ferrucci
- Division of Intramural Research, National Institute on Aging, Baltimore, MD, USA.
| | - Flora Guerra
- Department of Biological and Environmental Sciences and Technologies, Università del Salento, Lecce, Italy
| | - Cecilia Bucci
- Department of Experimental Medicine, Università del Salento, Lecce, Italy
| | - Emanuele Marzetti
- Department of Geriatrics, Orthopedics and Rheumatology, Università Cattolica del Sacro Cuore, Rome, Italy; Fondazione Policlinico Universitario "A. Gemelli" IRCCS, Rome, Italy
| | - Anna Picca
- Fondazione Policlinico Universitario "A. Gemelli" IRCCS, Rome, Italy; Department of Medicine and Surgery, LUM University, Casamassima, Italy.
| |
Collapse
|
6
|
Ramos SV, Distefano G, Lui LY, Cawthon PM, Kramer P, Sipula IJ, Bello FM, Mau T, Jurczak MJ, Molina AJ, Kershaw EE, Marcinek DJ, Shankland E, Toledo FG, Newman AB, Hepple RT, Kritchevsky SB, Goodpaster BH, Cummings SR, Coen PM. Role of Cardiorespiratory Fitness and Mitochondrial Oxidative Capacity in Reduced Walk Speed of Older Adults With Diabetes. Diabetes 2024; 73:1048-1057. [PMID: 38551899 PMCID: PMC11189829 DOI: 10.2337/db23-0827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Accepted: 03/26/2024] [Indexed: 04/09/2024]
Abstract
Cardiorespiratory fitness and mitochondrial oxidative capacity are associated with reduced walking speed in older adults, but their impact on walking speed in older adults with diabetes has not been clearly defined. We examined differences in cardiorespiratory fitness and skeletal muscle mitochondrial oxidative capacity between older adults with and without diabetes, as well as determined their relative contribution to slower walking speed in older adults with diabetes. Participants with diabetes (n = 159) had lower cardiorespiratory fitness and mitochondrial respiration in permeabilized fiber bundles compared with those without diabetes (n = 717), following adjustments for covariates including BMI, chronic comorbid health conditions, and physical activity. Four-meter and 400-m walking speeds were slower in those with diabetes. Mitochondrial oxidative capacity alone or combined with cardiorespiratory fitness mediated ∼20-70% of the difference in walking speed between older adults with and without diabetes. Additional adjustments for BMI and comorbidities further explained the group differences in walking speed. Cardiorespiratory fitness and skeletal muscle mitochondrial oxidative capacity contribute to slower walking speeds in older adults with diabetes. ARTICLE HIGHLIGHTS
Collapse
Affiliation(s)
| | | | - Li-Yung Lui
- San Francisco Coordinating Center, California Pacific Medical Center Research Institute, San Francisco, CA
- Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, CA
| | - Peggy M. Cawthon
- San Francisco Coordinating Center, California Pacific Medical Center Research Institute, San Francisco, CA
- Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, CA
| | - Philip Kramer
- Department of Internal Medicine, Wake Forest University School of Medicine, Winston-Salem, NC
| | - Ian J. Sipula
- Division of Endocrinology and Metabolism, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Fiona M. Bello
- Division of Endocrinology and Metabolism, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Theresa Mau
- San Francisco Coordinating Center, California Pacific Medical Center Research Institute, San Francisco, CA
- Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, CA
| | - Michael J. Jurczak
- Division of Endocrinology and Metabolism, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Anthony J. Molina
- Department of Medicine, University of California, San Diego, La Jolla, CA
| | - Erin E. Kershaw
- Division of Endocrinology and Metabolism, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - David J. Marcinek
- Department of Radiology, University of Washington School of Medicine, Seattle, WA
| | - Eric Shankland
- Department of Radiology, University of Washington School of Medicine, Seattle, WA
| | - Frederico G.S. Toledo
- Division of Endocrinology and Metabolism, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Anne B. Newman
- Division of Endocrinology and Metabolism, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Russell T. Hepple
- Department of Physical Therapy, University of Florida, Gainesville, FL
| | - Stephen B. Kritchevsky
- Department of Internal Medicine, Wake Forest University School of Medicine, Winston-Salem, NC
| | | | - Steven R. Cummings
- San Francisco Coordinating Center, California Pacific Medical Center Research Institute, San Francisco, CA
- Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, CA
| | - Paul M. Coen
- Translational Research Institute, AdventHealth, Orlando, FL
| |
Collapse
|
7
|
Seo D, Lee CM, Apio C, Heo G, Timsina J, Kohlfeld P, Boada M, Orellana A, Fernandez MV, Ruiz A, Morris JC, Schindler SE, Park T, Cruchaga C, Sung YJ. Sex and aging signatures of proteomics in human cerebrospinal fluid identify distinct clusters linked to neurodegeneration. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.06.18.24309102. [PMID: 38947020 PMCID: PMC11213043 DOI: 10.1101/2024.06.18.24309102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/02/2024]
Abstract
Sex and age are major risk factors for chronic diseases. Recent studies examining age-related molecular changes in plasma provided insights into age-related disease biology. Cerebrospinal fluid (CSF) proteomics can provide additional insights into brain aging and neurodegeneration. By comprehensively examining 7,006 aptamers targeting 6,139 proteins in CSF obtained from 660 healthy individuals aged from 43 to 91 years old, we subsequently identified significant sex and aging effects on 5,097 aptamers in CSF. Many of these effects on CSF proteins had different magnitude or even opposite direction as those on plasma proteins, indicating distinctive CSF-specific signatures. Network analysis of these CSF proteins revealed not only modules associated with healthy aging but also modules showing sex differences. Through subsequent analyses, several modules were highlighted for their proteins implicated in specific diseases. Module 2 and 6 were enriched for many aging diseases including those in the circulatory systems, immune mechanisms, and neurodegeneration. Together, our findings fill a gap of current aging research and provide mechanistic understanding of proteomic changes in CSF during a healthy lifespan and insights for brain aging and diseases.
Collapse
|
8
|
Gibril BAA, Xiong X, Chai X, Xu Q, Gong J, Xu J. Unlocking the Nexus of Sirtuins: A Comprehensive Review of Their Role in Skeletal Muscle Metabolism, Development, and Disorders. Int J Biol Sci 2024; 20:3219-3235. [PMID: 38904020 PMCID: PMC11186354 DOI: 10.7150/ijbs.96885] [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: 04/02/2024] [Accepted: 05/23/2024] [Indexed: 06/22/2024] Open
Abstract
The sirtuins constitute a group of histone deacetylases reliant on NAD+ for their activity that have gained recognition for their critical roles as regulators of numerous biological processes. These enzymes have various functions in skeletal muscle biology, including development, metabolism, and the body's response to disease. This comprehensive review seeks to clarify sirtuins' complex role in skeletal muscle metabolism, including glucose uptake, fatty acid oxidation, mitochondrial dynamics, autophagy regulation, and exercise adaptations. It also examines their critical roles in developing skeletal muscle, including myogenesis, the determination of muscle fiber type, regeneration, and hypertrophic responses. Moreover, it sheds light on the therapeutic potential of sirtuins by examining their impact on a range of skeletal muscle disorders. By integrating findings from various studies, this review outlines the context of sirtuin-mediated regulation in skeletal muscle, highlighting their importance and possible consequences for health and disease.
Collapse
Affiliation(s)
| | | | | | | | | | - Jiguo Xu
- Jiangxi Provincial Key Laboratory of Poultry Genetic Improvement, Institute of Biological Technology, Nanchang Normal University, Nanchang, 330032, China
| |
Collapse
|
9
|
Lee MJC, Saner NJ, Ferri A, García-Domínguez E, Broatch JR, Bishop DJ. Delineating the contribution of ageing and physical activity to changes in mitochondrial characteristics across the lifespan. Mol Aspects Med 2024; 97:101272. [PMID: 38626488 DOI: 10.1016/j.mam.2024.101272] [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: 12/28/2023] [Revised: 03/20/2024] [Accepted: 03/22/2024] [Indexed: 04/18/2024]
Abstract
Ageing is associated with widespread physiological changes prominent within all tissues, including skeletal muscle and the brain, which lead to a decline in physical function. To tackle the growing health and economic burdens associated with an ageing population, the concept of healthy ageing has become a major research priority. Changes in skeletal muscle mitochondrial characteristics have been suggested to make an important contribution to the reductions in skeletal muscle function with age, and age-related changes in mitochondrial content, respiratory function, morphology, and mitochondrial DNA have previously been reported. However, not all studies report changes in mitochondrial characteristics with ageing, and there is increasing evidence to suggest that physical activity (or inactivity) throughout life is a confounding factor when interpreting age-associated changes. Given that physical activity is a potent stimulus for inducing beneficial adaptations to mitochondrial characteristics, delineating the influence of physical activity on the changes in skeletal muscle that occur with age is complicated. This review aims to summarise our current understanding and knowledge gaps regarding age-related changes to mitochondrial characteristics within skeletal muscle, as well as to provide some novel insights into brain mitochondria, and to propose avenues of future research and targeted interventions. Furthermore, where possible, we incorporate discussions of the modifying effects of physical activity, exercise, and training status, to purported age-related changes in mitochondrial characteristics.
Collapse
Affiliation(s)
- Matthew J-C Lee
- The Exercise Prescription Lab (EPL), Institute for Health and Sport (IHES), Victoria University, Melbourne, Victoria, Australia
| | - Nicholas J Saner
- The Exercise Prescription Lab (EPL), Institute for Health and Sport (IHES), Victoria University, Melbourne, Victoria, Australia
| | - Alessandra Ferri
- The Exercise Prescription Lab (EPL), Institute for Health and Sport (IHES), Victoria University, Melbourne, Victoria, Australia
| | - Esther García-Domínguez
- Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria, Australia; Freshage Research Group, Department of Physiology, Faculty of Medicine, University of Valencia and CIBERFES, Fundación Investigación Hospital Clínico Universitario/INCLIVA, Valencia, Spain
| | - James R Broatch
- The Exercise Prescription Lab (EPL), Institute for Health and Sport (IHES), Victoria University, Melbourne, Victoria, Australia
| | - David J Bishop
- The Exercise Prescription Lab (EPL), Institute for Health and Sport (IHES), Victoria University, Melbourne, Victoria, Australia.
| |
Collapse
|
10
|
Cummings SR, Coen PM, Ferrucci L. The cellular bases of mobility from the Study of Muscle, Mobility and Aging (SOMMA). Aging Cell 2024; 23:e14129. [PMID: 38429931 PMCID: PMC11166358 DOI: 10.1111/acel.14129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Accepted: 02/14/2024] [Indexed: 03/03/2024] Open
Abstract
Findings from the Study of Muscle, Mobility and Aging (SOMMA) in this issue of Aging Cell show that several biological pathways in skeletal muscle cells play an important role in determining mobility in older adults. These are based on assays in skeletal muscle biopsies obtained from participants, aged 70 years and older in SOMMA tested for association with assessments related to mobility, including muscle mass, strength, power, cardiopulmonary fitness, and 400 m walking speed. The papers show that, using mass spectrometry, oxidative modifications of proteins essential to myocellular function are associated with poorer mobility. Using RNA-seq to quantify gene expression, lower levels of expression of antioxidant enzymes located in mitochondria, autophagy, patterns of expression of genes involved in autophagy, and higher levels of RNA transcripts that increase with denervation were associated with poorer performance on tests of mobility. These results extend previous research from the Baltimore Longitudinal Study of Aging and recent studies from SOMMA showing the importance of mitochondrial energetics in mobility. Together, these findings are painting a picture of how fundamental cellular processes influence the loss of mobility with aging. They may also be a window on aging in other cells, tissues, and systems. The data collected in SOMMA are publicly available and SOMMA welcomes collaborations with scientists who are interested in research about human aging.
Collapse
Affiliation(s)
- Steven R. Cummings
- San Francisco Coordinating CenterCalifornia Pacific Research InstituteSan FranciscoCaliforniaUSA
- Department of Epidemiology and BiostatisticsUniversity of CaliforniaSan FranciscoCaliforniaUSA
| | - Paul M. Coen
- Translational Research Institute, AdventHealthOrlandoFloridaUSA
| | - Luigi Ferrucci
- Intramural Research Program of the National Institute on Aging, NIA, NIHBaltimoreMarylandUSA
| |
Collapse
|
11
|
Tian Q, Greig EE, Walker KA, Fishbein KW, Spencer RG, Resnick SM, Ferrucci L. Plasma metabolomic markers underlying skeletal muscle mitochondrial function relationships with cognition and motor function. Age Ageing 2024; 53:afae079. [PMID: 38615247 PMCID: PMC11484644 DOI: 10.1093/ageing/afae079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Indexed: 04/15/2024] Open
Abstract
BACKGROUND Lower skeletal muscle mitochondrial function is associated with future cognitive impairment and mobility decline, but the biological underpinnings for these associations are unclear. We examined metabolomic markers underlying skeletal muscle mitochondrial function, cognition and motor function. METHODS We analysed data from 560 participants from the Baltimore Longitudinal Study of Aging (mean age: 68.4 years, 56% women, 28% Black) who had data on skeletal muscle oxidative capacity (post-exercise recovery rate of phosphocreatine, kPCr) via 31P magnetic resonance spectroscopy and targeted plasma metabolomics using LASSO model. We then examined which kPCr-related markers were also associated with cognition and motor function in a larger sample (n = 918, mean age: 69.4, 55% women, 27% Black). RESULTS The LASSO model revealed 24 metabolites significantly predicting kPCr, with the top 5 being asymmetric dimethylarginine, lactic acid, lysophosphatidylcholine a C18:1, indoleacetic acid and triacylglyceride (17:1_34:3), also significant in multivariable linear regression. The kPCr metabolite score was associated with cognitive or motor function, with 2.5-minute usual gait speed showing the strongest association (r = 0.182). Five lipids (lysophosphatidylcholine a C18:1, phosphatidylcholine ae C42:3, cholesteryl ester 18:1, sphingomyelin C26:0, octadecenoic acid) and 2 amino acids (leucine, cystine) were associated with both cognitive and motor function measures. CONCLUSION Our findings add evidence to the hypothesis that mitochondrial function is implicated in the pathogenesis of cognitive and physical decline with aging and suggest that targeting specific metabolites may prevent cognitive and mobility decline through their effects on mitochondria. Future omics studies are warranted to confirm these findings and explore mechanisms underlying mitochondrial dysfunction in aging phenotypes.
Collapse
Affiliation(s)
- Qu Tian
- Translational Gerontology Branch, National Institute on Aging, NIH, Baltimore, MD 21224, USA
| | - Erin E Greig
- Translational Gerontology Branch, National Institute on Aging, NIH, Baltimore, MD 21224, USA
| | - Keenan A Walker
- Laboratory of Behavioral Neuroscience, National Institute on Aging, NIH, Baltimore, MD 21224, USA
| | - Kenneth W Fishbein
- Laboratory of Clinical Investigation, National Institute on Aging, NIH, Baltimore, MD 21224, USA
| | - Richard G Spencer
- Laboratory of Clinical Investigation, National Institute on Aging, NIH, Baltimore, MD 21224, USA
| | - Susan M Resnick
- Laboratory of Behavioral Neuroscience, National Institute on Aging, NIH, Baltimore, MD 21224, USA
| | - Luigi Ferrucci
- Translational Gerontology Branch, National Institute on Aging, NIH, Baltimore, MD 21224, USA
| |
Collapse
|
12
|
Cummings SR, Lui LY, Glynn NW, Mau T, Cawthon PM, Kritchevsky SB, Coen PM, Goodpaster B, Marcinek DJ, Hepple RT, Patel S, Newman AB. Energetics and clinical factors for the time required to walk 400 m: The Study of Muscle, Mobility and Aging (SOMMA). J Am Geriatr Soc 2024; 72:1035-1047. [PMID: 38243364 DOI: 10.1111/jgs.18763] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 10/31/2023] [Accepted: 12/16/2023] [Indexed: 01/21/2024]
Abstract
BACKGROUND Walking slows with aging often leading to mobility disability. Mitochondrial energetics has been found to be associated with gait speed over short distances. Additionally, walking is a complex activity but few clinical factors that may be associated with walk time have been studied. METHODS We examined 879 participants ≥70 years and measured the time to walk 400 m. We tested the hypothesis that decreased mitochondrial energetics by respirometry in muscle biopsies and magnetic resonance spectroscopy in the thigh and is associated with longer time to walk 400 m. We also used cardiopulmonary exercise testing to assess the energetic costs of walking: maximum oxygen consumption (VO2peak) and energy cost-capacity (the ratio of VO2, at a slow speed to VO2peak). In addition, we tested the hypothesis that selected clinical factors would also be associated with 400-m walk time. RESULTS Lower Max OXPHOS was associated with longer walk time, and the association was explained by the energetic costs of walking, leg power, and weight. Additionally, a multivariate model revealed that longer walk time was also significantly associated with lower VO2peak, greater cost-capacity ratio, weaker leg power, heavier weight, hip and knee stiffness, peripheral neuropathy, greater perceived exertion while walking slowly, greater physical fatigability, less moderate-to-vigorous exercise, less sedentary time, and anemia. Significant associations between age, sex, muscle mass, and peripheral artery disease with 400-m walk time were explained by other clinical and physiologic factors. CONCLUSIONS Lower mitochondrial energetics is associated with needing more time to walk 400 m. This supports the value of developing interventions to improve mitochondrial energetics. Additionally, doing more moderate-to-vigorous exercise, increasing leg power, reducing weight, treating hip and knee stiffness, and screening for and treating anemia may reduce the time required to walk 400 m and reduce the risk of mobility disability.
Collapse
Affiliation(s)
- Steven R Cummings
- San Francisco Coordinating Center, California Pacific Medical Center Research Institute, San Francisco, California, USA
- Department of Epidemiology and Biostatistics, University of California, San Francisco, California, USA
| | - Li-Yung Lui
- San Francisco Coordinating Center, California Pacific Medical Center Research Institute, San Francisco, California, USA
| | - Nancy W Glynn
- Department of Epidemiology, University of Pittsburgh Graduate School of Public Health, Pittsburgh, Pennsylvania, USA
| | - Theresa Mau
- San Francisco Coordinating Center, California Pacific Medical Center Research Institute, San Francisco, California, USA
- Department of Epidemiology and Biostatistics, University of California, San Francisco, California, USA
| | - Peggy M Cawthon
- San Francisco Coordinating Center, California Pacific Medical Center Research Institute, San Francisco, California, USA
- Department of Epidemiology and Biostatistics, University of California, San Francisco, California, USA
| | - Stephen B Kritchevsky
- Department of Internal Medicine-Gerontology and Geriatric Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
| | - Paul M Coen
- Department of Internal Medicine-Gerontology and Geriatric Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
- AdventHealth, Translational Research Institute, Orlando, Florida, USA
| | - Bret Goodpaster
- Department of Internal Medicine-Gerontology and Geriatric Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
| | - David J Marcinek
- Department of Radiology, University of Washington, Seattle, Washington, USA
| | - Russell T Hepple
- Department of Physical Therapy, University of Florida, Gainesville, Florida, USA
| | - Sheena Patel
- San Francisco Coordinating Center, California Pacific Medical Center Research Institute, San Francisco, California, USA
| | - Anne B Newman
- Department of Epidemiology, University of Pittsburgh Graduate School of Public Health, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
13
|
Marzetti E, Lozanoska-Ochser B, Calvani R, Landi F, Coelho-Júnior HJ, Picca A. Restoring Mitochondrial Function and Muscle Satellite Cell Signaling: Remedies against Age-Related Sarcopenia. Biomolecules 2024; 14:415. [PMID: 38672432 PMCID: PMC11048011 DOI: 10.3390/biom14040415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Revised: 03/24/2024] [Accepted: 03/27/2024] [Indexed: 04/28/2024] Open
Abstract
Sarcopenia has a complex pathophysiology that encompasses metabolic dysregulation and muscle ultrastructural changes. Among the drivers of intracellular and ultrastructural changes of muscle fibers in sarcopenia, mitochondria and their quality control pathways play relevant roles. Mononucleated muscle stem cells/satellite cells (MSCs) have been attributed a critical role in muscle repair after an injury. The involvement of mitochondria in supporting MSC-directed muscle repair is unclear. There is evidence that a reduction in mitochondrial biogenesis blunts muscle repair, thus indicating that the delivery of functional mitochondria to injured muscles can be harnessed to limit muscle fibrosis and enhance restoration of muscle function. Injection of autologous respiration-competent mitochondria from uninjured sites to damaged tissue has been shown to reduce infarct size and enhance cell survival in preclinical models of ischemia-reperfusion. Furthermore, the incorporation of donor mitochondria into MSCs enhances lung and cardiac tissue repair. This strategy has also been tested for regeneration purposes in traumatic muscle injuries. Indeed, the systemic delivery of mitochondria promotes muscle regeneration and restores muscle mass and function while reducing fibrosis during recovery after an injury. In this review, we discuss the contribution of altered MSC function to sarcopenia and illustrate the prospect of harnessing mitochondrial delivery and restoration of MSCs as a therapeutic strategy against age-related sarcopenia.
Collapse
Affiliation(s)
- Emanuele Marzetti
- Fondazione Policlinico Universitario “Agostino Gemelli” IRCCS, L.go A. Gemelli 8, 00168 Rome, Italy; (R.C.); (F.L.)
- Department of Geriatrics, Orthopedics and Rheumatology, Università Cattolica del Sacro Cuore, L.go F. Vito 1, 00168 Rome, Italy;
| | - Biliana Lozanoska-Ochser
- Department of Medicine and Surgery, LUM University, 70010 Casamassima, Italy;
- DAHFMO Unit of Histology and Medical Embryology, Sapienza Università di Roma, P.le Aldo Moro 5, 00185 Rome, Italy
| | - Riccardo Calvani
- Fondazione Policlinico Universitario “Agostino Gemelli” IRCCS, L.go A. Gemelli 8, 00168 Rome, Italy; (R.C.); (F.L.)
- Department of Geriatrics, Orthopedics and Rheumatology, Università Cattolica del Sacro Cuore, L.go F. Vito 1, 00168 Rome, Italy;
| | - Francesco Landi
- Fondazione Policlinico Universitario “Agostino Gemelli” IRCCS, L.go A. Gemelli 8, 00168 Rome, Italy; (R.C.); (F.L.)
- Department of Geriatrics, Orthopedics and Rheumatology, Università Cattolica del Sacro Cuore, L.go F. Vito 1, 00168 Rome, Italy;
| | - Hélio José Coelho-Júnior
- Department of Geriatrics, Orthopedics and Rheumatology, Università Cattolica del Sacro Cuore, L.go F. Vito 1, 00168 Rome, Italy;
| | - Anna Picca
- Fondazione Policlinico Universitario “Agostino Gemelli” IRCCS, L.go A. Gemelli 8, 00168 Rome, Italy; (R.C.); (F.L.)
- Department of Medicine and Surgery, LUM University, 70010 Casamassima, Italy;
| |
Collapse
|
14
|
Marzetti E, Calvani R, Coelho-Júnior HJ, Landi F, Picca A. Mitochondrial Quantity and Quality in Age-Related Sarcopenia. Int J Mol Sci 2024; 25:2052. [PMID: 38396729 PMCID: PMC10889427 DOI: 10.3390/ijms25042052] [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: 12/19/2023] [Revised: 02/01/2024] [Accepted: 02/06/2024] [Indexed: 02/25/2024] Open
Abstract
Sarcopenia, the age-associated decline in skeletal muscle mass and strength, is a condition with a complex pathophysiology. Among the factors underlying the development of sarcopenia are the progressive demise of motor neurons, the transition from fast to slow myosin isoform (type II to type I fiber switch), and the decrease in satellite cell number and function. Mitochondrial dysfunction has been indicated as a key contributor to skeletal myocyte decline and loss of physical performance with aging. Several systems have been implicated in the regulation of muscle plasticity and trophism such as the fine-tuned and complex regulation between the stimulator of protein synthesis, mechanistic target of rapamycin (mTOR), and the inhibitor of mTOR, AMP-activated protein kinase (AMPK), that promotes muscle catabolism. Here, we provide an overview of the molecular mechanisms linking mitochondrial signaling and quality with muscle homeostasis and performance and discuss the main pathways elicited by their imbalance during age-related muscle wasting. We also discuss lifestyle interventions (i.e., physical exercise and nutrition) that may be exploited to preserve mitochondrial function in the aged muscle. Finally, we illustrate the emerging possibility of rescuing muscle tissue homeostasis through mitochondrial transplantation.
Collapse
Affiliation(s)
- Emanuele Marzetti
- Fondazione Policlinico Universitario “Agostino Gemelli” IRCCS, L.go A. Gemelli 8, 00168 Rome, Italy; (R.C.); (F.L.)
- Department of Geriatrics, Orthopedics and Rheumatology, Università Cattolica del Sacro Cuore, L.go F. Vito 1, 00618 Rome, Italy;
| | - Riccardo Calvani
- Fondazione Policlinico Universitario “Agostino Gemelli” IRCCS, L.go A. Gemelli 8, 00168 Rome, Italy; (R.C.); (F.L.)
- Department of Geriatrics, Orthopedics and Rheumatology, Università Cattolica del Sacro Cuore, L.go F. Vito 1, 00618 Rome, Italy;
| | - Hélio José Coelho-Júnior
- Department of Geriatrics, Orthopedics and Rheumatology, Università Cattolica del Sacro Cuore, L.go F. Vito 1, 00618 Rome, Italy;
| | - Francesco Landi
- Fondazione Policlinico Universitario “Agostino Gemelli” IRCCS, L.go A. Gemelli 8, 00168 Rome, Italy; (R.C.); (F.L.)
- Department of Geriatrics, Orthopedics and Rheumatology, Università Cattolica del Sacro Cuore, L.go F. Vito 1, 00618 Rome, Italy;
| | - Anna Picca
- Fondazione Policlinico Universitario “Agostino Gemelli” IRCCS, L.go A. Gemelli 8, 00168 Rome, Italy; (R.C.); (F.L.)
- Department of Medicine and Surgery, LUM University, SS100 km 18, 70010 Casamassima, Italy
| |
Collapse
|
15
|
Gonsalves SG, Saligan LN, Bergeron CM, Lee PR, Fishbein KW, Spencer RG, Zampino M, Sun X, Sheng JYS, Stearns V, Carducci M, Ferrucci L, Lukkahatai N. Exploring the links of skeletal muscle mitochondrial oxidative capacity, physical functionality, and mental well-being of cancer survivors. Sci Rep 2024; 14:2669. [PMID: 38302539 PMCID: PMC10834492 DOI: 10.1038/s41598-024-52570-x] [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: 10/06/2023] [Accepted: 01/20/2024] [Indexed: 02/03/2024] Open
Abstract
Physical impairments following cancer treatment have been linked with the toxic effects of these treatments on muscle mass and strength, through their deleterious effects on skeletal muscle mitochondrial oxidative capacity. Accordingly, we designed the present study to explore relationships of skeletal muscle mitochondrial oxidative capacity with physical performance and perceived cancer-related psychosocial experiences of cancer survivors. We assessed skeletal muscle mitochondrial oxidative capacity using in vivo phosphorus-31 magnetic resonance spectroscopy (31P MRS), measuring the postexercise phosphocreatine resynthesis time constant, τPCr, in 11 post-chemotherapy participants aged 34-70 years. During the MRS procedure, participants performed rapid ballistic knee extension exercise to deplete phosphocreatine (PCr); hence, measuring the primary study outcome, which was the recovery rate of PCr (τPCr). Patient-reported outcomes of psychosocial symptoms and well-being were assessed using the Patient-Reported Outcomes Measurement Information System and the 36-Item Short Form health survey (SF-36). Rapid bioenergetic recovery, reflected through a smaller value of τPCr was associated with worse depression (rho ρ = - 0.69, p = 0.018, and Cohen's d = - 1.104), anxiety (ρ = - 0.61, p = .046, d = - 0.677), and overall mental health (ρ = 0.74, p = 0.010, d = 2.198) scores, but better resilience (ρ = 0.65, p = 0.029), and coping-self efficacy (ρ = 0.63, p = 0.04) scores. This is the first study to link skeletal muscle mitochondrial oxidative capacity with subjective reports of cancer-related behavioral toxicities. Further investigations are warranted to confirm these findings probing into the role of disease status and personal attributes in these preliminary results.
Collapse
Affiliation(s)
- Stephen G Gonsalves
- National Institute of Nursing Research, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Leorey N Saligan
- National Institute of Nursing Research, National Institutes of Health, Bethesda, MD, 20892, USA.
| | - Christopher M Bergeron
- Laboratory of Clinical Investigation, Intramural Research Program, National Institute On Aging, National Institutes of Health, Baltimore, MD, USA
| | - Philip R Lee
- Laboratory of Clinical Investigation, Intramural Research Program, National Institute On Aging, National Institutes of Health, Baltimore, MD, USA
| | - Kenneth W Fishbein
- Laboratory of Clinical Investigation, Intramural Research Program, National Institute On Aging, National Institutes of Health, Baltimore, MD, USA
| | - Richard G Spencer
- Laboratory of Clinical Investigation, Intramural Research Program, National Institute On Aging, National Institutes of Health, Baltimore, MD, USA
| | - Marta Zampino
- Department of Internal Medicine, University of Maryland, Baltimore, MD, USA
| | - Xinyi Sun
- School of Nursing, Johns Hopkins University, Baltimore, MD, USA
| | | | - Vered Stearns
- School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Michael Carducci
- School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Luigi Ferrucci
- Laboratory of Clinical Investigation, Intramural Research Program, National Institute On Aging, National Institutes of Health, Baltimore, MD, USA
| | - Nada Lukkahatai
- National Institute of Nursing Research, National Institutes of Health, Bethesda, MD, 20892, USA
- School of Nursing, Johns Hopkins University, Baltimore, MD, USA
| |
Collapse
|
16
|
Hayden CMT, Nagarajan R, Smith ZH, Gilmore S, Kent JA. Postcontraction [acetylcarnitine] reflects interindividual variation in skeletal muscle ATP production patterns in vivo. Am J Physiol Regul Integr Comp Physiol 2024; 326:R66-R78. [PMID: 37955131 DOI: 10.1152/ajpregu.00027.2023] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Revised: 11/02/2023] [Accepted: 11/02/2023] [Indexed: 11/14/2023]
Abstract
In addition to its role in substrate selection (carbohydrate vs. fat) for oxidative metabolism in muscle, acetylcarnitine production may be an important modulator of the energetic pathway by which ATP is produced. A combination of noninvasive magnetic resonance spectroscopy measures of cytosolic acetylcarnitine and ATP production pathways was used to investigate the link between [acetylcarnitine] and energy production in vivo. Intracellular metabolites were measured in the vastus lateralis muscle of eight males (mean: 28.4 yr, range: 25-35) during 8 min of incremental, dynamic contractions (0.5 Hz, 2-min stages at 6%, 9%, 12%, and 15% maximal torque) that increased [acetylcarnitine] approximately fivefold from resting levels. ATP production via oxidative phosphorylation, glycolysis, and the creatine kinase reaction was calculated based on phosphorus metabolites and pH. Spearman rank correlations indicated that postcontraction [acetylcarnitine] was positively associated with both absolute (mM) and relative (% total ATP) glycolytic ATP production (rs = 0.95, P = 0.001; rs = 0.93, P = 0.002), and negatively associated with relative (rs = -0.81, P = 0.02) but not absolute (rs = -0.14, P = 0.75) oxidative ATP production. Thus, acetylcarnitine accumulated more when there was a greater reliance on "nonoxidative" glycolysis and a relatively lower contribution from oxidative phosphorylation, reflecting the fate of pyruvate in working skeletal muscle. Furthermore, these data indicate striking interindividual variation in responses to the energy demand of submaximal contractions. Overall, the results of this preliminary study provide novel evidence of the coupling in vivo between ATP production pathways and the carnitine system.NEW & NOTEWORTHY Production of acetylcarnitine from acetyl-CoA and free carnitine may be important for energy pathway regulation in contracting skeletal muscle. Noninvasive magnetic resonance spectroscopy was used to investigate the link between acetylcarnitine and energy production in the vastus lateralis muscle during dynamic contractions (n = 8 individuals). A positive correlation between acetylcarnitine accumulation and "nonoxidative" glycolysis and an inverse relationship with oxidative phosphorylation, provides novel evidence of the coupling between ATP production and the carnitine system in vivo.
Collapse
Affiliation(s)
- Christopher M T Hayden
- Muscle Physiology Laboratory, Department of Kinesiology, University of Massachusetts, Amherst, Massachusetts, United States
| | - Rajakumar Nagarajan
- Human Magnetic Resonance Center, Institute for Applied Life Sciences, University of Massachusetts, Amherst, Massachusetts, United States
| | - Zoe H Smith
- Muscle Physiology Laboratory, Department of Kinesiology, University of Massachusetts, Amherst, Massachusetts, United States
| | - Samantha Gilmore
- Muscle Physiology Laboratory, Department of Kinesiology, University of Massachusetts, Amherst, Massachusetts, United States
| | - Jane A Kent
- Muscle Physiology Laboratory, Department of Kinesiology, University of Massachusetts, Amherst, Massachusetts, United States
| |
Collapse
|
17
|
Picca A, Faitg J, Auwerx J, Ferrucci L, D'Amico D. Mitophagy in human health, ageing and disease. Nat Metab 2023; 5:2047-2061. [PMID: 38036770 DOI: 10.1038/s42255-023-00930-8] [Citation(s) in RCA: 103] [Impact Index Per Article: 51.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Accepted: 10/13/2023] [Indexed: 12/02/2023]
Abstract
Maintaining optimal mitochondrial function is a feature of health. Mitophagy removes and recycles damaged mitochondria and regulates the biogenesis of new, fully functional ones preserving healthy mitochondrial functions and activities. Preclinical and clinical studies have shown that impaired mitophagy negatively affects cellular health and contributes to age-related chronic diseases. Strategies to boost mitophagy have been successfully tested in model organisms, and, recently, some have been translated into clinics. In this Review, we describe the basic mechanisms of mitophagy and how mitophagy can be assessed in human blood, the immune system and tissues, including muscle, brain and liver. We outline mitophagy's role in specific diseases and describe mitophagy-activating approaches successfully tested in humans, including exercise and nutritional and pharmacological interventions. We describe how mitophagy is connected to other features of ageing through general mechanisms such as inflammation and oxidative stress and forecast how strengthening research on mitophagy and mitophagy interventions may strongly support human health.
Collapse
Affiliation(s)
- Anna Picca
- Department of Medicine and Surgery, LUM University, Casamassima, Italy
- Fondazione Policlinico Universitario 'A. Gemelli' IRCCS, Rome, Italy
| | - Julie Faitg
- Amazentis, EPFL Innovation Park, Lausanne, Switzerland
| | - Johan Auwerx
- Laboratory of Integrative Systems Physiology, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Luigi Ferrucci
- Division of Intramural Research, National Institute on Aging, Baltimore, MD, USA.
| | | |
Collapse
|
18
|
Cummings SR, Lui LY, Glynn NW, Mau T, Cawthon PM, Kritchevsky SB, Coen PM, Goodpaster B, Marcinek DJ, Hepple RT, Patel S, Newman AB. Energetics and Clinical Factors for the Time Required to Walk 400 Meters The Study of Muscle, Mobility and Aging (SOMMA). MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.11.10.23298299. [PMID: 37986884 PMCID: PMC10659495 DOI: 10.1101/2023.11.10.23298299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2023]
Abstract
Background Walking slows with aging often leading to mobility disability. Mitochondrial energetics has been found to influence gait speed over short distances. Additionally, walking is a complex activity but few clinical factors that may influence walk time have been studied. Methods We examined 879 participants ≥70 years and measured the time to walk 400m. We tested the hypothesis that decreased mitochondrial energetics by respirometry in muscle biopsies and magnetic resonance spectroscopy in the thigh, is associated with longer time to walk 400m. We also used cardiopulmonary exercise testing to assess the energetic costs of walking: maximum oxygen consumption (VO 2 peak) and energy cost-capacity (the ratio of VO2, at a slow speed to VO 2 peak). In addition, we tested the hypothesis that selected clinical factors would also be associated with 400m walk time. Results Lower Max OXPHOS was associated with longer walk time and the association was explained by the energetics costs of walking, leg power and weight. Additionally, a multivariate model revealed that longer walk time was also significantly associated with lower VO 2 peak, greater cost-capacity ratio, weaker leg power, heavier weight, hip and knee stiffness, peripheral neuropathy, greater perceived exertion while walking slowly, greater physical fatigability, less moderate-to-vigorous exercise, less sedentary time and anemia. Significant associations between age, sex, muscle mass, and peripheral artery disease with 400m walk time were explained by other clinical and physiologic factors. Conclusions Lower mitochondrial energetics is associated with needing more time to walk 400m. This supports the value of developing interventions to improve mitochondrial energetics. Additionally, doing more moderate-to-vigorous exercise, increasing leg power, reducing weight, treating hip and knee stiffness, and screening for and treating anemia may reduce the time required to walk 400m and reduce the risk of mobility disability.
Collapse
|
19
|
Ramos SV, Distefano G, Lui LY, Cawthon PM, Kramer P, Sipula IJ, Bello FM, Mau T, Jurczak MJ, Molina AJ, Kershaw EE, Marcinek DJ, Toledo FGS, Newman AB, Hepple RT, Kritchevsky SB, Goodpaster BH, Cummings SR, Coen PM. Role of Cardiorespiratory Fitness and Mitochondrial Energetics in Reduced Walk Speed of Older Adults with Diabetes in the Study of Muscle, Mobility and Aging (SOMMA). MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.11.03.23297992. [PMID: 37986814 PMCID: PMC10659460 DOI: 10.1101/2023.11.03.23297992] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2023]
Abstract
Rationale Cardiorespiratory fitness and mitochondrial energetics are associated with reduced walking speed in older adults. The impact of cardiorespiratory fitness and mitochondrial energetics on walking speed in older adults with diabetes has not been clearly defined. Objective To examine differences in cardiorespiratory fitness and skeletal muscle mitochondrial energetics between older adults with and without diabetes. We also assessed the contribution of cardiorespiratory fitness and skeletal muscle mitochondrial energetics to slower walking speed in older adults with diabetes. Findings Participants with diabetes had lower cardiorespiratory fitness and mitochondrial energetics when compared to those without diabetes, following adjustments for covariates including BMI, chronic comorbid health conditions, and physical activity. 4-m and 400-m walking speeds were slower in those with diabetes. Mitochondrial oxidative capacity alone or combined with cardiorespiratory fitness mediated ∼20-70% of the difference in walk speed between older adults with and without diabetes. Further adjustments of BMI and co-morbidities further explained the group differences in walk speed. Conclusions Skeletal muscle mitochondrial energetics and cardiorespiratory fitness contribute to slower walking speeds in older adults with diabetes. Cardiorespiratory fitness and mitochondrial energetics may be therapeutic targets to maintain or improve mobility in older adults with diabetes. ARTICLE HIGHLIGHTS Why did we undertake this study? To determine if mitochondrial energetics and cardiorespiratory fitness contribute to slower walking speed in older adults with diabetes. What is the specific question(s) we wanted to answer? Are mitochondrial energetics and cardiorespiratory fitness in older adults with diabetes lower than those without diabetes? How does mitochondrial energetics and cardiorespiratory fitness impact walking speed in older adults with diabetes? What did we find? Mitochondrial energetics and cardiorespiratory fitness were lower in older adults with diabetes compared to those without diabetes, and energetics, and cardiorespiratory fitness, contributed to slower walking speed in those with diabetes. What are the implications of our findings? Cardiorespiratory fitness and mitochondrial energetics may be key therapeutic targets to maintain or improve mobility in older adults with diabetes.
Collapse
|
20
|
Tian Q, Bilgel M, Walker KA, Moghekar AR, Fishbein KW, Spencer RG, Resnick SM, Ferrucci L. Skeletal muscle mitochondrial function predicts cognitive impairment and is associated with biomarkers of Alzheimer's disease and neurodegeneration. Alzheimers Dement 2023; 19:4436-4445. [PMID: 37530130 PMCID: PMC10592411 DOI: 10.1002/alz.13388] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 06/08/2023] [Accepted: 06/12/2023] [Indexed: 08/03/2023]
Abstract
INTRODUCTION Mitochondrial dysfunction is implicated in the pathophysiology of many chronic diseases. Whether it is related to cognitive impairment and pathological markers is unknown. METHODS We examined the associations of in vivo skeletal muscle mitochondrial function (post-exercise recovery rate of phosphocreatine [kPCr] via magnetic resonance [MR] spectroscopy with future mild cognitive impairment (MCI) or dementia, and with positron emission tomography (PET) and blood biomarkers of Alzheimer's disease [AD] and neurodegeneration (i.e., Pittsburgh Compound-B [PiB] distribution volume ratio [DVR] for amyloid beta [Aβ], flortaucipir (FTP) standardized uptake value ratio [SUVR] for tau, Aβ42 /40 ratio, phosphorylated tau 181 [p-tau181], neurofilament light chain [NfL], and glial fibrillary acidic protein [GFAP]). RESULTS After covariate adjustment, each standard deviation (SD) higher kPCr level was associated with 52% lower hazards of developing MCI/dementia, and with 59% lower odds of being PiB positive with specific associations in DVR of frontal, parietal, and temporal regions, and cingulate cortex and pallidum. Higher kPCr level was also associated with lower plasma GFAP. DISCUSSION In aging, mitochondrial dysfunction may play a vital role in AD pathological changes and neuroinflammation. Highlights Higher in vivo mitochondrial function is related to lower risk of mild cognitive impairment (MCI)/dementia. Higher in vivo mitochondrial function is related to lower amyloid tracer uptake. Higher in vivo mitochondrial function is related to lower plasma neuroinflammation. Mitochondrial dysfunction may play a key role in Alzheimer's disease (AD) and neurodegeneration.
Collapse
Affiliation(s)
- Qu Tian
- Longitudinal Studies Section, Translational Gerontology Branch, National Institute on Aging, 251 Bayview Blvd., Suite 100, Baltimore, MD 21224
| | - Murat Bilgel
- Laboratory of Behavioral Neuroscience, National Institute on Aging, 251 Bayview Blvd., Suite 100, Baltimore, MD 21224
| | - Keenan A. Walker
- Laboratory of Behavioral Neuroscience, National Institute on Aging, 251 Bayview Blvd., Suite 100, Baltimore, MD 21224
| | - Abhay R. Moghekar
- Johns Hopkins University School of Medicine, 600 N Wolfe St, Baltimore, MD 21287
| | - Kenneth W. Fishbein
- Laboratory of Clinical Investigation, National Institute on Aging, 251 Bayview Blvd., Suite 100, Baltimore, MD 21224
| | - Richard G. Spencer
- Laboratory of Clinical Investigation, National Institute on Aging, 251 Bayview Blvd., Suite 100, Baltimore, MD 21224
| | - Susan M. Resnick
- Laboratory of Behavioral Neuroscience, National Institute on Aging, 251 Bayview Blvd., Suite 100, Baltimore, MD 21224
| | - Luigi Ferrucci
- Longitudinal Studies Section, Translational Gerontology Branch, National Institute on Aging, 251 Bayview Blvd., Suite 100, Baltimore, MD 21224
| |
Collapse
|
21
|
Tian Q, Lee PR, Walker KA, Ferrucci L. Energizing Mitochondria to Prevent Mobility Loss in Aging: Rationale and Hypotheses. Exerc Sport Sci Rev 2023; 51:96-102. [PMID: 37057904 PMCID: PMC10258139 DOI: 10.1249/jes.0000000000000315] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/15/2023]
Abstract
Based on recent studies from our group and others, we hypothesize that mitochondrial dysfunction during aging may be the root cause of mobility decline through deficits in the musculoskeletal and central nervous systems. Mitochondrial dysfunction could be a therapeutic target to prevent mobility decline in aging.
Collapse
Affiliation(s)
- Qu Tian
- Longitudinal Studies Section, Translational Gerontology Branch, National Institute on Aging, Baltimore, MD 21224
| | - Philip R. Lee
- Longitudinal Studies Section, Translational Gerontology Branch, National Institute on Aging, Baltimore, MD 21224
| | - Keenan A. Walker
- Laboratory of Behavioral Neuroscience, National Institute on Aging, Baltimore, MD 21224
| | - Luigi Ferrucci
- Longitudinal Studies Section, Translational Gerontology Branch, National Institute on Aging, Baltimore, MD 21224
| |
Collapse
|
22
|
Reiter DA, Bellissimo MP, Zhou L, Boebinger S, Wells GD, Jones DP, Ziegler TR, Alvarez JA, Fleischer CC. Increased adiposity is associated with altered skeletal muscle energetics. J Appl Physiol (1985) 2023; 134:1083-1092. [PMID: 36759162 PMCID: PMC10125027 DOI: 10.1152/japplphysiol.00387.2022] [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: 07/05/2022] [Revised: 02/08/2023] [Accepted: 02/08/2023] [Indexed: 02/11/2023] Open
Abstract
The objective of this pilot study was to characterize relationships between skeletal muscle energy metabolism and body composition in healthy adults with varied amounts and distribution of adipose tissue. In vivo muscle energetics were quantified using dynamic 31P magnetic resonance spectroscopy with knee extension exercise standardized to subject lean body mass. Spearman's correlation analysis examined relationships between muscle metabolism indices and measures of adiposity including body mass index (BMI), total body fat, and quadriceps intermuscular adipose tissue (IMAT). Post hoc partial correlations were examined controlling for additional body composition measures. Kruskal-Wallis tests with Dunn-Sidak post hoc comparisons evaluated group differences in energy metabolism based on body composition profiles (i.e., lean, normal-weight obese, and overweight-obese) and IMAT tertiles. BMI negatively correlated with end-exercise muscle pH after correcting for IMAT and total body fat (r = -0.46, P = 0.034). Total adiposity negatively correlated with maximum oxidative capacity after correcting for IMAT (r = -0.54, P = 0.013). IMAT positively correlated with muscle proton buffering capacity after correcting for total body fat (r = 0.53, P = 0.023). Body composition groups showed differences in end-exercise fall in [PCr] with normalized workload (P = 0.036; post hoc: overweight-obese < lean, P = 0.029) and maximum oxidative capacity (P = 0.021; post hoc: normal-weight obese < lean, P = 0.016). IMAT tertiles showed differences in end-exercise fall in [PCr] with normalized workload (P = 0.035; post hoc: 3rd < 1st, P = 0.047). Taken together, these results support increased adiposity is associated with reduced muscle energetic efficiency with more reliance on glycolysis, and when accompanied with reduced lean mass, is associated with reduced maximum oxidative capacity.NEW & NOTEWORTHY Skeletal muscle energy production is influenced by both lean body mass and adipose tissue but the effect of their distribution on energy metabolism is unclear. This study examined variations in quadriceps muscle energy metabolism in healthy adults with varied relative amounts of lean and adipose tissue. Results suggest increased adiposity is associated with reduced muscle energetic efficiency with more reliance on glycolysis, and when accompanied with reduced lean mass, is associated with reduced maximum oxidative capacity.
Collapse
Affiliation(s)
- David A Reiter
- Department of Radiology and Imaging Sciences, Emory University School of Medicine, Atlanta, Georgia, United States
- Department of Orthopedics, Emory University School of Medicine, Atlanta, Georgia, United States
- Department of Biomedical Engineering, Emory University and Georgia Tech, Atlanta, Georgia, United States
| | - Moriah P Bellissimo
- Department of Medicine, Emory University School of Medicine, Atlanta, Georgia, United States
- Department of Internal Medicine, Virginia Commonwealth University School of Medicine, Richmond, Virginia, United States
| | - Lei Zhou
- Department of Radiology and Imaging Sciences, Emory University School of Medicine, Atlanta, Georgia, United States
| | - Scott Boebinger
- Department of Biomedical Engineering, Emory University and Georgia Tech, Atlanta, Georgia, United States
| | - Greg D Wells
- Department of Translational Medicine, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Dean P Jones
- Department of Medicine, Emory University School of Medicine, Atlanta, Georgia, United States
| | - Thomas R Ziegler
- Department of Medicine, Emory University School of Medicine, Atlanta, Georgia, United States
| | - Jessica A Alvarez
- Department of Medicine, Emory University School of Medicine, Atlanta, Georgia, United States
| | - Candace C Fleischer
- Department of Radiology and Imaging Sciences, Emory University School of Medicine, Atlanta, Georgia, United States
- Department of Biomedical Engineering, Emory University and Georgia Tech, Atlanta, Georgia, United States
| |
Collapse
|
23
|
Brisendine MH, Drake JC. Early-stage Alzheimer's disease: are skeletal muscle and exercise the key? J Appl Physiol (1985) 2023; 134:515-520. [PMID: 36656981 PMCID: PMC9970658 DOI: 10.1152/japplphysiol.00659.2022] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 01/13/2023] [Accepted: 01/13/2023] [Indexed: 01/21/2023] Open
Abstract
Alzheimer's disease (AD) is the most common form of dementia affecting approximately 6.5 million people in the United States alone. The development of AD progresses over a span of years to possibly decades before resulting in cognitive impairment and clinically diagnosed AD. The time leading up to a clinical diagnosis is known as the preclinical phase, a time in which recent literature has noted a more severe loss of body mass and more specifically lean muscle mass and strength prior to diagnosis. Mitochondria dysfunction in neurons is also closely associated with AD, and mitochondrial dysfunction has been seen to occur in skeletal muscle with mild cognitive impairment prior to AD manifestation. Evidence from animal models of AD suggests a close link among skeletal muscle mass, mitochondria function, and cognition. Exercise is a powerful stimulus for improving mitochondria function and muscle health, and its benefits to cognition have been suggested as a possible therapeutic strategy for AD. However, evidence for beneficial effects of exercise in AD-afflicted populations and animal models has produced conflicting results. In this mini-review, we discuss these findings and highlight potential avenues for further investigation that may lead to the implementation of exercise as a therapeutic intervention to delay or prevent the development of AD.
Collapse
Affiliation(s)
- Matthew H Brisendine
- Department of Human Nutrition, Foods, and Exercise, Virginia Polytechnic Institute and State University, Blacksburg, Virginia, United States
| | - Joshua C Drake
- Department of Human Nutrition, Foods, and Exercise, Virginia Polytechnic Institute and State University, Blacksburg, Virginia, United States
| |
Collapse
|
24
|
Boyer KA, Hayes KL, Umberger BR, Adamczyk PG, Bean JF, Brach JS, Clark BC, Clark DJ, Ferrucci L, Finley J, Franz JR, Golightly YM, Hortobágyi T, Hunter S, Narici M, Nicklas B, Roberts T, Sawicki G, Simonsick E, Kent JA. Age-related changes in gait biomechanics and their impact on the metabolic cost of walking: Report from a National Institute on Aging workshop. Exp Gerontol 2023; 173:112102. [PMID: 36693530 PMCID: PMC10008437 DOI: 10.1016/j.exger.2023.112102] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 01/09/2023] [Accepted: 01/19/2023] [Indexed: 01/22/2023]
Abstract
Changes in old age that contribute to the complex issue of an increased metabolic cost of walking (mass-specific energy cost per unit distance traveled) in older adults appear to center at least in part on changes in gait biomechanics. However, age-related changes in energy metabolism, neuromuscular function and connective tissue properties also likely contribute to this problem, of which the consequences are poor mobility and increased risk of inactivity-related disease and disability. The U.S. National Institute on Aging convened a workshop in September 2021 with an interdisciplinary group of scientists to address the gaps in research related to the mechanisms and consequences of changes in mobility in old age. The goal of the workshop was to identify promising ways to move the field forward toward improving gait performance, decreasing energy cost, and enhancing mobility for older adults. This report summarizes the workshop and brings multidisciplinary insight into the known and potential causes and consequences of age-related changes in gait biomechanics. We highlight how gait mechanics and energy cost change with aging, the potential neuromuscular mechanisms and role of connective tissue in these changes, and cutting-edge interventions and technologies that may be used to measure and improve gait and mobility in older adults. Key gaps in the literature that warrant targeted research in the future are identified and discussed.
Collapse
Affiliation(s)
- Katherine A Boyer
- Department of Kinesiology, University of Massachusetts Amherst, MA, USA; Department of Orthopedics and Physical Rehabilitation, University of Massachusetts Medical School, Worcester, MA, USA.
| | - Kate L Hayes
- Department of Kinesiology, University of Massachusetts Amherst, MA, USA
| | | | | | - Jonathan F Bean
- New England GRECC, VA Boston Healthcare System, Boston, MA, USA; Department of PM&R, Harvard Medical School, Boston, MA, USA; Spaulding Rehabilitation Hospital, Boston, MA, USA
| | - Jennifer S Brach
- Department of Physical Therapy, University of Pittsburgh, Pittsburgh, PA, USA
| | - Brian C Clark
- Ohio Musculoskeletal and Neurological Institute and the Department of Biomedical Sciences, Ohio University, Athens, OH, USA
| | - David J Clark
- Brain Rehabilitation Research Center, Malcom Randall VA Medical Center, Gainesville, FL, USA; Department of Physiology and Aging, University of Florida, Gainesville, FL, USA
| | - Luigi Ferrucci
- Intramural Research Program of the National Institute on Aging, NIH, Baltimore, MD, USA
| | - James Finley
- Division of Biokinesiology and Physical Therapy, University of Southern California, Los Angeles, CA, USA
| | - Jason R Franz
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Chapel Hill, NC, USA
| | - Yvonne M Golightly
- College of Allied Health Professions, University of Nebraska Medical Center, Omaha, NE, USA; Thurston Arthritis Research Center, Division of Rheumatology, Allergy, and Immunology, Department of Medicine, University of North Carolina, Chapel Hill, NC, USA
| | - Tibor Hortobágyi
- Hungarian University of Sports Science, Department of Kinesiology, Budapest, Hungary; Institute of Sport Sciences and Physical Education, University of Pécs, Hungary; Somogy County Kaposi Mór Teaching Hospital, Kaposvár, Hungary; Center for Human Movement Sciences, University of Groningen Medical Center, Groningen, the Netherlands
| | - Sandra Hunter
- Department of Physical Therapy, Marquette University, Milwaukee, WI, USA
| | - Marco Narici
- Neuromuscular Physiology Laboratory, Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Barbara Nicklas
- Section on Gerontology and Geriatric Medicine, Wake Forest University School of Medicine, USA
| | - Thomas Roberts
- Department of Ecology and Evolutionary Biology, Brown University, USA
| | - Gregory Sawicki
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, USA
| | - Eleanor Simonsick
- Intramural Research Program of the National Institute on Aging, NIH, Baltimore, MD, USA
| | - Jane A Kent
- Department of Kinesiology, University of Massachusetts Amherst, MA, USA
| |
Collapse
|
25
|
Eck BL, Yang M, Elias JJ, Winalski CS, Altahawi F, Subhas N, Li X. Quantitative MRI for Evaluation of Musculoskeletal Disease: Cartilage and Muscle Composition, Joint Inflammation, and Biomechanics in Osteoarthritis. Invest Radiol 2023; 58:60-75. [PMID: 36165880 PMCID: PMC10198374 DOI: 10.1097/rli.0000000000000909] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
ABSTRACT Magnetic resonance imaging (MRI) is a valuable tool for evaluating musculoskeletal disease as it offers a range of image contrasts that are sensitive to underlying tissue biochemical composition and microstructure. Although MRI has the ability to provide high-resolution, information-rich images suitable for musculoskeletal applications, most MRI utilization remains in qualitative evaluation. Quantitative MRI (qMRI) provides additional value beyond qualitative assessment via objective metrics that can support disease characterization, disease progression monitoring, or therapy response. In this review, musculoskeletal qMRI techniques are summarized with a focus on techniques developed for osteoarthritis evaluation. Cartilage compositional MRI methods are described with a detailed discussion on relaxometric mapping (T 2 , T 2 *, T 1ρ ) without contrast agents. Methods to assess inflammation are described, including perfusion imaging, volume and signal changes, contrast-enhanced T 1 mapping, and semiquantitative scoring systems. Quantitative characterization of structure and function by bone shape modeling and joint kinematics are described. Muscle evaluation by qMRI is discussed, including size (area, volume), relaxometric mapping (T 1 , T 2 , T 1ρ ), fat fraction quantification, diffusion imaging, and metabolic assessment by 31 P-MR and creatine chemical exchange saturation transfer. Other notable technologies to support qMRI in musculoskeletal evaluation are described, including magnetic resonance fingerprinting, ultrashort echo time imaging, ultrahigh-field MRI, and hybrid MRI-positron emission tomography. Challenges for adopting and using qMRI in musculoskeletal evaluation are discussed, including the need for metal artifact suppression and qMRI standardization.
Collapse
Affiliation(s)
- Brendan L. Eck
- Program of Advanced Musculoskeletal Imaging, Cleveland Clinic, Cleveland, OH, USA
- Imaging Instute, Cleveland Clinic, Cleveland, OH, USA
| | - Mingrui Yang
- Program of Advanced Musculoskeletal Imaging, Cleveland Clinic, Cleveland, OH, USA
- Department of Biomedical Engineering, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - John J. Elias
- Program of Advanced Musculoskeletal Imaging, Cleveland Clinic, Cleveland, OH, USA
- Department of Research, Cleveland Clinic Akron General, Akron, OH, USA
| | - Carl S. Winalski
- Program of Advanced Musculoskeletal Imaging, Cleveland Clinic, Cleveland, OH, USA
- Imaging Instute, Cleveland Clinic, Cleveland, OH, USA
- Department of Biomedical Engineering, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Faysal Altahawi
- Program of Advanced Musculoskeletal Imaging, Cleveland Clinic, Cleveland, OH, USA
- Imaging Instute, Cleveland Clinic, Cleveland, OH, USA
| | - Naveen Subhas
- Program of Advanced Musculoskeletal Imaging, Cleveland Clinic, Cleveland, OH, USA
- Imaging Instute, Cleveland Clinic, Cleveland, OH, USA
| | - Xiaojuan Li
- Program of Advanced Musculoskeletal Imaging, Cleveland Clinic, Cleveland, OH, USA
- Imaging Instute, Cleveland Clinic, Cleveland, OH, USA
- Department of Biomedical Engineering, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| |
Collapse
|
26
|
Yan ZW, Yang Z, Yang J, Chen YF, Zhang XB, Song CL. Tai Chi for spatiotemporal gait features and dynamic balancing capacity in elderly female patients with non-specific low back pain: A six-week randomized controlled trial. J Back Musculoskelet Rehabil 2022; 35:1311-1319. [PMID: 35599469 DOI: 10.3233/bmr-210247] [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] [Indexed: 02/04/2023]
Abstract
BACKGROUND Non-specific low back pain (NS-LBP) is a serious public health problem. Tai Chi is promising in reducing the risk of falls and alleviating symptoms in this population. OBJECTIVE To investigate the effect of Tai Chi on gait and dynamic balance in elderly women with NS-LBP. METHODS 20 women (age > 65 yr.) with NS-LBP were randomly assigned to a Tai Chi group (n= 10) or a control group (n= 10). The Tai Chi group practiced Tai Chi exercise 3 times a week for 6 weeks. Each session lasted 60 minutes. Pain, spatiotemporal gait features and dynamic balancing capacity were assessed at 0 and 6 weeks. RESULTS Compared to the control group at 6 weeks, the Tai Chi group had a significant decrease in VAS (p= 0.027) and stride width (p= 0.019), significant improvement in gait velocity, stride length (p< 0.001). Regarding dynamic balance capacity, the Tai Chi group had significant improvements in anterior (Left: p= 0.001; Right: p= 0.038), postero-lateral (Left: p< 0.001; Right: p= 0.038), and postero-medial (Left: p= 0.015; Right: p= 0.018). CONCLUSION 6-week Tai Chi can relieve pain and improve gait and dynamic balance in elderly women with NS-LBP, which suggests Tai Chi could be a promising rehabilitation intervention to reduce the risk of falls in this population.
Collapse
Affiliation(s)
- Zhi-Wei Yan
- Department of Sports Rehabilitation, College of Human Kinesiology, Shenyang Sport University, Liaoning, China
| | - Zhen Yang
- Moray House School of Education and Sport, University of Edinburgh, Edinburgh, UK
| | - Jinghui Yang
- Department of Physical Therapy, New York University, New York, NY, USA
| | - Ya-Feng Chen
- Affiliated Hospital of Liaoning University of Traditional Chinese Medicine, Shenyang, Liaoning, China
| | - Xue-Bin Zhang
- Department of Sports Rehabilitation, College of Human Kinesiology, Shenyang Sport University, Liaoning, China
| | - Cheng-Lin Song
- Department of Sports Rehabilitation, College of Human Kinesiology, Shenyang Sport University, Liaoning, China
| |
Collapse
|
27
|
Age-Related Changes in Skeletal Muscle Oxygen Utilization. J Funct Morphol Kinesiol 2022; 7:jfmk7040087. [PMID: 36278748 PMCID: PMC9590092 DOI: 10.3390/jfmk7040087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 09/07/2022] [Accepted: 09/08/2022] [Indexed: 11/17/2022] Open
Abstract
The cardiovascular and skeletal muscle systems are intrinsically interconnected, sharing the goal of delivering oxygen to metabolically active tissue. Deficiencies within those systems that affect oxygen delivery to working tissues are a hallmark of advancing age. Oxygen delivery and utilization are reflected as muscle oxygen saturation (SmO2) and are assessed using near-infrared resonance spectroscopy (NIRS). SmO2 has been observed to be reduced by ~38% at rest, ~24% during submaximal exercise, and ~59% during maximal exercise with aging (>65 y). Furthermore, aging prolongs restoration of SmO2 back to baseline by >50% after intense exercise. Regulatory factors that contribute to reduced SmO2 with age include blood flow, capillarization, endothelial cells, nitric oxide, and mitochondrial function. These mechanisms are governed by reactive oxygen species (ROS) at the cellular level. However, mishandling of ROS with age ultimately leads to alterations in structure and function of the regulatory factors tasked with maintaining SmO2. The purpose of this review is to provide an update on the current state of the literature regarding age-related effects in SmO2. Furthermore, we attempt to bridge the gap between SmO2 and associated underlying mechanisms affected by aging.
Collapse
|
28
|
Walker KA, Basisty N, Wilson DM, Ferrucci L. Connecting aging biology and inflammation in the omics era. J Clin Invest 2022; 132:e158448. [PMID: 35838044 PMCID: PMC9282936 DOI: 10.1172/jci158448] [Citation(s) in RCA: 83] [Impact Index Per Article: 27.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Aging is characterized by the accumulation of damage to macromolecules and cell architecture that triggers a proinflammatory state in blood and solid tissues, termed inflammaging. Inflammaging has been implicated in the pathogenesis of many age-associated chronic diseases as well as loss of physical and cognitive function. The search for mechanisms that underlie inflammaging focused initially on the hallmarks of aging, but it is rapidly expanding in multiple directions. Here, we discuss the threads connecting cellular senescence and mitochondrial dysfunction to impaired mitophagy and DNA damage, which may act as a hub for inflammaging. We explore the emerging multi-omics efforts that aspire to define the complexity of inflammaging - and identify molecular signatures and novel targets for interventions aimed at counteracting excessive inflammation and its deleterious consequences while preserving the physiological immune response. Finally, we review the emerging evidence that inflammation is involved in brain aging and neurodegenerative diseases. Our goal is to broaden the research agenda for inflammaging with an eye on new therapeutic opportunities.
Collapse
Affiliation(s)
- Keenan A. Walker
- Intramural Research Program of the National Institute on Aging, NIH, Baltimore, Maryland, USA
| | - Nathan Basisty
- Intramural Research Program of the National Institute on Aging, NIH, Baltimore, Maryland, USA
| | - David M. Wilson
- Biomedical Research Institute, Faculty of Medicine and Life Sciences, Hasselt University, Diepenbeek, Belgium
| | - Luigi Ferrucci
- Intramural Research Program of the National Institute on Aging, NIH, Baltimore, Maryland, USA
| |
Collapse
|
29
|
Abstract
Frailty is a complex syndrome affecting a growing sector of the global population as medical developments have advanced human mortality rates across the world. Our current understanding of frailty is derived from studies conducted in the laboratory as well as the clinic, which have generated largely phenotypic information. Far fewer studies have uncovered biological underpinnings driving the onset and progression of frailty, but the stage is set to advance the field with preclinical and clinical assessment tools, multiomics approaches together with physiological and biochemical methodologies. In this article, we provide comprehensive coverage of topics regarding frailty assessment, preclinical models, interventions, and challenges as well as clinical frameworks and prevalence. We also identify central biological mechanisms that may be at play including mitochondrial dysfunction, epigenetic alterations, and oxidative stress that in turn, affect metabolism, stress responses, and endocrine and neuromuscular systems. We review the role of metabolic syndrome, insulin resistance and visceral obesity, focusing on glucose homeostasis, adenosine monophosphate-activated protein kinase (AMPK), mammalian target of rapamycin (mTOR), and nicotinamide adenine dinucleotide (NAD+ ) as critical players influencing the age-related loss of health. We further focus on how immunometabolic dysfunction associates with oxidative stress in promoting sarcopenia, a key contributor to slowness, weakness, and fatigue. We explore the biological mechanisms involved in stem cell exhaustion that affect regeneration and may contribute to the frailty-associated decline in resilience and adaptation to stress. Together, an overview of the interplay of aging biology with genetic, lifestyle, and environmental factors that contribute to frailty, as well as potential therapeutic targets to lower risk and slow the progression of ongoing disease is covered. © 2022 American Physiological Society. Compr Physiol 12:1-46, 2022.
Collapse
Affiliation(s)
- Laís R. Perazza
- Department of Physical Therapy and Athletic Training, Boston University, Boston, Massachusetts, USA
| | - Holly M. Brown-Borg
- Department of Biomedical Sciences, University of North Dakota, Grand Forks, North Dakota, USA
| | - LaDora V. Thompson
- Department of Physical Therapy and Athletic Training, Boston University, Boston, Massachusetts, USA
| |
Collapse
|
30
|
Metabolites Associated with Memory and Gait: A Systematic Review. Metabolites 2022; 12:metabo12040356. [PMID: 35448544 PMCID: PMC9024701 DOI: 10.3390/metabo12040356] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 04/08/2022] [Accepted: 04/13/2022] [Indexed: 01/19/2023] Open
Abstract
We recently found that dual decline in memory and gait speed was consistently associated with an increased risk of dementia compared to decline in memory or gait only or no decline across six aging cohorts. The mechanisms underlying this relationship are unknown. We hypothesize that individuals who experience dual decline may have specific pathophysiological pathways to dementia which can be indicated by specific metabolomic signatures. Here, we summarize blood-based metabolites that are associated with memory and gait from existing literature and discuss their relevant pathways. A total of 39 eligible studies were included in this systematic review. Metabolites that were associated with memory and gait belonged to five shared classes: sphingolipids, fatty acids, phosphatidylcholines, amino acids, and biogenic amines. The sphingolipid metabolism pathway was found to be enriched in both memory and gait impairments. Existing data may suggest that metabolites from sphingolipids and the sphingolipid metabolism pathway are important for both memory and gait impairments. Future studies using empirical data across multiple cohorts are warranted to identify metabolomic signatures of dual decline in memory and gait and to further understand its relationship with future dementia risk.
Collapse
|
31
|
Longitudinal associations between blood lysophosphatidylcholines and skeletal muscle mitochondrial function. GeroScience 2022; 44:2213-2221. [PMID: 35389191 PMCID: PMC9616971 DOI: 10.1007/s11357-022-00548-w] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Accepted: 03/16/2022] [Indexed: 12/11/2022] Open
Abstract
Lysophosphatidylcholines (LPCs) are phospholipids critical in the synthesis of cardiolipin, an essential component of mitochondrial membranes. Lower plasma LPCs have been cross-sectionally associated with lower skeletal muscle mitochondrial function, but whether lower LPCs and their decline over time are longitudinally associated with an accelerated decline of mitochondria function is unknown. We analyzed data from 184 participants in the Baltimore Longitudinal Study of Aging (mean age: 74.5 years, 57% women, 25% black) who had repeated measures of plasma LPCs (16:0, 16:1, 17:0, 18:0, 18:1, 18:2, 20:3, 20:4, 24:0, and 28:1) by liquid chromatography-tandem mass spectrometry and repeated measures of skeletal muscle oxidative capacity (kPCr) assessed by 31P magnetic resonance spectroscopy over an average of 2.4 years. Rates of change in kPCr and each LPC were first estimated using simple linear regression. In multivariable linear regression models adjusted for baseline demographics and PCr % depletion, lower baseline LPC 16:1 and faster rates of decline in LPC 16:1 and 18:1 were significantly associated with a faster rate of decline in kPCr (B = - 0.169, 95% CI: - 0.328, - 0.010, p = 0.038; B = 0.209, 95% CI: 0.065, 0.352, p = 0.005; B = 0.156, 95% CI: 0.011, 0.301, p = 0.035, respectively). Rates of change in other LPCs were not significantly associated with change in kPCr (all p > 0.05). Lower baseline concentrations and faster decline in selected plasma lysophosphatidylcholines over time are associated with faster decline in skeletal muscle mitochondrial function. Strategies to prevent the decline of plasma LPCs at an early stage may slow down mitochondrial function decline and impairment during aging.
Collapse
|
32
|
Linoleic Acid Intake and Physical Function: Pilot Results from the Health ABC Energy Expenditure Sub-Study. ADVANCES IN GERIATRIC MEDICINE AND RESEARCH 2022; 4. [PMID: 35368862 PMCID: PMC8975246 DOI: 10.20900/agmr20220001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Background: Dietary fat quality is important for health and physical functioning in older adults. Linoleic acid is a dietary polyunsaturated fatty acid that is necessary for optimal inner-mitochondrial membrane function. However, limited evidence exists for examining the role of linoleic acid intake on indices of mobility and physical function. In this pilot study, we sought to examine the associations between linoleic acid intake and physical functioning in older adults. Methods: This secondary analysis of data from the Health, Aging, and Body Composition energy expenditure sub-study was conducted for our investigation. Ability to complete physical tasks such as climbing a flight of stairs, walking a quarter mile, and lifting 10 lbs. was self-reported. Daily linoleic acid intake was estimated from a food frequency questionnaire. Persons with daily linoleic acid intake below approximately 85% of Adequate Intake were considered as having low linoleic acid intake. Covariate-adjusted logistic models were used for the analyses. Results: The final analytical sample included 317 participants aged 74.4 ± 2.8 years who consumed 18.9 ± 11.4 g/day of linoleic acid, with 78 (24.6%) participants having low daily linoleic acid intake. Persons with low daily linoleic acid intake had 2.58 (95% confidence interval: 1.27–5.24) greater odds for a limitation in climbing stairs. Conclusions: Our pilot investigation found that low daily linoleic acid intake could be associated with physical function in older adults. Dietitians working with older patients may want to consider the importance of daily linoleic acid intake for health and certain physical function tasks.
Collapse
|
33
|
Oberdier MT, AlGhatrif M, Adelnia F, Zampino M, Morrell CH, Simonsick E, Fishbein K, Lakatta EG, McDermott MM, Ferrucci L. Ankle-Brachial Index and Energy Production in People Without Peripheral Artery Disease: The BLSA. J Am Heart Assoc 2022; 11:e019014. [PMID: 35253449 PMCID: PMC9075330 DOI: 10.1161/jaha.120.019014] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Accepted: 10/04/2021] [Indexed: 11/16/2022]
Abstract
Background Lower ankle-brachial index (ABI) values within the 0.90 to 1.40 range are associated with poorer mitochondrial oxidative capacity of thigh muscles in cross-sectional analyses. Whether ABI decline is associated with greater declines in thigh muscle oxidative capacity with aging is unknown. Method and Results We analyzed data from 228 participants (100 men) of the BLSA (Baltimore Longitudinal Study of Aging), aged 39 to 97 years, with an ABI between 0.9 and 1.40 at baseline and at follow-up (mean follow-up period of 2.8 years). We examined mitochondrial oxidative capacity of the left thigh muscle, by measuring the postexercise phosphocreatine recovery rate constant (kPCr) from phosphorus-31 magnetic resonance spectroscopy. Greater kPCr indicated higher mitochondrial oxidative capacity. Although kPCr was available on the left leg only, ABI was measured in both legs. Longitudinal rates of change (Change) of left and right ABI and kPCr of the left thigh muscle were estimated using linear mixed effects models, and their association was analyzed by standardized multiple linear regressions. In multivariate analysis including sex, age, baseline kPCr, both left and right baseline ABI, and ABI change in both legs, (kPCr)Change was directly associated with ipsilateral (left) (ABI)Change (standardized [STD]-β=0.14; P=0.0168) but not with contralateral (right) (ABI)Change (P=0.22). Adjusting for traditional cardiovascular risk factors, this association remained significant (STD-β=0.18; P=0.0051). (kPCr)Change was steeper in White race participants (STD-β=0.16; P=0.0122) and body mass index (STD-β=0.13; P=0.0479). There was no significant association with current smoking status (P=0.63), fasting glucose (P=0.28), heart rate (P=0.67), mean blood pressure (P=0.78), and low-density lipoprotein (P=0.75), high-density lipoprotein (P=0.82), or triglycerides (P=0.15). Conclusions In people without peripheral arterial disease, greater decline in ABI over time, but not baseline ABI, was associated with faster decline in thigh mitochondrial oxidative capacity in the ipsilateral leg. Further studies are needed to examine whether early interventions that improve lower extremity muscle perfusion can improve and prevent the decline of muscle energetics.
Collapse
Affiliation(s)
- Matt T. Oberdier
- Laboratory of Cardiovascular ScienceNational Institute on AgingBaltimoreMD
- Longitudinal Studies SectionNational Institute on AgingBaltimoreMD
| | - Majd AlGhatrif
- Laboratory of Cardiovascular ScienceNational Institute on AgingBaltimoreMD
- Longitudinal Studies SectionNational Institute on AgingBaltimoreMD
- Department of MedicineJohns Hopkins School of MedicineBaltimoreMD
| | - Fatemeh Adelnia
- Longitudinal Studies SectionNational Institute on AgingBaltimoreMD
| | - Marta Zampino
- Longitudinal Studies SectionNational Institute on AgingBaltimoreMD
| | - Christopher H. Morrell
- Laboratory of Cardiovascular ScienceNational Institute on AgingBaltimoreMD
- Loyola University MarylandBaltimoreMD
| | | | - Kenneth Fishbein
- Laboratory of Clinical InvestigationNational Institute on AgingBaltimoreMD
| | - Edward G. Lakatta
- Laboratory of Cardiovascular ScienceNational Institute on AgingBaltimoreMD
| | - Mary M. McDermott
- Department of MedicineNorthwestern University Feinberg School of MedicineChicagoIL
| | - Luigi Ferrucci
- Longitudinal Studies SectionNational Institute on AgingBaltimoreMD
| |
Collapse
|
34
|
Tian Q, Mitchell BA, Zampino M, Fishbein KW, Spencer RG, Ferrucci L. Muscle mitochondrial energetics predicts mobility decline in well-functioning older adults: The baltimore longitudinal study of aging. Aging Cell 2022; 21:e13552. [PMID: 35048491 PMCID: PMC8844110 DOI: 10.1111/acel.13552] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 11/18/2021] [Accepted: 01/05/2022] [Indexed: 12/31/2022] Open
Abstract
Background Muscle mitochondrial dysfunction is associated with poor mobility in aging. Whether mitochondrial dysfunction predicts subsequent mobility decline is unknown. Methods We examined 380 cognitively normal participants aged 60 and older (53%women, 22%Black) who were well‐functioning (gait speed ≥ 1.0 m/s) and free of Parkinson's disease and stroke at baseline and had data on baseline skeletal muscle oxidative capacity and one or more mobility assessments during an average 2.5 years. Muscle oxidative capacity was measured by phosphorus magnetic resonance spectroscopy as the post‐exercise recovery rate of phosphocreatine (kPCr). Mobility was measured by four walking tests. Associations of baseline kPCr with mobility changes were examined using linear mixed‐effects models, adjusted for covariates. In a subset, we examined whether changes in muscle strength and mass affected these associations by adjusting for longitudinal muscle strength, lean mass, and fat mass. Results Lower baseline kPCr was associated with greater decline in all four mobility measures (β, p‐value: (0.036, 0.020) 6‐m usual gait speed; (0.029, 0.038) 2.5‐min usual gait speed; (0.034, 0.011) 6‐m rapid gait speed; (−0.042, <0.001) 400‐m time). In the subset, further adjustment for longitudinal muscle strength, lean mass, and fat mass attenuated longitudinal associations with changes in mobility (Δβ reduced 26–63%). Conclusion Among initially well‐functioning older adults, worse muscle mitochondrial function predicts mobility decline, and part of this longitudinal association is explained by decline in muscle strength and mass. Our findings suggest that worse mitochondrial function contributes to mobility decline with aging. These findings need to be verified in studies correlating longitudinal changes in mitochondrial function, muscle, and mobility performance.
Collapse
Affiliation(s)
- Qu Tian
- Translational Gerontology Branch National Institute on Aging National Institutes of Health Baltimore Maryland USA
| | - Brendan A. Mitchell
- Translational Gerontology Branch National Institute on Aging National Institutes of Health Baltimore Maryland USA
| | - Marta Zampino
- Translational Gerontology Branch National Institute on Aging National Institutes of Health Baltimore Maryland USA
| | - Kenneth W. Fishbein
- Laboratory of Clinical Investigation National Institute on Aging National Institutes of Health Baltimore Maryland USA
| | - Richard G. Spencer
- Laboratory of Clinical Investigation National Institute on Aging National Institutes of Health Baltimore Maryland USA
| | - Luigi Ferrucci
- Translational Gerontology Branch National Institute on Aging National Institutes of Health Baltimore Maryland USA
| |
Collapse
|
35
|
Impact of exercise training on muscle mitochondria modifications in older adults: a systematic review of randomized controlled trials. Aging Clin Exp Res 2022; 34:1495-1510. [PMID: 35079977 DOI: 10.1007/s40520-021-02073-w] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2021] [Accepted: 12/29/2021] [Indexed: 12/12/2022]
Abstract
BACKGROUND Previous evidence showed that cellular aging is a multifactorial process that is associated with decline in mitochondrial function. Physical exercise has been proposed as an effective and safe therapeutical intervention to improve the mitochondria network in the adult myocytes. AIMS The aim of this systematic review of randomized controlled trials (RCTs) was to assess the exercise-induced muscle mitochondria modifications in older adults, underlining the differences related to different exercise modalities. METHODS On November 28th, 2021, five databases (PubMed, Scopus, Web of Science, Cochrane, and PEDro) were systematically searched for RCTs to include articles with: healthy older people as participants; physical exercise (endurance training (ET), resistance training (RT), and combined training (CT)) as intervention; other different exercise modalities or physical inactivity as comparator; mitochondrial modifications (quality, density and dynamics, oxidative, and antioxidant capacity) as outcomes. The quality assessment was performed according to the PEDro scale; the bias risk was evaluated by Cochrane risk-of-bias assessment tool. RESULTS Out of 2940 records, 6 studies were included (2 assessing ET, 2 RT, 1 CT, and 1 both ET and RT). Taken together, 164 elderly subjects were included in the present systematic review. Significant positive effects were reported in terms of mitochondrial quality, density, dynamics, oxidative and antioxidant capacity, even though with different degrees according to the exercise type. The quality assessment reported one good-quality study, whereas the other five studies had a fair quality. DISCUSSION The overall low quality of the studies on this topic indicate that further research is needed. CONCLUSION RT seems to be the most studied physical exercise modality improving mitochondrial density and dynamics, while ET have been related to mitochondrial antioxidant capacity improvements. However, these exercise-induced specific effects should be better explored in older people.
Collapse
|
36
|
DeVito LM, Barzilai N, Cuervo AM, Niedernhofer LJ, Milman S, Levine M, Promislow D, Ferrucci L, Kuchel GA, Mannick J, Justice J, Gonzales MM, Kirkland JL, Cohen P, Campisi J. Extending human healthspan and longevity: a symposium report. Ann N Y Acad Sci 2022; 1507:70-83. [PMID: 34498278 PMCID: PMC10231756 DOI: 10.1111/nyas.14681] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Accepted: 08/09/2021] [Indexed: 12/13/2022]
Abstract
For many years, it was believed that the aging process was inevitable and that age-related diseases could not be prevented or reversed. The geroscience hypothesis, however, posits that aging is, in fact, malleable and, by targeting the hallmarks of biological aging, it is indeed possible to alleviate age-related diseases and dysfunction and extend longevity. This field of geroscience thus aims to prevent the development of multiple disorders with age, thereby extending healthspan, with the reduction of morbidity toward the end of life. Experts in the field have made remarkable advancements in understanding the mechanisms underlying biological aging and identified ways to target aging pathways using both novel agents and repurposed therapies. While geroscience researchers currently face significant barriers in bringing therapies through clinical development, proof-of-concept studies, as well as early-stage clinical trials, are underway to assess the feasibility of drug evaluation and lay a regulatory foundation for future FDA approvals in the future.
Collapse
Affiliation(s)
| | - Nir Barzilai
- Albert Einstein College of Medicine, Bronx, New York
| | | | | | - Sofiya Milman
- Albert Einstein College of Medicine, Bronx, New York
| | | | | | - Luigi Ferrucci
- National Institute on Aging, National Institutes of Health, Bethesda, Maryland
| | - George A Kuchel
- University of Connecticut School of Medicine, Farmington, Connecticut
| | | | - Jamie Justice
- Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Mitzi M Gonzales
- University of Texas Health Sciences Center San Antonio, San Antonio, Texas
| | | | - Pinchas Cohen
- USC Leonard Davis School of Gerontology, Los Angeles, California
| | - Judith Campisi
- The Buck Institute for Research on Aging, Novato, California
- Lawrence Berkeley National Laboratory, Berkley, California
| |
Collapse
|
37
|
Avgerinos KI, Mullins RJ, Egan JM, Kapogiannis D. Ketone Ester Effects on Biomarkers of Brain Metabolism and Cognitive Performance in Cognitively Intact Adults ≥ 55 Years Old. A Study Protocol for a Double-Blinded Randomized Controlled Clinical Trial. J Prev Alzheimers Dis 2022; 9:54-66. [PMID: 35098974 PMCID: PMC9359666 DOI: 10.14283/jpad.2022.3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
BACKGROUND Ketone bodies have been proposed as an "energy rescue" for the Alzheimer's disease (AD) brain, which underutilizes glucose. Prior research has shown that oral ketone monoester (KME) safely induces robust ketosis in humans and has demonstrated cognitive-enhancing and pathology-reducing properties in animal models of AD. However, human evidence that KME may enhance brain ketone metabolism, improve cognitive performance and engage AD pathogenic cascades is scarce. OBJECTIVES To investigate the effects of ketone monoester (KME) on brain metabolism, cognitive performance and AD pathogenic cascades in cognitively normal older adults with metabolic syndrome and therefore at higher risk for AD. DESIGN Double-blinded randomized placebo-controlled clinical trial. SETTING Clinical Unit of the National Institute on Aging, Baltimore, US. PARTICIPANTS Fifty cognitively intact adults ≥ 55 years old, with metabolic syndrome. INTERVENTION Drinks containing 25 g of KME or isocaloric placebo consumed three times daily for 28 days. OUTCOMES Primary: concentration of beta-hydroxybutyrate (BHB) in precuneus measured with Magnetic Resonance Spectroscopy (MRS). Exploratory: plasma and urine BHB, multiple brain and muscle metabolites detected with MRS, cognition assessed with the PACC and NIH toolbox, biomarkers of AD and metabolic mediators in plasma extracellular vesicles, and stool microbiome. DISCUSSION This is the first study to investigate the AD-biomarker and cognitive effects of KME in humans. Ketone monoester is safe, tolerable, induces robust ketosis, and animal studies indicate that it can modify AD pathology. By conducting a study of KME in a population at risk for AD, we hope to bridge the existing gap between pre-clinical evidence and the potential for brain-metabolic, pro-cognitive, and anti-Alzheimer's effects in humans.
Collapse
Affiliation(s)
- K I Avgerinos
- Dimitrios Kapogiannis, Laboratory of Clinical Investigation, National Institute on Aging, National Institutes of Health, 251 Bayview Blvd, 8C228, Baltimore, MD 21224, USA; Email address: ; Telephone: +1 410 350 3953
| | | | | | | |
Collapse
|
38
|
Tian Q, Moore AZ, Oppong R, Ding J, Zampino M, Fishbein KW, Spencer RG, Ferrucci L. Mitochondrial DNA copy number and heteroplasmy load correlate with skeletal muscle oxidative capacity by P31 MR spectroscopy. Aging Cell 2021; 20:e13487. [PMID: 34612579 PMCID: PMC8590093 DOI: 10.1111/acel.13487] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 06/24/2021] [Accepted: 09/12/2021] [Indexed: 12/31/2022] Open
Abstract
The association between blood‐based estimates of mitochondrial DNA parameters, mitochondrial DNA copy number (mtDNA‐CN) and heteroplasmy load, with skeletal muscle bioenergetic capacity was evaluated in 230 participants of the Baltimore Longitudinal Study of Aging (mean age:74.7 years, 53% women). Participants in the study sample had concurrent data on muscle oxidative capacity (τPCr) assessed by 31P magnetic resonance spectroscopy, and mitochondrial DNA parameters estimated from whole‐genome sequencing data. In multivariable linear regression models, adjusted for age, sex, extent of phosphocreatine (PCr) depletion, autosomal sequencing coverage, white blood cell total, and differential count, as well as platelet count, mtDNA‐CN and heteroplasmy load were not significantly associated with τPCr (both p > 0.05). However, in models evaluating whether the association between mtDNA‐CN and τPCr varied by heteroplasmy load, there was a significant interaction between mtDNA‐CN and heteroplasmy load (p = 0.037). In stratified analysis, higher mtDNA‐CN was significantly associated with lower τPCr among participants with high heteroplasmy load (n = 84, β (SE) = −0.236 (0.115), p‐value = 0.044), but not in those with low heteroplasmy load (n = 146, β (SE) = 0.046 (0.119), p‐value = 0.702). Taken together, mtDNA‐CN and heteroplasmy load provide information on muscle bioenergetics. Thus, mitochondrial DNA parameters may be considered proxy measures of mitochondrial function that can be used in large epidemiological studies, especially when comparing subgroups.
Collapse
Affiliation(s)
- Qu Tian
- Translational Gerontology Branch National Institute on Aging Baltimore Maryland USA
| | - Ann Zenobia Moore
- Translational Gerontology Branch National Institute on Aging Baltimore Maryland USA
| | - Richard Oppong
- Translational Gerontology Branch National Institute on Aging Baltimore Maryland USA
| | - Jun Ding
- Translational Gerontology Branch National Institute on Aging Baltimore Maryland USA
| | - Marta Zampino
- Translational Gerontology Branch National Institute on Aging Baltimore Maryland USA
| | - Kenneth W. Fishbein
- Laboratory of Clinical Investigation National Institute on Aging Baltimore Maryland USA
| | - Richard G. Spencer
- Laboratory of Clinical Investigation National Institute on Aging Baltimore Maryland USA
| | - Luigi Ferrucci
- Translational Gerontology Branch National Institute on Aging Baltimore Maryland USA
| |
Collapse
|
39
|
Monaco CMF, Tarnopolsky MA, Dial AG, Nederveen JP, Rebalka IA, Nguyen M, Turner LV, Perry CGR, Ljubicic V, Hawke TJ. Normal to enhanced intrinsic mitochondrial respiration in skeletal muscle of middle- to older-aged women and men with uncomplicated type 1 diabetes. Diabetologia 2021; 64:2517-2533. [PMID: 34392397 DOI: 10.1007/s00125-021-05540-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Accepted: 05/20/2021] [Indexed: 12/26/2022]
Abstract
AIMS/HYPOTHESIS This study interrogated mitochondrial respiratory function and content in skeletal muscle biopsies of healthy adults between 30 and 72 years old with and without uncomplicated type 1 diabetes. METHODS Participants (12 women/nine men) with type 1 diabetes (48 ± 11 years of age), without overt complications, were matched for age, sex, BMI and level of physical activity to participants without diabetes (control participants) (49 ± 12 years of age). Participants underwent a Bergström biopsy of the vastus lateralis to assess mitochondrial respiratory function using high-resolution respirometry and citrate synthase activity. Electron microscopy was used to quantify mitochondrial content and cristae (pixel) density. RESULTS Mean mitochondrial area density was 27% lower (p = 0.006) in participants with type 1 diabetes compared with control participants. This was largely due to smaller mitochondrial fragments in women with type 1 diabetes (-18%, p = 0.057), as opposed to a decrease in the total number of mitochondrial fragments in men with diabetes (-28%, p = 0.130). Mitochondrial respiratory measures, whether estimated per milligram of tissue (i.e. mass-specific) or normalised to area density (i.e. intrinsic mitochondrial function), differed between cohorts, and demonstrated sexual dimorphism. Mass-specific mitochondrial oxidative phosphorylation (OXPHOS) capacity with the substrates for complex I and complex II (CI + II) was significantly lower (-24%, p = 0.033) in women with type 1 diabetes compared with control participants, whereas mass-specific OXPHOS capacities with substrates for complex I only (pyruvate [CI pyr] or glutamate [CI glu]) or complex II only (succinate [CII succ]) were not different (p > 0.404). No statistical differences (p > 0.397) were found in mass-specific OXPHOS capacity in men with type 1 diabetes compared with control participants despite a 42% non-significant increase in CI glu OXPHOS capacity (p = 0.218). In contrast, intrinsic CI + II OXPHOS capacity was not different in women with type 1 diabetes (+5%, p = 0.378), whereas in men with type 1 diabetes it was 25% higher (p = 0.163) compared with control participants. Men with type 1 diabetes also demonstrated higher intrinsic OXPHOS capacity for CI pyr (+50%, p = 0.159), CI glu (+88%, p = 0.033) and CII succ (+28%, p = 0.123), as well as higher intrinsic respiratory rates with low (more physiological) concentrations of either ADP, pyruvate, glutamate or succinate (p < 0.012). Women with type 1 diabetes had higher (p < 0.003) intrinsic respiratory rates with low concentrations of succinate only. Calculated aerobic fitness (Physical Working Capacity Test [PWC130]) showed a strong relationship with mitochondrial respiratory function and content in the type 1 diabetes cohort. CONCLUSIONS/INTERPRETATION In middle- to older-aged adults with uncomplicated type 1 diabetes, we conclude that skeletal muscle mitochondria differentially adapt to type 1 diabetes and demonstrate sexual dimorphism. Importantly, these cellular alterations were significantly associated with our metric of aerobic fitness (PWC130) and preceded notable impairments in skeletal mass and strength.
Collapse
Affiliation(s)
- Cynthia M F Monaco
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON, Canada
| | | | - Athan G Dial
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON, Canada
| | | | - Irena A Rebalka
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON, Canada
| | - Maria Nguyen
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON, Canada
| | - Lauren V Turner
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON, Canada
| | - Christopher G R Perry
- School of Kinesiology and Health Sciences, Muscle Health Research Centre, York University, Toronto, ON, Canada
| | - Vladimir Ljubicic
- Department of Kinesiology, McMaster University, Hamilton, ON, Canada
| | - Thomas J Hawke
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON, Canada.
| |
Collapse
|
40
|
Grevendonk L, Connell NJ, McCrum C, Fealy CE, Bilet L, Bruls YMH, Mevenkamp J, Schrauwen-Hinderling VB, Jörgensen JA, Moonen-Kornips E, Schaart G, Havekes B, de Vogel-van den Bosch J, Bragt MCE, Meijer K, Schrauwen P, Hoeks J. Impact of aging and exercise on skeletal muscle mitochondrial capacity, energy metabolism, and physical function. Nat Commun 2021; 12:4773. [PMID: 34362885 PMCID: PMC8346468 DOI: 10.1038/s41467-021-24956-2] [Citation(s) in RCA: 124] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 07/08/2021] [Indexed: 12/13/2022] Open
Abstract
The relationship between the age-associated decline in mitochondrial function and its effect on skeletal muscle physiology and function remain unclear. In the current study, we examined to what extent physical activity contributes to the decline in mitochondrial function and muscle health during aging and compared mitochondrial function in young and older adults, with similar habitual physical activity levels. We also studied exercise-trained older adults and physically impaired older adults. Aging was associated with a decline in mitochondrial capacity, exercise capacity and efficiency, gait stability, muscle function, and insulin sensitivity, even when maintaining an adequate daily physical activity level. Our data also suggest that a further increase in physical activity level, achieved through regular exercise training, can largely negate the effects of aging. Finally, mitochondrial capacity correlated with exercise efficiency and insulin sensitivity. Together, our data support a link between mitochondrial function and age-associated deterioration of skeletal muscle. Aging is associated with a progressive loss of muscle function. Here the authors characterize mitochondrial capacity and muscle function in young and older adults with similar habitual physical activity and also compared to older adults with exercise training or with physical impairment.
Collapse
Affiliation(s)
- L Grevendonk
- Department of Nutrition and Movement Sciences, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, The Netherlands.,TI Food and Nutrition, Wageningen, The Netherlands
| | - N J Connell
- Department of Nutrition and Movement Sciences, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, The Netherlands.,TI Food and Nutrition, Wageningen, The Netherlands
| | - C McCrum
- Department of Nutrition and Movement Sciences, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, The Netherlands
| | - C E Fealy
- Department of Nutrition and Movement Sciences, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, The Netherlands.,TI Food and Nutrition, Wageningen, The Netherlands
| | - L Bilet
- Department of Nutrition and Movement Sciences, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, The Netherlands.,TI Food and Nutrition, Wageningen, The Netherlands
| | - Y M H Bruls
- Department of Radiology and Nuclear Medicine, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+, Maastricht, The Netherlands
| | - J Mevenkamp
- Department of Radiology and Nuclear Medicine, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+, Maastricht, The Netherlands
| | - V B Schrauwen-Hinderling
- Department of Nutrition and Movement Sciences, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, The Netherlands.,Department of Radiology and Nuclear Medicine, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+, Maastricht, The Netherlands
| | - J A Jörgensen
- Department of Nutrition and Movement Sciences, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, The Netherlands
| | - E Moonen-Kornips
- Department of Nutrition and Movement Sciences, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, The Netherlands
| | - G Schaart
- Department of Nutrition and Movement Sciences, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, The Netherlands
| | - B Havekes
- Department of Nutrition and Movement Sciences, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, The Netherlands.,Department of Internal Medicine, Division of Endocrinology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+, Maastricht, The Netherlands
| | | | - M C E Bragt
- Friesland-Campina, Amersfoort, The Netherlands
| | - K Meijer
- Department of Nutrition and Movement Sciences, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, The Netherlands
| | - P Schrauwen
- Department of Nutrition and Movement Sciences, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, The Netherlands.,TI Food and Nutrition, Wageningen, The Netherlands
| | - J Hoeks
- Department of Nutrition and Movement Sciences, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, The Netherlands. .,TI Food and Nutrition, Wageningen, The Netherlands.
| |
Collapse
|
41
|
Roshanravan B, Liu SZ, Ali AS, Shankland EG, Goss C, Amory JK, Robertson HT, Marcinek DJ, Conley KE. In vivo mitochondrial ATP production is improved in older adult skeletal muscle after a single dose of elamipretide in a randomized trial. PLoS One 2021; 16:e0253849. [PMID: 34264994 PMCID: PMC8282018 DOI: 10.1371/journal.pone.0253849] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Accepted: 03/03/2021] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Loss of mitochondrial function contributes to fatigue, exercise intolerance and muscle weakness, and is a key factor in the disability that develops with age and a wide variety of chronic disorders. Here, we describe the impact of a first-in-class cardiolipin-binding compound that is targeted to mitochondria and improves oxidative phosphorylation capacity (Elamipretide, ELAM) in a randomized, double-blind, placebo-controlled clinical trial. METHODS Non-invasive magnetic resonance and optical spectroscopy provided measures of mitochondrial capacity (ATPmax) with exercise and mitochondrial coupling (ATP supply per O2 uptake; P/O) at rest. The first dorsal interosseous (FDI) muscle was studied in 39 healthy older adult subjects (60 to 85 yrs of age; 46% female) who were enrolled based on the presence of poorly functioning mitochondria. We measured volitional fatigue resistance by force-time integral over repetitive muscle contractions. RESULTS A single ELAM dose elevated mitochondrial energetic capacity in vivo relative to placebo (ΔATPmax; P = 0.055, %ΔATPmax; P = 0.045) immediately after a 2-hour infusion. No difference was found on day 7 after treatment, which is consistent with the half-life of ELAM in human blood. No significant changes were found in resting muscle mitochondrial coupling. Despite the increase in ATPmax there was no significant effect of treatment on fatigue resistance in the FDI. CONCLUSIONS These results highlight that ELAM rapidly and reversibly elevates mitochondrial capacity after a single dose. This response represents the first demonstration of a pharmacological intervention that can reverse mitochondrial dysfunction in vivo immediately after treatment in aging human muscle.
Collapse
Affiliation(s)
- Baback Roshanravan
- Department of Medicine, Division of Nephrology, University of California Davis, Sacramento, California, United States of America
| | - Sophia Z. Liu
- Department of Radiology, University of Washington, Seattle, Washington, United States of America
| | - Amir S. Ali
- Department of Radiology, University of Washington, Seattle, Washington, United States of America
| | - Eric G. Shankland
- Department of Radiology, University of Washington, Seattle, Washington, United States of America
| | - Chessa Goss
- Institute of Translational Health Sciences, University of Washington, Seattle, Washington, United States of America
| | - John K. Amory
- Department of Medicine, University of Washington, Seattle, Washington, United States of America
| | - H. Thomas Robertson
- Department of Medicine, University of Washington, Seattle, Washington, United States of America
| | - David J. Marcinek
- Department of Radiology, University of Washington, Seattle, Washington, United States of America
- Department of Bioengineering, University of Washington, Seattle, Washington, United States of America
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington, United States of America
| | - Kevin E. Conley
- Department of Radiology, University of Washington, Seattle, Washington, United States of America
- Department of Bioengineering, University of Washington, Seattle, Washington, United States of America
- Department of Physiology & Biophysics, University of Washington, Seattle, Washington, United States of America
| |
Collapse
|
42
|
Zampino M, Spencer RG, Fishbein KW, Simonsick EM, Ferrucci L. Cardiovascular Health and Mitochondrial Function: Testing an Association. J Gerontol A Biol Sci Med Sci 2021; 76:361-367. [PMID: 33249505 DOI: 10.1093/gerona/glaa297] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Although mitochondrial dysfunction appears to be a contributing factor in the pathogenesis of cardiovascular and metabolic diseases, empirical data on this association are still lacking. This study evaluated whether mitochondrial oxidative capacity, as assessed by phosphorus magnetic resonance spectroscopy, was associated with cardiovascular risk, as estimated by the Framingham Risk Score (FRS), and with a clinical history of cardiovascular disease (CVD), in community-dwelling adults. METHOD A total of 616 subjects from the Baltimore Longitudinal Study of Aging (mean age 66 years) underwent a comprehensive clinical evaluation. Mitochondrial oxidative capacity in skeletal muscle was assessed as post-exercise phosphocreatine recovery time constant by phosphorus magnetic resonance spectroscopy. Multivariate regression models were employed to determine the cross-sectional association of mitochondrial oxidative capacity with FRS and history of CVD. RESULTS Decreased mitochondrial oxidative capacity was strongly associated with higher FRS independent of age, body composition, and physical activity. Lower oxidative capacity was also associated with a history of positive of CVD and higher number of CVD events. CONCLUSIONS We speculate that the observed association could reflect the effect of an excessive production of oxidative species by dysfunctional mitochondria. Furthermore, decreased energy production could hamper the functionality of heart and vessels. In turn, a malfunctioning cardiovascular apparatus could fail to deliver the oxygen necessary for optimal mitochondrial energy production, therefore creating a vicious cycle. Longitudinal studies are necessary to ascertain the directionality of the association and the eventual presence of common pathogenetic roots. In conclusion, mitochondria could represent an important target for intervention in cardiovascular health.
Collapse
Affiliation(s)
- Marta Zampino
- National Institute on Aging (NIA), National Institutes of Health (NIH), Baltimore, Maryland
| | - Richard G Spencer
- National Institute on Aging (NIA), National Institutes of Health (NIH), Baltimore, Maryland
| | - Kenneth W Fishbein
- National Institute on Aging (NIA), National Institutes of Health (NIH), Baltimore, Maryland
| | - Eleanor M Simonsick
- National Institute on Aging (NIA), National Institutes of Health (NIH), Baltimore, Maryland
| | - Luigi Ferrucci
- National Institute on Aging (NIA), National Institutes of Health (NIH), Baltimore, Maryland
| |
Collapse
|
43
|
Chungath RR, Witham MD, Clarke CL, Hutcheon A, Gandy S, Gingles C, Priba L, Nicholas SR, Cavin I, Sumukadas D, Struthers AD, George J. Association between mitochondrial function measured by 31P magnetic resonance spectroscopy and physical performance in older people with functional impairment. JCSM CLINICAL REPORTS 2021. [DOI: 10.1002/crt2.33] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Affiliation(s)
- Rebecca R. Chungath
- AGE Research Group, NIHR Newcastle Biomedical Research Centre Newcastle University and Newcastle upon Tyne Hospitals NHS Foundation Trust Newcastle upon Tyne UK
| | - Miles D. Witham
- AGE Research Group, NIHR Newcastle Biomedical Research Centre Newcastle University and Newcastle upon Tyne Hospitals NHS Foundation Trust Newcastle upon Tyne UK
- Division of Molecular and Clinical Medicine University of Dundee, Ninewells Hospital and Medical School Dundee UK
| | - Clare L. Clarke
- Division of Molecular and Clinical Medicine University of Dundee, Ninewells Hospital and Medical School Dundee UK
| | - Anita Hutcheon
- Division of Molecular and Clinical Medicine University of Dundee, Ninewells Hospital and Medical School Dundee UK
| | - Stephen Gandy
- Department of Medical Physics Ninewells Hospital, NHS Tayside Dundee UK
| | - Christopher Gingles
- Division of Molecular and Clinical Medicine University of Dundee, Ninewells Hospital and Medical School Dundee UK
| | - Lukasz Priba
- Department of Medical Physics Ninewells Hospital, NHS Tayside Dundee UK
| | | | - Ian Cavin
- Department of Medical Physics NHS Lothian Edinburgh UK
| | - Deepa Sumukadas
- Department of Medicine for the Elderly Ninewells Hospital, NHS Tayside Dundee UK
| | - Allan D. Struthers
- Division of Molecular and Clinical Medicine University of Dundee, Ninewells Hospital and Medical School Dundee UK
| | - Jacob George
- Division of Molecular and Clinical Medicine University of Dundee, Ninewells Hospital and Medical School Dundee UK
| |
Collapse
|
44
|
Bittel DC, Bittel AJ, Varadhachary AS, Pietka T, Sinacore DR. Deficits in the Skeletal Muscle Transcriptome and Mitochondrial Coupling in Progressive Diabetes-Induced CKD Relate to Functional Decline. Diabetes 2021; 70:1130-1144. [PMID: 33526590 PMCID: PMC8173802 DOI: 10.2337/db20-0688] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Accepted: 01/25/2021] [Indexed: 12/14/2022]
Abstract
Two-thirds of people with type 2 diabetes mellitus (T2DM) have or will develop chronic kidney disease (CKD), which is characterized by rapid renal decline that, together with superimposed T2DM-related metabolic sequelae, synergistically promotes early frailty and mobility deficits that increase the risk of mortality. Distinguishing the mechanisms linking renal decline to mobility deficits in CKD progression and/or increasing severity in T2DM is instrumental both in identifying those at high risk for functional decline and in formulating effective treatment strategies to prevent renal failure. While evidence suggests that skeletal muscle energetics may relate to the development of these comorbidities in advanced CKD, this has never been assessed across the spectrum of CKD progression, especially in T2DM-induced CKD. Here, using next-generation sequencing, we first report significant downregulation in transcriptional networks governing oxidative phosphorylation, coupled electron transport, electron transport chain (ETC) complex assembly, and mitochondrial organization in both middle- and late-stage CKD in T2DM. Furthermore, muscle mitochondrial coupling is impaired as early as stage 3 CKD, with additional deficits in ETC respiration, enzymatic activity, and increased redox leak. Moreover, mitochondrial ETC function and coupling strongly relate to muscle performance and physical function. Our results indicate that T2DM-induced CKD progression impairs physical function, with implications for altered metabolic transcriptional networks and mitochondrial functional deficits as primary mechanistic factors early in CKD progression in T2DM.
Collapse
Affiliation(s)
- Daniel C Bittel
- Program in Physical Therapy, Washington University School of Medicine, St. Louis, MO
| | - Adam J Bittel
- Program in Physical Therapy, Washington University School of Medicine, St. Louis, MO
| | - Arun S Varadhachary
- Department of Neurology, Washington University School of Medicine, St. Louis, MO
| | - Terri Pietka
- Center for Human Nutrition, Washington University School of Medicine, St. Louis, MO
| | - David R Sinacore
- Program in Physical Therapy, Washington University School of Medicine, St. Louis, MO
- Department of Physical Therapy, Congdon School of Health Sciences, High Point University, High Point, NC
| |
Collapse
|
45
|
Liu F, Wanigatunga AA, Zampino M, Knuth ND, Simonsick EM, Schrack JA, Ferrucci L. Association of Mitochondrial Function, Substrate Utilization, and Anaerobic Metabolism With Age-Related Perceived Fatigability. J Gerontol A Biol Sci Med Sci 2021; 76:426-433. [PMID: 32803242 DOI: 10.1093/gerona/glaa201] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Indexed: 11/13/2022] Open
Abstract
Previous work has shown that poorer mitochondrial function is associated with age-related perceived fatigability. However, whether glucose oxidation and anaerobic metabolism are intermediate factors underlying this association remains unclear. We examined the total cross-sectional association between mitochondrial function and perceived fatigability in 554 adults aged 22-99 years. Mitochondrial function was assessed by skeletal muscle oxidative capacity (kPCr) using 31P magnetic resonance spectroscopy. Perceived fatigability was measured by rating of perceived exertion after a 5-minute (0.67 m/s) treadmill walk. The intermediate role of glucose oxidation (measured by the rate of change of respiratory exchange ratio [RER change rate] during the 5-minute treadmill walk) and anaerobic metabolism (measured by ventilatory threshold [VeT] during a maximal treadmill test) was evaluated by examining their cross-sectional associations with kPCr and perceived exertion. For each 0.01/s lower kPCr, perceived fatigability was 0.47 points higher (p = .002). A 0.01/s lower kPCr was also associated with 8.3 L/min lower VeT (p < .001). Lower VeT was associated with higher fatigability at lower levels of kPCr but not at higher kPCr levels (β for interaction = 0.017, p = .002). kPCr and RER change rate were not significantly associated (p = .341), but a 0.01/min higher RER change rate was associated with 0.12-point higher fatigability (p = .001). Poorer mitochondrial function potentially contributes to higher perceived fatigability through higher glucose oxidation and higher anaerobic metabolism. Future studies to further explore the longitudinal mechanisms between these metabolic changes and fatigability are warranted.
Collapse
Affiliation(s)
- Fangyu Liu
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland.,Center on Aging and Health, Johns Hopkins University, Baltimore, Maryland
| | - Amal A Wanigatunga
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland.,Center on Aging and Health, Johns Hopkins University, Baltimore, Maryland
| | - Marta Zampino
- National Institute on Aging, National Institutes of Health, Baltimore, Maryland
| | | | - Eleanor M Simonsick
- National Institute on Aging, National Institutes of Health, Baltimore, Maryland
| | - Jennifer A Schrack
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland.,Center on Aging and Health, Johns Hopkins University, Baltimore, Maryland
| | - Luigi Ferrucci
- National Institute on Aging, National Institutes of Health, Baltimore, Maryland
| |
Collapse
|
46
|
Berg OK, Kwon OS, Hureau TJ, Clifton HL, Thurston TS, Le Fur Y, Jeong EK, Trinity JD, Richardson RS, Wang E, Layec G. Skeletal Muscle Mitochondrial Adaptations to Maximal Strength Training in Older Adults. J Gerontol A Biol Sci Med Sci 2021; 75:2269-2277. [PMID: 32253421 DOI: 10.1093/gerona/glaa082] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Indexed: 01/06/2023] Open
Abstract
Maximal strength training (MST) results in robust improvements in skeletal muscle force production, efficiency, and mass. However, the effects of MST on muscle mitochondria are still unknown. Accordingly, the purpose of this study was to examine, from the molecular level to whole-muscle, mitochondrial adaptations induced by 8 weeks of knee-extension MST in the quadriceps of 10 older adults using immunoblotting, spectrophotometry, high-resolution respirometry in permeabilized muscle fibers, in vivo 31P magnetic resonance spectroscopy (31P-MRS), and gas exchange. As anticipated, MST resulted in an increased isometric knee-extensor force from 133 ± 36 to 147 ± 49 Nm (p < .05) and quadriceps muscle volume from 1,410 ± 103 to 1,555 ± 455 cm3 (p < .05). Mitochondrial complex (I-V) protein abundance and citrate synthase activity were not significantly altered by MST. Assessed ex vivo, maximal ADP-stimulated respiration (state 3CI+CII, PRE: 23 ± 6 and POST: 14 ± 5 ρM·mg-1·s-1, p < .05), was decreased by MST, predominantly, as a result of a decline in complex I-linked respiration (p < .05). Additionally, state 3 free-fatty acid linked respiration was decreased following MST (PRE: 19 ± 5 and POST: 14 ± 3 ρM·mg-1·s-1, p < .05). Assessed in vivo, MST slowed the PCr recovery time constant (PRE: 49 ± 13 and POST: 57 ± 16 seconds, p < .05) and lowered, by ~20% (p = .055), the quadriceps peak rate of oxidative ATP synthesis, but did not significantly alter the oxidation of lipid. Although these, likely qualitative, mitochondrial adaptations are potentially negative in terms of skeletal muscle energetic capacity, they need to be considered in light of the many improvements in muscle function that MST affords older adults.
Collapse
Affiliation(s)
- Ole Kristian Berg
- Faculty of Health and Social Sciences, Molde University College, Norway
| | - Oh Sung Kwon
- Department of Kinesiology, University of Connecticut, Storrs
| | - Thomas J Hureau
- Department of Internal Medicine, Division of Geriatrics, University of Utah, Salt Lake City.,Geriatric Research, Education, and Clinical Center, George E. Whalen VA Medical Center, Salt Lake City, Utah
| | - Heather L Clifton
- Department of Internal Medicine, Division of Geriatrics, University of Utah, Salt Lake City.,Geriatric Research, Education, and Clinical Center, George E. Whalen VA Medical Center, Salt Lake City, Utah
| | - Taylor S Thurston
- Department of Internal Medicine, Division of Geriatrics, University of Utah, Salt Lake City.,Geriatric Research, Education, and Clinical Center, George E. Whalen VA Medical Center, Salt Lake City, Utah.,Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City
| | - Yann Le Fur
- CRMBM, Aix-Marseille Universite, CNRS 7339, France
| | - Eun-Kee Jeong
- Department of Radiology, Utah Center for Advanced Imaging Research, University of Utah, Salt Lake City
| | - Joel D Trinity
- Department of Internal Medicine, Division of Geriatrics, University of Utah, Salt Lake City.,Geriatric Research, Education, and Clinical Center, George E. Whalen VA Medical Center, Salt Lake City, Utah.,Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City
| | - Russell S Richardson
- Department of Internal Medicine, Division of Geriatrics, University of Utah, Salt Lake City.,Geriatric Research, Education, and Clinical Center, George E. Whalen VA Medical Center, Salt Lake City, Utah.,Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City
| | - Eivind Wang
- Faculty of Health and Social Sciences, Molde University College, Norway.,Department of Internal Medicine, Division of Geriatrics, University of Utah, Salt Lake City.,Department of Medicine, Norwegian University of Science and Technology, Trondheim, Norway
| | - Gwenael Layec
- Department of Internal Medicine, Division of Geriatrics, University of Utah, Salt Lake City.,Geriatric Research, Education, and Clinical Center, George E. Whalen VA Medical Center, Salt Lake City, Utah.,Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City.,Department of Kinesiology, University of Massachusetts, Amherst.,Institute for Applied Life Sciences, University of Massachusetts, Amherst
| |
Collapse
|
47
|
Zampino M, Semba RD, Adelnia F, Spencer RG, Fishbein KW, Schrack JA, Simonsick EM, Ferrucci L. Greater Skeletal Muscle Oxidative Capacity Is Associated With Higher Resting Metabolic Rate: Results From the Baltimore Longitudinal Study of Aging. J Gerontol A Biol Sci Med Sci 2021; 75:2262-2268. [PMID: 32201887 DOI: 10.1093/gerona/glaa071] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Indexed: 12/20/2022] Open
Abstract
Resting metabolic rate (RMR) tends to decline with aging. The age-trajectory of decline in RMR is similar to changes that occur in muscle mass, muscle strength, and fitness, but while the decline in these phenotypes has been related to changes of mitochondrial function and oxidative capacity, whether lower RMR is associated with poorer mitochondrial oxidative capacity is unknown. In 619 participants of the Baltimore Longitudinal Study of Aging, we analyzed the cross-sectional association between RMR (kcal/day), assessed by indirect calorimetry, and skeletal muscle maximal oxidative phosphorylation capacity, assessed as postexercise phosphocreatine recovery time constant (τ PCr), by phosphorous magnetic resonance spectroscopy. Linear regression models were used to evaluate the relationship between τ PCr and RMR, adjusting for potential confounders. Independent of age, sex, lean body mass, muscle density, and fat mass, higher RMR was significantly associated with shorter τ PCr, indicating greater mitochondrial oxidative capacity. Higher RMR is associated with a higher mitochondrial oxidative capacity in skeletal muscle. This association may reflect a relationship between better muscle quality and greater mitochondrial health.
Collapse
Affiliation(s)
- Marta Zampino
- National Institute on Aging, National Institutes of Health, Baltimore, Maryland
| | - Richard D Semba
- Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Fatemeh Adelnia
- Institute of Imaging Science, Vanderbilt University, Nashville, Tennessee
| | - Richard G Spencer
- National Institute on Aging, National Institutes of Health, Baltimore, Maryland
| | - Kenneth W Fishbein
- National Institute on Aging, National Institutes of Health, Baltimore, Maryland
| | - Jennifer A Schrack
- Center on Aging and Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| | - Eleanor M Simonsick
- National Institute on Aging, National Institutes of Health, Baltimore, Maryland
| | - Luigi Ferrucci
- National Institute on Aging, National Institutes of Health, Baltimore, Maryland
| |
Collapse
|
48
|
Malis V, Sinha U, Sinha S. 3D Muscle Deformation Mapping at Submaximal Isometric Contractions: Applications to Aging Muscle. Front Physiol 2020; 11:600590. [PMID: 33343396 PMCID: PMC7744822 DOI: 10.3389/fphys.2020.600590] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2020] [Accepted: 11/09/2020] [Indexed: 11/24/2022] Open
Abstract
3D strain or strain rate tensor mapping comprehensively captures regional muscle deformation. While compressive strain along the muscle fiber is a potential measure of the force generated, radial strains in the fiber cross-section may provide information on the material properties of the extracellular matrix. Additionally, shear strain may potentially inform on the shearing of the extracellular matrix; the latter has been hypothesized as the mechanism of lateral transmission of force. Here, we implement a novel fast MR method for velocity mapping to acquire multi-slice images at different % maximum voluntary contraction (MVC) for 3D strain mapping to explore deformation in the plantar-flexors under isometric contraction in a cohort of young and senior subjects. 3D strain rate and strain tensors were computed and eigenvalues and two invariants (maximum shear and volumetric strain) were extracted. Strain and strain rate indices (contractile and in-plane strain/strain rate, shear strain/strain rate) changed significantly with %MVC (30 and 60% MVC) and contractile and shear strain with age in the medial gastrocnemius. In the soleus, significant differences with age in contractile and shear strain were seen. Univariate regression revealed weak but significant correlation of in-plane and shear strain and shear strain rate indices to %MVC and correlation of contractile and shear strain indices to force. The ability to map strain tensor components provides unique insights into muscle physiology: with contractile strain providing an index of the force generated by the muscle fibers while the shear strain could potentially be a marker of lateral transmission of force.
Collapse
Affiliation(s)
- Vadim Malis
- Department of Physics, University of California, San Diego, San Diego, CA, United States.,Department of Radiology, University of California, San Diego, San Diego, CA, United States
| | - Usha Sinha
- Department of Physics, San Diego State University, San Diego, CA, United States
| | - Shantanu Sinha
- Department of Radiology, University of California, San Diego, San Diego, CA, United States
| |
Collapse
|
49
|
A Plasma Proteomic Signature of Skeletal Muscle Mitochondrial Function. Int J Mol Sci 2020; 21:ijms21249540. [PMID: 33333910 PMCID: PMC7765442 DOI: 10.3390/ijms21249540] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Revised: 12/09/2020] [Accepted: 12/10/2020] [Indexed: 12/29/2022] Open
Abstract
Although mitochondrial dysfunction has been implicated in aging, physical function decline, and several age-related diseases, an accessible and affordable measure of mitochondrial health is still lacking. In this study we identified the proteomic signature of muscular mitochondrial oxidative capacity in plasma. In 165 adults, we analyzed the association between concentrations of plasma proteins, measured using the SOMAscan assay, and skeletal muscle maximal oxidative phosphorylation capacity assessed as post-exercise phosphocreatine recovery time constant (τPCr) by phosphorous magnetic resonance spectroscopy. Out of 1301 proteins analyzed, we identified 87 proteins significantly associated with τPCr, adjusting for age, sex, and phosphocreatine depletion. Sixty proteins were positively correlated with better oxidative capacity, while 27 proteins were correlated with poorer capacity. Specific clusters of plasma proteins were enriched in the following pathways: homeostasis of energy metabolism, proteostasis, response to oxidative stress, and inflammation. The generalizability of these findings would benefit from replication in an independent cohort and in longitudinal analyses.
Collapse
|
50
|
Ng TKS, Kovalik JP, Ching J, Chan AW, Matchar DB. Novel metabolomics markers are associated with pre-clinical decline in hand grip strength in community-dwelling older adults. Mech Ageing Dev 2020; 193:111405. [PMID: 33217429 DOI: 10.1016/j.mad.2020.111405] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 10/26/2020] [Accepted: 11/11/2020] [Indexed: 12/30/2022]
Abstract
BACKGROUND Hand grip strength (HGS) has been proposed as a robust predictor for frailty and sarcopenia. Hence, identifying biomarkers for declining HGS accompanying aging could deepen our understanding of the biological underpinnings, informing pre-emptive intervention. Acylcarnitines (ACs) are metabolites generated by fatty acid metabolism in the mitochondria and are dysregulated in multiple disorders affecting the musculature. However, they have not been comprehensively profiled and examined regarding their utility in predicting variability in declining HGS, longitudinally. Thus, we aimed to: 1) validate previous findings on insignificant cross-sectional association between ACs and HGS, and 2) examine whether baseline ACs were associated with both decline and variability in HGS over 18 months, in community-dwelling older adults. METHODS We included participants who had HGS measured with dynamometer longitudinally (N = 121). We quantified ACs by targeted plasma metabolomics profiling. Multivariable linear regressions were then performed. RESULTS Cross-sectionally, ACs were not significantly associated with HGS. Longitudinally, baseline short-chain dicarboxylic and hydroxylated acylcarnitines (AC-DC/-OH) levels were inversely associated with and significantly explained the variability in 18-month decline in HGS. A specific AC species, the C4-OH, accounted for most of the variance explained. CONCLUSIONS We showed novel biomarkers for declining HGS, furthering molecular understanding and informing nutritional pre-emptive programs.
Collapse
Affiliation(s)
- Ted Kheng Siang Ng
- Duke-National University of Singapore Medical School, Center for Aging, Research and Education Singapore.
| | - Jean-Paul Kovalik
- Duke-National University of Singapore Medical School, Cardiovascular and Metabolic Disorders Programme, Singapore
| | - Jianhong Ching
- Duke-National University of Singapore Medical School, Cardiovascular and Metabolic Disorders Programme, Singapore
| | - Angelique W Chan
- Duke-National University of Singapore Medical School, Center for Aging, Research and Education Singapore; Duke-National University of Singapore Medical School, Program in Health Services and Systems Research, Singapore; National University of Singapore, Department of Sociology, Faculty of Arts and Social Sciences, Singapore
| | - David Bruce Matchar
- Duke-National University of Singapore Medical School, Center for Aging, Research and Education Singapore; Duke-National University of Singapore Medical School, Program in Health Services and Systems Research, Singapore; Duke University School of Medicine, Department of Medicine (General Internal Medicine), United States.
| |
Collapse
|