1
|
Aslan DH, Sayre MK, Bharadwaj PK, Ally M, Maltagliati S, Lai MHC, Wilcox RR, Klimentidis YC, Alexander GE, Raichlen DA. Associations Between Walking Pace, APOE-ε4 Genotype, and Brain Health in Middle-Aged to Older Adults. Med Sci Sports Exerc 2025; 57:1212-1220. [PMID: 39780372 DOI: 10.1249/mss.0000000000003646] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2025]
Abstract
PURPOSE This study aimed to investigate whether self-reported walking pace (a marker of physical function) and the presence of APOE-ε4 allele interact to modify brain health outcomes. METHODS We used data from a prospective cohort study of middle-aged to older adults from the UK Biobank who self-reported walking pace (slow or steady-to-brisk) and who were initially free of dementia ( n = 415,110). Incident all-cause dementia was obtained from hospital and death registry records, and structural brain volumes (right and left hippocampus volumes, total gray matter volume, and volume of white matter hyperintensities) were measured from a subset of participants ( n = 33,113). Cox proportional hazard models and generalized linear models were used to assess associations between exposures and outcomes. RESULTS Slow walking pace and the presence of APOE-ε4 allele were associated with increased dementia risk (HR = 1.79 [95% CI = 1.66-1.93], P < 0.001; HR = 3.06 [2.90-3.23], P < 0.001, respectively), and there was an interaction between these associations, indicating that the association of walking pace with dementia risk is modified by APOE-ε4 status (reference group: HR Steady-Brisk/APOE-ε4- = 1; HR Slow/APOE-ε4- = 2.03 [1.84-2.25], P < 0.001; HR Steady-Brisk/APOE-ε4+ = 3.21 [3.02-3.41], P < 0.001; HR Slow/APOE-ε4+ = 4.99 [4.48-5.58], P < 0.001). Slow self-reported walking pace was associated with worse brain volume outcomes, and these associations were not modified by APOE-ε4 genotype. CONCLUSIONS These results suggest walking pace and APOE-ε4 status independently influence brain volume outcomes, but both factors independently and jointly contribute to increased dementia risk. Individuals with both risk factors (slow walking pace and APOE-ε4 allele) show the strongest associations with dementia risk.
Collapse
Affiliation(s)
- Daniel H Aslan
- Human and Evolutionary Biology Section, Department of Biological Sciences, University of Southern California, CA
| | - M Katherine Sayre
- Department of Anthropology, University of California, Santa Barbara, CA
| | | | - Madeline Ally
- Department of Psychology, University of Arizona, Tucson, AZ
| | - Silvio Maltagliati
- Human and Evolutionary Biology Section, Department of Biological Sciences, University of Southern California, CA
| | - Mark H C Lai
- Department of Psychology, University of Southern California, CA
| | - Rand R Wilcox
- Department of Psychology, University of Southern California, CA
| | | | | | | |
Collapse
|
2
|
Castro Sepulveda M, Lagarrigue S, Amati F. The association of mitochondrial morphology and supercomplex redistribution with skeletal muscle oxidative capacity in older adults. Physiol Rep 2025; 13:e70359. [PMID: 40343403 PMCID: PMC12059264 DOI: 10.14814/phy2.70359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Revised: 04/24/2025] [Accepted: 04/24/2025] [Indexed: 05/11/2025] Open
Abstract
Skeletal muscle maximal oxidative capacity (ATPmax) is a key component of age-related sarcopenia and muscle health. The contribution of mitochondrial morphology and electron transport chain supercomplex (SC) assemblies to ATPmax has yet to be determined in human muscle. ATPmax measured in vivo by 31phosphorus magnetic resonance spectroscopy in the quadriceps femoris of nine volunteers (65.5 ± 3.3 years old) was correlated with muscle biopsy outcomes before and after 4 months of supervised exercise. Mitochondrial morphology was assessed in electron micrographs, and SCs were measured by blue native gel electrophoresis. In the sedentary conditions, ATPmax was positively associated with complex (C) I and CIII in SC I+III2+IVn and negatively associated with CI and CIII in SC I+III2. Regarding mitochondrial morphology, ATPmax was positively associated with markers of mitochondrial elongation. Exercise training-induced increases in ATPmax were accompanied by mitochondrial elongation and by the redistribution of free complex III. Indicators of mitochondrial elongation were associated with the redistribution of specific complexes to SC I+III2+IVn. Higher skeletal muscle oxidative capacity in older adults is associated with mitochondrial elongation and the redistribution of electron transport chain complexes into higher rank SCs in the same muscle. Further, we provide evidence that mitochondrial elongation favors mitochondrial SC assembly.
Collapse
Affiliation(s)
- Mauricio Castro Sepulveda
- Aging and Muscle Metabolism Laboratory, Department of Biomedical SciencesUniversity of LausanneLausanneSwitzerland
| | - Sylviane Lagarrigue
- Aging and Muscle Metabolism Laboratory, Department of Biomedical SciencesUniversity of LausanneLausanneSwitzerland
| | - Francesca Amati
- Aging and Muscle Metabolism Laboratory, Department of Biomedical SciencesUniversity of LausanneLausanneSwitzerland
- Service of Endocrinology, Diabetology, and Metabolism, Department of MedicineLausanne University HospitalLausanneSwitzerland
| |
Collapse
|
3
|
Gay EL, Coen PM, Harrison S, Garcia RE, Qiao YS, Goodpaster BH, Forman DE, Toledo FGS, Distefano G, Kramer PA, Ramos SV, Molina AJA, Nicklas BJ, Cummings SR, Cawthon PM, Hepple RT, Newman AB, Glynn NW. Sex differences in the association between skeletal muscle energetics and perceived physical fatigability: the Study of Muscle, Mobility and Aging (SOMMA). GeroScience 2025; 47:1999-2013. [PMID: 39436549 PMCID: PMC11979008 DOI: 10.1007/s11357-024-01373-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Accepted: 09/27/2024] [Indexed: 10/23/2024] Open
Abstract
Greater perceived physical fatigability and lower skeletal muscle energetics are both predictors of mobility decline. Characterizing associations between muscle energetics and perceived fatigability may provide insight into potential targets to prevent mobility decline. We examined associations of in vivo (maximal ATP production, ATPmax) and ex vivo (maximal carbohydrate supported oxidative phosphorylation [max OXPHOS] and maximal fatty acid supported OXPHOS [max FAO OXPHOS]) measures of mitochondrial energetics with two measures of perceived physical fatigability, Pittsburgh Fatigability Scale (PFS, 0-50, higher = greater) and Rating of Perceived Exertion (RPE Fatigability, 6-20, higher = greater) after a slow treadmill walk. Participants from the Study of Muscle, Mobility and Aging (N = 873) were 76.3 ± 5.0 years old, 59.2% women, and 85.3% White. Higher muscle energetics (both in vivo and ex vivo) were associated with lower perceived physical fatigability, all p < 0.03. When stratified by sex, higher ATPmax was associated with lower PFS Physical for men only; higher max OXPHOS and max FAO OXPHOS were associated with lower RPE Fatigability for both sexes. Higher skeletal muscle energetics were associated with 40-55% lower odds of being in the most (PFS ≥ 25, RPE Fatigability ≥ 12) vs least (PFS 0-4, RPE Fatigability 6-7) severe fatigability strata, all p < 0.03. Being a woman was associated with 2-3 times higher odds of being in the most severe fatigability strata when controlling for ATPmax but not the ex vivo measures (p < 0.05). Better mitochondrial energetics were linked to lower fatigability and less severe fatigability in older adults. Findings imply that improving skeletal muscle energetics may mitigate perceived physical fatigability and prolong healthy aging.
Collapse
Affiliation(s)
- Emma L Gay
- Department of Epidemiology, School of Public Health, University of Pittsburgh, 130 De Soto Street, Pittsburgh, PA, 15261, USA
| | - Paul M Coen
- Translational Research Institute, AdventHealth, Orlando, FL, 32804, USA
| | - Stephanie Harrison
- San Francisco Coordinating Center, California Pacific Medical Center Research Institute, San Francisco, CA, 94143, USA
| | - Reagan E Garcia
- Department of Epidemiology, School of Public Health, University of Pittsburgh, 130 De Soto Street, Pittsburgh, PA, 15261, USA
| | - Yujia Susanna Qiao
- San Francisco Coordinating Center, California Pacific Medical Center Research Institute, San Francisco, CA, 94143, USA
| | - Bret H Goodpaster
- Translational Research Institute, AdventHealth, Orlando, FL, 32804, USA
| | - Daniel E Forman
- Department of Medicine (Geriatrics and Cardiology), University of Pittsburgh, and Geriatrics, Research, Education, and Clinical Center (GRECC), VA Pittsburgh Healthcare System, Pittsburgh, PA, 15261, USA
| | - Frederico G S Toledo
- Division of Endocrinology and Metabolism, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| | | | - Philip A Kramer
- Department of Internal Medicine-Gerontology and Geriatric Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, 27109, USA
| | - Sofhia V Ramos
- Translational Research Institute, AdventHealth, Orlando, FL, 32804, USA
| | - Anthony J A Molina
- University of California San Diego, School of Medicine, Division of Geriatrics, Gerontology, and Palliative Care, La Jolla, CA, 92093, USA
| | - Barbara J Nicklas
- Department of Internal Medicine-Gerontology and Geriatric Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, 27109, USA
| | - Steven R Cummings
- San Francisco Coordinating Center, California Pacific Medical Center Research Institute, San Francisco, CA, 94143, USA
| | - Peggy M Cawthon
- San Francisco Coordinating Center, California Pacific Medical Center Research Institute, San Francisco, CA, 94143, USA
- Department of Epidemiology and Biostatistics, University of California, San Francisco, California, USA
| | - Russell T Hepple
- Department of Physical Therapy, University of Florida, Gainesville, FL, 32610, USA
| | - Anne B Newman
- Department of Epidemiology, School of Public Health, University of Pittsburgh, 130 De Soto Street, Pittsburgh, PA, 15261, USA
| | - Nancy W Glynn
- Department of Epidemiology, School of Public Health, University of Pittsburgh, 130 De Soto Street, Pittsburgh, PA, 15261, USA.
| |
Collapse
|
4
|
Cefis M, Marcangeli V, Hammad R, Granet J, Leduc-Gaudet JP, Gaudreau P, Trumpff C, Huang Q, Picard M, Aubertin-Leheudre M, Bélanger M, Robitaille R, Morais JA, Gouspillou G. Impact of physical activity on physical function, mitochondrial energetics, ROS production, and Ca 2+ handling across the adult lifespan in men. Cell Rep Med 2025; 6:101968. [PMID: 39933528 PMCID: PMC11866497 DOI: 10.1016/j.xcrm.2025.101968] [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: 07/16/2024] [Revised: 11/05/2024] [Accepted: 01/20/2025] [Indexed: 02/13/2025]
Abstract
Aging-related muscle atrophy and weakness contribute to loss of mobility, falls, and disability. Mitochondrial dysfunction is widely considered a key contributing mechanism to muscle aging. However, mounting evidence positions physical activity as a confounding factor, making unclear whether muscle mitochondria accumulate bona fide defects with aging. To disentangle aging from physical activity-related mitochondrial adaptations, we functionally profiled skeletal muscle mitochondria in 51 inactive and 88 active men aged 20-93. Physical activity status confers partial protection against age-related decline in physical performance. Mitochondrial respiration remains unaltered in active participants, indicating that aging per se does not alter mitochondrial respiratory capacity. Mitochondrial reactive oxygen species (ROS) production is unaffected by aging and higher in active participants. In contrast, mitochondrial calcium retention capacity decreases with aging regardless of physical activity and correlates with muscle mass, performance, and the stress-responsive metabokine/mitokine growth differentiation factor 15 (GDF15). Targeting mitochondrial calcium handling may hold promise for treating aging-related muscle impairments.
Collapse
Affiliation(s)
- Marina Cefis
- Département des sciences de l'activité physique, Université du Québec À Montréal, Montreal, QC, Canada; Groupe de recherche en Activité Physique Adaptée, Montréal, QC, Canada; INSERM UMR1093-CAPS, UFR des Sciences de santé, Université de Bourgogne, Dijon, France
| | - Vincent Marcangeli
- Département des sciences de l'activité physique, Université du Québec À Montréal, Montreal, QC, Canada; Groupe de recherche en Activité Physique Adaptée, Montréal, QC, Canada; Département des sciences biologiques, Université du Québec À Montréal, Montreal, QC, Canada
| | - Rami Hammad
- Département des sciences de l'activité physique, Université du Québec À Montréal, Montreal, QC, Canada; Groupe de recherche en Activité Physique Adaptée, Montréal, QC, Canada; Département des sciences biologiques, Université du Québec À Montréal, Montreal, QC, Canada; Centre de Recherche de l'Institut Universitaire de Gériatrie de Montréal, Montreal, QC, Canada; Al-Ahliyya Amman university, Faculty of educational sciences, Department of physical and health education, Amman, Jordan
| | - Jordan Granet
- Département des sciences biologiques, Université du Québec À Montréal, Montreal, QC, Canada; Centre de Recherche de l'Institut Universitaire de Gériatrie de Montréal, Montreal, QC, Canada
| | - Jean-Philippe Leduc-Gaudet
- Département des sciences de l'activité physique, Université du Québec À Montréal, Montreal, QC, Canada; Research Group in Cellular Signaling, Department of Medical Biology, Université du Québec À Trois-Rivières, Trois-Rivières, Canada
| | - Pierrette Gaudreau
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal, Département de médecine, Université de Montréal, Montreal, QC, Canada
| | - Caroline Trumpff
- Division of Behavioral Medicine, Department of Psychiatry, Columbia University Irving Medical Center, and Robert N Butler Columbia Aging Center, Mailman School of Public Health, Columbia University, New York, NY, USA
| | - Qiuhan Huang
- Division of Behavioral Medicine, Department of Psychiatry, Columbia University Irving Medical Center, and Robert N Butler Columbia Aging Center, Mailman School of Public Health, Columbia University, New York, NY, USA
| | - Martin Picard
- Division of Behavioral Medicine, Department of Psychiatry, Columbia University Irving Medical Center, and Robert N Butler Columbia Aging Center, Mailman School of Public Health, Columbia University, New York, NY, USA
| | - Mylène Aubertin-Leheudre
- Département des sciences de l'activité physique, Université du Québec À Montréal, Montreal, QC, Canada; Groupe de recherche en Activité Physique Adaptée, Montréal, QC, Canada; Centre de Recherche de l'Institut Universitaire de Gériatrie de Montréal, Montreal, QC, Canada
| | - Marc Bélanger
- Département des sciences de l'activité physique, Université du Québec À Montréal, Montreal, QC, Canada; Groupe de recherche en Activité Physique Adaptée, Montréal, QC, Canada
| | - Richard Robitaille
- Groupe de recherche en Activité Physique Adaptée, Montréal, QC, Canada; Département de neurosciences, Université de Montréal, Montreal, QC, Canada; Centre interdisciplinaire de recherche sur le cerveau et l'apprentissage, Montreal, QC, Canada
| | - José A Morais
- Groupe de recherche en Activité Physique Adaptée, Montréal, QC, Canada; Research Institute of the McGill University Health Centre, Montreal, QC, Canada; Division of Geriatric Medicine, Faculty of Medicine, McGill University, Montreal, QC, Canada
| | - Gilles Gouspillou
- Département des sciences de l'activité physique, Université du Québec À Montréal, Montreal, QC, Canada; Groupe de recherche en Activité Physique Adaptée, Montréal, QC, Canada; Centre de Recherche de l'Institut Universitaire de Gériatrie de Montréal, Montreal, QC, Canada; Meakins-Christie Laboratories, Department of Medicine, McGill University, Montreal, QC, Canada.
| |
Collapse
|
5
|
Tian Q, Zweibaum DA, Qian Y, Oppong RF, Pilling LC, Casanova F, Atkins JL, Melzer D, Ding J, Ferrucci L. Mitochondrial DNA copy number associated dementia risk by somatic mutations and frailty. GeroScience 2025; 47:825-835. [PMID: 39313624 PMCID: PMC11872790 DOI: 10.1007/s11357-024-01355-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Accepted: 09/13/2024] [Indexed: 09/25/2024] Open
Abstract
Mitochondrial dysfunction is linked to physical impairment and dementia. Mitochondrial DNA copy number (mtDNAcn) from blood may predict cognitive decline and dementia risk, but the effect of somatic mutations or frailty is unknown. We estimated mtDNAcn using fastMitoCalc and microheteroplasmies using mitoCaller, from Whole Genome Sequencing (WGS) data. In 189,566 participants free of dementia at study entry (mean age = 56 ± 8), we examined the association between mtDNAcn and subsequent dementia diagnosis using Cox regression. Cognition was assessed in a subset on average 8.9 years later. We examined the associations between mtDNAcn and cognitive measures using multivariable linear regression, adjusted for demographic factors, mtDNAcn-related parameters, and apolipoprotein E ε4 status. We further stratified by frailty and microheteroplasmies. Over an average follow-up of 13.2 years, 3533 participants developed dementia. Each SD higher mtDNAcn (16) was associated with 4.2% lower all-cause dementia hazard (HR = 0.958, p = 0.030), 6% lower non-AD dementia hazard (HR = 0.933, p = 0.022), and not-AD dementia hazard. The associations between mtDNAcn and all-cause dementia and non-AD dementia were stronger among those who were pre-frail or frail or with higher microheteroplasmies. Higher mtDNAcn was associated with higher DSST scores (p = 0.036) and significant only among those with higher microheteroplasmies or frailty (p = 0.029 and 0.048, respectively). mtDNAcn was also associated with delta TMT and paired associate learning only in pre-frail/frail participants (p = 0.007 and 0.045, respectively). Higher WGS-based mtDNAcn in human blood is associated with lower dementia risk, specifically non-AD dementia, and specific cognitive function. The relationships appear stronger in high somatic mutations or frailty. Future studies are warranted to investigate biological underpinnings.
Collapse
Affiliation(s)
- Qu Tian
- Translational Gerontology Branch, National Institute on Aging Intramural Research Program, 251 Bayview Blvd., Suite 100, Baltimore, MD, 21224, USA.
| | - David A Zweibaum
- Translational Gerontology Branch, National Institute on Aging Intramural Research Program, 251 Bayview Blvd., Suite 100, Baltimore, MD, 21224, USA
| | - Yong Qian
- Translational Gerontology Branch, National Institute on Aging Intramural Research Program, 251 Bayview Blvd., Suite 100, Baltimore, MD, 21224, USA
| | - Richard F Oppong
- Translational Gerontology Branch, National Institute on Aging Intramural Research Program, 251 Bayview Blvd., Suite 100, Baltimore, MD, 21224, USA
| | - Luke C Pilling
- Epidemiology & Public Health Group, Department of Clinical & Biomedical Science, Faculty of Health & Life Sciences, University of Exeter, College House, University of Exeter, St Luke's Campus, Heavitree Road, Exeter Devon, EX1 2LU, UK
| | - Francesco Casanova
- Epidemiology & Public Health Group, Department of Clinical & Biomedical Science, Faculty of Health & Life Sciences, University of Exeter, College House, University of Exeter, St Luke's Campus, Heavitree Road, Exeter Devon, EX1 2LU, UK
| | - Janice L Atkins
- Epidemiology & Public Health Group, Department of Clinical & Biomedical Science, Faculty of Health & Life Sciences, University of Exeter, College House, University of Exeter, St Luke's Campus, Heavitree Road, Exeter Devon, EX1 2LU, UK
| | - David Melzer
- Epidemiology & Public Health Group, Department of Clinical & Biomedical Science, Faculty of Health & Life Sciences, University of Exeter, College House, University of Exeter, St Luke's Campus, Heavitree Road, Exeter Devon, EX1 2LU, UK
| | - Jun Ding
- Translational Gerontology Branch, National Institute on Aging Intramural Research Program, 251 Bayview Blvd., Suite 100, Baltimore, MD, 21224, USA
| | - Luigi Ferrucci
- Translational Gerontology Branch, National Institute on Aging Intramural Research Program, 251 Bayview Blvd., Suite 100, Baltimore, MD, 21224, USA
| |
Collapse
|
6
|
Goulding RP, Charlton BT, Breedveld EA, van der Laan M, Strating AR, Noort W, Kolodyazhna A, Appelman B, van Vugt M, Grootemaat AE, van der Wel NN, de Koning JJ, Bloemers FW, Wüst RCI. Skeletal muscle mitochondrial fragmentation predicts age-associated decline in physical capacity. Aging Cell 2025; 24:e14386. [PMID: 39630001 PMCID: PMC11822651 DOI: 10.1111/acel.14386] [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: 04/03/2024] [Revised: 08/09/2024] [Accepted: 10/04/2024] [Indexed: 02/14/2025] Open
Abstract
Ageing substantially impairs skeletal muscle metabolic and physical function. Skeletal muscle mitochondrial health is also impaired with ageing, but the role of skeletal muscle mitochondrial fragmentation in age-related functional decline remains imprecisely characterized. Here, using a cross-sectional study design, we performed a detailed comparison of skeletal muscle mitochondrial characteristics in relation to in vivo markers of exercise capacity between young and middle-aged individuals. Despite similar overall oxidative phosphorylation capacity (young: 99 ± 17 vs. middle-aged: 99 ± 27 pmol O2.s-1.mg-1, p = 0.95) and intermyofibrillar mitochondrial density (young: 5.86 ± 0.57 vs. middle-aged: 5.68 ± 1.48%, p = 0.25), older participants displayed a more fragmented intermyofibrillar mitochondrial network (young: 1.15 ± 0.17 vs. middle-aged: 1.55 ± 0.15 A.U., p < 0.0001), a lower mitochondrial cristae density (young: 23.40 ± 7.12 vs. middle-aged: 13.55 ± 4.10%, p = 0.002) and a reduced subsarcolemmal mitochondrial density (young: 22.39 ± 6.50 vs. middle-aged: 13.92 ± 4.95%, p = 0.005). Linear regression analysis showed that 87% of the variance associated with maximal oxygen uptake could be explained by skeletal muscle mitochondrial fragmentation and cristae density alone, whereas subsarcolemmal mitochondrial density was positively associated with the capacity for oxygen extraction during exercise. Intramuscular lipid accumulation was positively associated with mitochondrial fragmentation and negatively associated with cristae density. Collectively, our work highlights the critical role of skeletal muscle mitochondria in age-associated declines in physical function.
Collapse
Affiliation(s)
- Richie P. Goulding
- Department of Human Movement Sciences, Faculty of Behavioural and Movement Sciences, Amsterdam Movement SciencesVrije Universiteit AmsterdamAmsterdamThe Netherlands
| | - Braeden T. Charlton
- Department of Human Movement Sciences, Faculty of Behavioural and Movement Sciences, Amsterdam Movement SciencesVrije Universiteit AmsterdamAmsterdamThe Netherlands
| | - Ellen A. Breedveld
- Department of Human Movement Sciences, Faculty of Behavioural and Movement Sciences, Amsterdam Movement SciencesVrije Universiteit AmsterdamAmsterdamThe Netherlands
| | - Matthijs van der Laan
- Department of Human Movement Sciences, Faculty of Behavioural and Movement Sciences, Amsterdam Movement SciencesVrije Universiteit AmsterdamAmsterdamThe Netherlands
| | - Anne R. Strating
- Department of Human Movement Sciences, Faculty of Behavioural and Movement Sciences, Amsterdam Movement SciencesVrije Universiteit AmsterdamAmsterdamThe Netherlands
| | - Wendy Noort
- Department of Human Movement Sciences, Faculty of Behavioural and Movement Sciences, Amsterdam Movement SciencesVrije Universiteit AmsterdamAmsterdamThe Netherlands
| | - Aryna Kolodyazhna
- Department of Human Movement Sciences, Faculty of Behavioural and Movement Sciences, Amsterdam Movement SciencesVrije Universiteit AmsterdamAmsterdamThe Netherlands
| | - Brent Appelman
- Center for Experimental and Molecular MedicineAmsterdam UMC Location University of AmsterdamAmsterdamThe Netherlands
- Amsterdam Institute for Infection and ImmunityAmsterdamThe Netherlands
| | - Michèle van Vugt
- Center for Experimental and Molecular MedicineAmsterdam UMC Location University of AmsterdamAmsterdamThe Netherlands
- Division of Infectious Diseases, Tropical Medicine, Department of MedicineAmsterdam UMC Location University of AmsterdamAmsterdamThe Netherlands
| | - Anita E. Grootemaat
- Electron Microscopy Centre AmsterdamAmsterdam UMC Location Academic Medical CentreAmsterdamThe Netherlands
| | - Nicole N. van der Wel
- Electron Microscopy Centre AmsterdamAmsterdam UMC Location Academic Medical CentreAmsterdamThe Netherlands
| | - Jos J. de Koning
- Department of Human Movement Sciences, Faculty of Behavioural and Movement Sciences, Amsterdam Movement SciencesVrije Universiteit AmsterdamAmsterdamThe Netherlands
| | - Frank W. Bloemers
- Department of Trauma Surgery, Amsterdam Movement Sciences, Amsterdam UMCVrije Universiteit AmsterdamAmsterdamThe Netherlands
| | - Rob C. I. Wüst
- Department of Human Movement Sciences, Faculty of Behavioural and Movement Sciences, Amsterdam Movement SciencesVrije Universiteit AmsterdamAmsterdamThe Netherlands
| |
Collapse
|
7
|
Marcinek DJ, Ferrucci L. Reduced oxidative capacity of skeletal muscle mitochondria IS a fundamental consequence of adult ageing. J Physiol 2025; 603:17-20. [PMID: 38970753 DOI: 10.1113/jp285040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Accepted: 04/03/2024] [Indexed: 07/08/2024] Open
Affiliation(s)
- David J Marcinek
- Department of Radiology, University of Washington, Seattle, Washington, USA
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington, USA
| | - Luigi Ferrucci
- Longitudinal Studies Section, Translational Gerontology Branch, National Institute on Aging, Baltimore, Maryland, USA
| |
Collapse
|
8
|
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
|
9
|
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
|
10
|
Kim H, Ranjit R, Claflin DR, Georgescu C, Wren JD, Brooks SV, Miller BF, Ahn B. Unacylated Ghrelin Protects Against Age-Related Loss of Muscle Mass and Contractile Dysfunction in Skeletal Muscle. Aging Cell 2024; 23:e14323. [PMID: 39223708 PMCID: PMC11634730 DOI: 10.1111/acel.14323] [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: 03/08/2024] [Revised: 07/20/2024] [Accepted: 08/10/2024] [Indexed: 09/04/2024] Open
Abstract
Sarcopenia, the progressive loss of muscle mass and function, universally affects older adults and is closely associated with frailty and reduced quality of life. Despite the inevitable consequences of sarcopenia and its relevance to healthspan, no pharmacological therapies are currently available. Ghrelin is a gut-released hormone that increases appetite and body weight through acylation. Acylated ghrelin activates its receptor, growth hormone secretagogue receptor 1a (GHSR1a), in the brain by binding to it. Studies have demonstrated that acyl and unacylated ghrelin (UnAG) both have protective effects against acute pathological conditions independent of receptor activation. Here, we investigated the long-term effects of UnAG in age-associated muscle atrophy and contractile dysfunction in mice. Four-month-old and 18-month-old mice were subjected to either UnAG or control treatment for 10 months. UnAG did not affect food consumption or body weight. Gastrocnemius and quadriceps muscle weights were reduced by 20%-30% with age, which was partially protected against by UnAG. Specific force, force per cross-sectional area, measured in isolated extensor digitorum longus muscle was diminished by 30% in old mice; however, UnAG prevented the loss of specific force. UnAG also protected from decreases in mitochondrial respiration and increases in hydrogen peroxide generation of skeletal muscle of old mice. Results of bulk mRNA-seq analysis and our contractile function data show that UnAG reversed neuromuscular junction impairment that occurs with age. Collectively, our data revealed the direct role of UnAG in mitigating sarcopenia in mice, independent of food consumption or body weight, implicating UnAG treatment as a potential therapy against sarcopenia.
Collapse
Affiliation(s)
- Hyunyoung Kim
- Department of Internal MedicineWake Forest University School of MedicineWinston‐SalemNorth CarolinaUSA
| | - Rojina Ranjit
- Aging and Metabolism Research ProgramOklahoma Medical Research FoundationOklahoma CityOklahomaUSA
- Department of BiochemistryUniversity of Oklahoma Health Sciences CenterOklahoma CityOklahomaUSA
| | - Dennis R. Claflin
- Department of Biomedical EngineeringUniversity of MichiganAnn ArborMichiganUSA
- Department of Molecular and Integrative PhysiologyUniversity of MichiganAnn ArborMichiganUSA
| | - Constantin Georgescu
- Genes and Human Disease Research ProgramOklahoma Medical Research FoundationOklahoma CityOklahomaUSA
| | - Jonathan D. Wren
- Genes and Human Disease Research ProgramOklahoma Medical Research FoundationOklahoma CityOklahomaUSA
| | - Susan V. Brooks
- Department of Biomedical EngineeringUniversity of MichiganAnn ArborMichiganUSA
- Department of Molecular and Integrative PhysiologyUniversity of MichiganAnn ArborMichiganUSA
| | - Benjamin F. Miller
- Aging and Metabolism Research ProgramOklahoma Medical Research FoundationOklahoma CityOklahomaUSA
- Oklahoma City VA Medical CenterOklahoma CityOklahomaUSA
| | - Bumsoo Ahn
- Department of Internal MedicineWake Forest University School of MedicineWinston‐SalemNorth CarolinaUSA
| |
Collapse
|
11
|
Wanigatunga AA, Liu F, Dougherty RJ, Roche KB, Urbanek JK, Zampino M, Simonsick EM, Tian Q, Schrack JA, Ferrucci L. Relationship between skeletal mitochondrial function and digital markers of free-living physical activity in older adults. GeroScience 2024; 46:6173-6182. [PMID: 38809390 PMCID: PMC11493922 DOI: 10.1007/s11357-024-01212-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Accepted: 05/16/2024] [Indexed: 05/30/2024] Open
Abstract
This study examined the association between in vivo skeletal mitochondrial function and digital free-living physical activity patterns-a measure that summarizes biological, phenotypic, functional, and environmental effects on mobility. Among 459 participants (mean age 68 years; 55% women) in the Baltimore Longitudinal Study of Aging, mitochondrial function was quantified as skeletal muscle oxidative capacity via post-exercise phosphocreatine recovery rate (τPCr) in the vastus lateralis muscle of the left thigh, using 31P magnetic resonance spectroscopy. Accelerometry was collected using a 7-day, 24-h wrist-worn protocol and summarized into activity amount, intensity, endurance, and accumulation patterning metrics. Linear regression, two-part linear and logistic (bout analyses), and linear mixed effects models (time-of-day analyses) were used to estimate associations between τPCr and each physical activity metric. Interactions by age, sex, and gait speed were tested. After covariate adjustment, higher τPCr (or poorer mitochondrial function) was associated with lower activity counts/day (β = - 6593.7, SE = 2406.0; p = 0.006) and activity intensity (- 81.5 counts, SE = 12.9; p < 0.001). For activity intensity, the magnitude of association was greater for men and those with slower gait speed (interaction p < 0.02 for both). Conversely, τPCr was not associated with daily active minutes/day (p = 0.15), activity fragmentation (p = 0.13), or endurance at any bout length (p > 0.05 for all). Time-of-day analyses show participants with high τPCr were less active from 6:00 a.m. to 12:00 a.m. than those with low τPCr. Results indicate that poorer skeletal mitochondrial function is primarily associated with lower engagement in high intensity activities. Our findings help define the connection between laboratory-measured mitochondrial function and real-world physical activity behavior.
Collapse
Affiliation(s)
- Amal A Wanigatunga
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA.
- Center On Aging and Health, Johns Hopkins University and Medical Institutions, Baltimore, MD, USA.
- , Baltimore, MD, 21025, USA.
| | - Fangyu Liu
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Ryan J Dougherty
- Center On Aging and Health, Johns Hopkins University and Medical Institutions, Baltimore, MD, USA
- Department of Neurology, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Karen Bandeen Roche
- Center On Aging and Health, Johns Hopkins University and Medical Institutions, Baltimore, MD, USA
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Jacek K Urbanek
- Center On Aging and Health, Johns Hopkins University and Medical Institutions, Baltimore, MD, USA
- Biostatistics and Data Management, Regeneron Pharmaceuticals, Tarrytown, NY, USA
| | - Marta Zampino
- Department of Internal Medicine, University of Maryland, Baltimore, MD, USA
| | - Eleanor M Simonsick
- Intramural Research Program, National Institute On Aging, National Institutes of Health, Baltimore, MD, USA
| | - Qu Tian
- Intramural Research Program, National Institute On Aging, National Institutes of Health, Baltimore, MD, USA
| | - Jennifer A Schrack
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
- Center On Aging and Health, Johns Hopkins University and Medical Institutions, Baltimore, MD, USA
| | - Luigi Ferrucci
- Intramural Research Program, National Institute On Aging, National Institutes of Health, Baltimore, MD, USA
| |
Collapse
|
12
|
Piergiovanni S, Terrier P. Validity of Linear and Nonlinear Measures of Gait Variability to Characterize Aging Gait with a Single Lower Back Accelerometer. SENSORS (BASEL, SWITZERLAND) 2024; 24:7427. [PMID: 39685964 DOI: 10.3390/s24237427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Revised: 11/15/2024] [Accepted: 11/19/2024] [Indexed: 12/18/2024]
Abstract
The attractor complexity index (ACI) is a recently developed gait analysis tool based on nonlinear dynamics. This study assesses ACI's sensitivity to attentional demands in gait control and its potential for characterizing age-related changes in gait patterns. Furthermore, we compare ACI with classical gait metrics to determine its efficacy relative to established methods. A 4 × 200 m indoor walking test with a triaxial accelerometer attached to the lower back was used to compare gait patterns of younger (N = 42) and older adults (N = 60) during normal and metronome walking. The other linear and non-linear gait metrics were movement intensity, gait regularity, local dynamic stability (maximal Lyapunov exponents), and scaling exponent (detrended fluctuation analysis). In contrast to other gait metrics, ACI demonstrated a specific sensitivity to metronome walking, with both young and old participants exhibiting altered stride interval correlations. Furthermore, there was a significant difference between the young and old groups (standardized effect size: -0.77). Additionally, older participants exhibited slower walking speeds, a reduced movement intensity, and a lower gait regularity. The ACI is likely a sensitive marker for attentional load and can effectively discriminate age-related changes in gait patterns. Its ease of measurement makes it a promising tool for gait analysis in unsupervised (free-living) conditions.
Collapse
Affiliation(s)
- Sophia Piergiovanni
- Haute-Ecole Arc Santé, HES-SO University of Applied Sciences and Arts Western Switzerland, 2000 Neuchâtel, Switzerland
| | - Philippe Terrier
- Haute-Ecole Arc Santé, HES-SO University of Applied Sciences and Arts Western Switzerland, 2000 Neuchâtel, Switzerland
| |
Collapse
|
13
|
Craige SM, Mammel RK, Amiri N, Willoughby OS, Drake JC. Interplay of ROS, mitochondrial quality, and exercise in aging: Potential role of spatially discrete signaling. Redox Biol 2024; 77:103371. [PMID: 39357424 PMCID: PMC11474192 DOI: 10.1016/j.redox.2024.103371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 09/05/2024] [Accepted: 09/23/2024] [Indexed: 10/04/2024] Open
Affiliation(s)
- Siobhan M Craige
- Department of Human Nutrition, Foods, and Exercise, Virginia Tech, Blacksburg, 24061, USA.
| | - Rebecca K Mammel
- Department of Human Nutrition, Foods, and Exercise, Virginia Tech, Blacksburg, 24061, USA
| | - Niloufar Amiri
- Department of Human Nutrition, Foods, and Exercise, Virginia Tech, Blacksburg, 24061, USA; Institute for Critical Technology and Applied Science, Virginia Tech, Blacksburg, 24061, USA
| | - Orion S Willoughby
- Department of Human Nutrition, Foods, and Exercise, Virginia Tech, Blacksburg, 24061, USA
| | - Joshua C Drake
- Department of Human Nutrition, Foods, and Exercise, Virginia Tech, Blacksburg, 24061, USA.
| |
Collapse
|
14
|
Delpino MV, Quarleri J. Aging mitochondria in the context of SARS-CoV-2: exploring interactions and implications. FRONTIERS IN AGING 2024; 5:1442323. [PMID: 39380657 PMCID: PMC11458564 DOI: 10.3389/fragi.2024.1442323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/01/2024] [Accepted: 09/16/2024] [Indexed: 10/10/2024]
Abstract
The coronavirus disease 2019 (COVID-19), caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has presented global challenges with a diverse clinical spectrum, including severe respiratory complications and systemic effects. This review explores the intricate relationship between mitochondrial dysfunction, aging, and obesity in COVID-19. Mitochondria are vital for cellular energy provision and resilience against age-related macromolecule damage accumulation. They manage energy allocation in cells, activating adaptive responses and stress signals such as redox imbalance and innate immunity activation. As organisms age, mitochondrial function diminishes. Aging and obesity, linked to mitochondrial dysfunction, compromise the antiviral response, affecting the release of interferons, and worsening COVID-19 severity. Furthermore, the development of post-acute sequelae of SARS-CoV-2 infection (PASC), also known as long COVID has been associated with altered energy metabolism, and chronic immune dysregulation derived from mitochondrial dysfunction. Understanding the interplay between mitochondria, aging, obesity, and viral infections provides insights into COVID-19 pathogenesis. Targeting mitochondrial health may offer potential therapeutic strategies to mitigate severe outcomes and address long-term consequences in infected individuals.
Collapse
|
15
|
Garcia RE, Blackwell TL, Forman DE, Coen PM, Nicklas BJ, Qiao Y(S, Cawthon PM, Toledo FGS, Goodpaster BH, Cummings SR, Newman AB, Glynn NW. Role of Walking Energetics and Perceived Fatigability Differs by Gait Speed: The Study of Muscle, Mobility and Aging (SOMMA). J Gerontol A Biol Sci Med Sci 2024; 79:glae187. [PMID: 39066507 PMCID: PMC11341985 DOI: 10.1093/gerona/glae187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Indexed: 07/28/2024] Open
Abstract
BACKGROUND Slower gait speed may be driven by greater energy deficits and fatigability among older adults. We examined associations of walking energetics and perceived physical fatigability with gait speed among slower and faster walkers. Additionally, we used statistical mediation to examine the role of fatigability in the associations of walking energetics and gait speed using the Study of Muscle, Mobility and Aging (SOMMA). METHODS Perceived physical fatigability was assessed using the Pittsburgh Fatigability Scale (PFS) Physical score (range 0-50, higher = greater). A 3-phase cardiopulmonary exercise treadmill test collected peak oxygen consumption (VO2peak, mL/kg/min), energetic cost of walking (ECW, mL/kg/m), and cost-capacity ratio (VO2/VO2peak*100, %). Slower (<1.01 m/s) versus faster (≥1.01 m/s) walkers were classified using median 4-m gait speed. Linear regressions and statistical mediation analyses were conducted. RESULTS Slower walkers had lower VO2peak, higher ECW at preferred walking speed (PWS), and greater PFS Physical score compared to faster walkers (all p < .05; N = 849). One standard deviation (1-SD) higher VO2peak was associated with 0.1 m/s faster gait speed, while 1-SD higher ECW PWS, cost-capacity ratio at PWS and slow walking speed (SWS), and PFS Physical score were associated with 0.02-0.23 m/s slower gait speed. PFS Physical score was a significant statistical mediator in the associations between VO2peak (15.2%), SWS cost-capacity ratio (15.9%), and ECW PWS (10.7%) with gait speed and was stronger among slower walkers. CONCLUSIONS Slower walkers may be more influenced by perceptions of fatigue in addition to walking energetics. Our work highlights the importance of targeting both energetics and perceived fatigability to prevent mobility decline.
Collapse
Affiliation(s)
- Reagan E Garcia
- Department of Epidemiology, School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Terri L Blackwell
- San Francisco Coordinating Center, California Pacific Medical Center Research Institute, San Francisco, California, USA
| | - Daniel E Forman
- Department of Medicine (Divisions of Cardiology and Geriatrics), University of Pittsburgh School of Medicine, and Geriatrics, Research, Education, and Clinical Center (GRECC), Pittsburgh, Pennsylvania, USA
| | - Paul M Coen
- Translational Research Institute for Metabolism and Diabetes, Advent Health, Orlando, Florida, USA
| | - Barbara J Nicklas
- Department of Internal Medicine, Section on Gerontology and Geriatric Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
| | - Yujia (Susanna) Qiao
- San Francisco Coordinating Center, California Pacific Medical Center Research Institute, 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, San Francisco, California, USA
| | - Frederico G S Toledo
- Department of Medicine (Divisions of Endocrinology and Metabolism), University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Bret H Goodpaster
- Translational Research Institute for Metabolism and Diabetes, Advent Health, Orlando, Florida, USA
| | - Steven R Cummings
- San Francisco Coordinating Center, California Pacific Medical Center Research Institute, San Francisco, California, USA
| | - Anne B Newman
- Department of Epidemiology, School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Nancy W Glynn
- Department of Epidemiology, School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
16
|
Berkman AM, Goodenough CG, Durakiewicz P, Howell CR, Wang Z, Easton J, Mulder HL, Armstrong GT, Hudson MM, Kundu M, Ness KK. Associations between mitochondrial copy number, exercise capacity, physiologic cost of walking, and cardiac strain in young adult survivors of childhood cancer. J Cancer Surviv 2024; 18:1154-1167. [PMID: 38635100 PMCID: PMC11324404 DOI: 10.1007/s11764-024-01590-7] [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: 10/31/2023] [Accepted: 04/04/2024] [Indexed: 04/19/2024]
Abstract
PURPOSE Childhood cancer survivors are at risk for cardiac dysfunction and impaired physical performance, though underlying cellular mechanisms are not well studied. In this cross-sectional study, we examined the association between peripheral blood mitochondrial DNA copy number (mtDNA-CN, a proxy for mitochondrial function) and markers of performance impairment and cardiac dysfunction. METHODS Whole-genome sequencing, validated by quantitative polymerase chain reaction, was used to estimate mtDNA-CN in 1720 adult survivors of childhood cancer (48.5% female; mean age = 30.7 years, standard deviation (SD) = 9.0). Multivariable logistic regression was performed to evaluate the associations between mtDNA-CN and exercise intolerance, walking inefficiency, and abnormal global longitudinal strain (GLS), adjusting for treatment exposures, age, sex, and race and ethnicity. RESULTS The prevalence of exercise intolerance, walking inefficiency, and abnormal GLS among survivors was 25.7%, 10.7%, and 31.7%, respectively. Each SD increase of mtDNA-CN was associated with decreased odds of abnormal GLS (adjusted odds ratio (OR) = 0.88, p = 0.04) but was not associated with exercise intolerance (OR = 1.02, p = 0.76) or walking inefficiency (OR = 1.06, p = 0.46). Alkylating agent exposure was associated with increased odds of exercise intolerance (OR = 2.25, p < 0.0001), walking inefficiency (OR = 2.37, p < 0.0001), and abnormal GLS (OR = 1.78, p = 0.0002). CONCLUSIONS Increased mtDNA-CN is associated with decreased odds of abnormal cardiac function in childhood cancer survivors. IMPLICATIONS FOR CANCER SURVIVORS These findings demonstrate a potential role for mtDNA-CN as a biomarker of early cardiac dysfunction in this population.
Collapse
Affiliation(s)
- Amy M Berkman
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Chelsea G Goodenough
- Department of Epidemiology and Cancer Control, St. Jude Children's Research Hospital, 262 Danny Thomas Place, MS-735, Memphis, TN, 38105, USA
| | - Paul Durakiewicz
- Department of Epidemiology and Cancer Control, St. Jude Children's Research Hospital, 262 Danny Thomas Place, MS-735, Memphis, TN, 38105, USA
| | - Carrie R Howell
- Division of Preventive Medicine, Department of Medicine, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Zhaoming Wang
- Department of Epidemiology and Cancer Control, St. Jude Children's Research Hospital, 262 Danny Thomas Place, MS-735, Memphis, TN, 38105, USA
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - John Easton
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Heather L Mulder
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Gregory T Armstrong
- Department of Epidemiology and Cancer Control, St. Jude Children's Research Hospital, 262 Danny Thomas Place, MS-735, Memphis, TN, 38105, USA
| | - Melissa M Hudson
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, TN, USA
- Department of Epidemiology and Cancer Control, St. Jude Children's Research Hospital, 262 Danny Thomas Place, MS-735, Memphis, TN, 38105, USA
| | - Mondira Kundu
- Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Kirsten K Ness
- Department of Epidemiology and Cancer Control, St. Jude Children's Research Hospital, 262 Danny Thomas Place, MS-735, Memphis, TN, 38105, USA.
| |
Collapse
|
17
|
Mau T, Blackwell TL, Cawthon PM, Molina AJA, Coen PM, Distefano G, Kramer PA, Ramos SV, Forman DE, Goodpaster BH, Toledo FGS, Duchowny KA, Sparks LM, Newman AB, Kritchevsky SB, Cummings SR. Muscle Mitochondrial Bioenergetic Capacities Are Associated With Multimorbidity Burden in Older Adults: The Study of Muscle, Mobility and Aging. J Gerontol A Biol Sci Med Sci 2024; 79:glae101. [PMID: 38605684 PMCID: PMC11167490 DOI: 10.1093/gerona/glae101] [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: 12/14/2023] [Indexed: 04/13/2024] Open
Abstract
BACKGROUND The geroscience hypothesis posits that aging biological processes contribute to many age-related deficits, including the accumulation of multiple chronic diseases. Though only one facet of mitochondrial function, declines in muscle mitochondrial bioenergetic capacities may contribute to this increased susceptibility to multimorbidity. METHODS The Study of Muscle, Mobility and Aging (SOMMA) assessed ex vivo muscle mitochondrial energetics in 764 older adults (mean age = 76.4, 56.5% women, and 85.9% non-Hispanic White) by high-resolution respirometry of permeabilized muscle fibers. We estimated the proportional odds ratio (POR [95% CI]) for the likelihood of greater multimorbidity (4 levels: 0 conditions, N = 332; 1 condition, N = 299; 2 conditions, N = 98; or 3+ conditions, N = 35) from an index of 11 conditions, per SD decrement in muscle mitochondrial energetic parameters. Distribution of conditions allowed for testing the associations of maximal muscle energetics with some individual conditions. RESULTS Lower oxidative phosphorylation supported by fatty acids and/or complex I- and II-linked carbohydrates (eg, Max OXPHOSCI+CII) was associated with a greater multimorbidity index score (POR = 1.32 [1.13, 1.54]) and separately with diabetes mellitus (OR = 1.62 [1.26, 2.09]), depressive symptoms (OR = 1.45 [1.04, 2.00]) and possibly chronic kidney disease (OR = 1.57 [0.98, 2.52]) but not significantly with other conditions (eg, cardiac arrhythmia, chronic obstructive pulmonary disease). CONCLUSIONS Lower muscle mitochondrial bioenergetic capacities were associated with a worse composite multimorbidity index score. Our results suggest that decrements in muscle mitochondrial energetics may contribute to a greater global burden of disease and are more strongly related to some conditions than others.
Collapse
Affiliation(s)
- 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, San Francisco, California, USA
| | - Terri L Blackwell
- San Francisco Coordinating Center, California Pacific Medical Center Research Institute, 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, San Francisco, California, USA
| | - Anthony J A Molina
- Division of Geriatrics, Gerontology, and Palliative Care, Department of Medicine, University of California San Diego School of Medicine, La Jolla, California, USA
| | - Paul M Coen
- Translational Research Institute, AdventHealth, Orlando, Florida, USA
| | | | - Philip A Kramer
- Gerontology and Geriatric Medicine, Department of Internal Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
| | - Sofhia V Ramos
- Translational Research Institute, AdventHealth, Orlando, Florida, USA
| | - Daniel E Forman
- Division of Geriatrics and Cardiology, Department of Medicine, University of Pittsburgh, Geriatrics Research, Education, and Clinical Care (GRECC), VA Pittsburgh Healthcare System, Pittsburgh, Pennsylvania, USA
| | - Bret H Goodpaster
- Translational Research Institute, AdventHealth, Orlando, Florida, USA
| | - Frederico G S Toledo
- Division of Endocrinology and Metabolism, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Kate A Duchowny
- Social Environment and Health, Survey Research Center, Institute for Social Research, University of Michigan, Ann Arbor, Michigan, USA
| | - Lauren M Sparks
- Translational Research Institute, AdventHealth, Orlando, Florida, USA
| | - Anne B Newman
- Department of Epidemiology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Stephen B Kritchevsky
- Gerontology and Geriatric Medicine, Department of Internal Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
| | - 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, San Francisco, California, USA
| |
Collapse
|
18
|
Zhang TR, Chiang CH, Hsu TC, Wang CY, Chen CY. Age and dietary restriction modulate mitochondrial quality in quadriceps femoris muscle of male mice. Biogerontology 2024; 25:447-459. [PMID: 38183523 DOI: 10.1007/s10522-023-10086-3] [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] [Received: 11/06/2023] [Accepted: 11/27/2023] [Indexed: 01/08/2024]
Abstract
Dietary restriction (DR) is a potential intervention for ameliorating ageing-related damages. Mitochondrial quality control is the key mechanism for regulating cellular functions in skeletal muscle. This study aimed to explore the effect of age and DR on the homeostasis of mitochondrial quality control in skeletal muscle. To study the effect of age on mitochondrial homeostasis, young (3 months old) male C57BL/6J mice were fed ad libitum (AL) until 7 (Young), 14 (Middle), and 19 months (Aged) of age. For the DR intervention, 60% of AL intake was given to the mice at 3 months of age until they reached 19 months of age (16 months). The quadriceps femoris muscle was collected for further analysis. Significant changes in the skeletal muscle were noticed during the transition between middle age and the elderly stages. An accumulation of collagen was observed in the muscle after middle age. Compared with the Middle muscle, Aged muscle displayed a greater expression of VDAC, and lower expressions of mitochondrial dynamic proteins and OXPHOS proteins. The DR intervention attenuated collagen content and elongated the sarcomere length in the skeletal muscle during ageing. In addition, DR adjusted the abnormalities in mitochondrial morphology in the Aged muscle. DR downregulated VDAC expression, but upregulated OPA1 and DRP1 expressions. Taken together, greater pathological changes were noticed in the skeletal muscle during ageing, especially in the transition between middle age and the elderly, whereas early-onset DR attenuated the muscular ageing via normalising partial functions of mitochondria.
Collapse
Affiliation(s)
- Ting-Rui Zhang
- Department of Animal Science and Technology, National Taiwan University, No. 50, Lane 155, Sec 3, Keelung Rd, Taipei, 10672, Taiwan
| | - Chun-Hsien Chiang
- Department of Animal Science and Technology, National Taiwan University, No. 50, Lane 155, Sec 3, Keelung Rd, Taipei, 10672, Taiwan
| | - Tzu-Chieh Hsu
- Department of Animal Science and Technology, National Taiwan University, No. 50, Lane 155, Sec 3, Keelung Rd, Taipei, 10672, Taiwan
| | - Chih-Yun Wang
- Department of Animal Science and Technology, National Taiwan University, No. 50, Lane 155, Sec 3, Keelung Rd, Taipei, 10672, Taiwan
| | - Ching-Yi Chen
- Department of Animal Science and Technology, National Taiwan University, No. 50, Lane 155, Sec 3, Keelung Rd, Taipei, 10672, Taiwan.
| |
Collapse
|
19
|
Irving BA, Kunz HE. Unraveling the roles of ectopic adipose depots and physical activity in age-related mitochondrial decline. Obesity (Silver Spring) 2024; 32:1043-1044. [PMID: 38803313 PMCID: PMC11863171 DOI: 10.1002/oby.24048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Accepted: 04/09/2024] [Indexed: 05/29/2024]
Affiliation(s)
- Brian A. Irving
- School of Kinesiology, Louisiana State University, Baton Rouge, Louisiana, USA
- Pennington Biomedical Research Center, Baton Rouge, Louisiana, USA
| | - Hawley E. Kunz
- Endocrine Research Unit, Division of Endocrinology, Department of Internal Medicine, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
20
|
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
|
21
|
Gay EL, Coen PM, Harrison S, Garcia RE, Qiao YS, Goodpaster BH, Forman DE, Toledo FGS, Distefano G, Kramer PA, Ramos SV, Molina AJA, Nicklas BJ, Cummings SR, Cawthon PM, Hepple RT, Newman AB, Glynn NW. Sex Differences in the Association between Skeletal Muscle Energetics and Perceived Physical Fatigability: The Study of Muscle, Mobility and Aging (SOMMA). MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.05.25.24307934. [PMID: 38853946 PMCID: PMC11160809 DOI: 10.1101/2024.05.25.24307934] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2024]
Abstract
Greater perceived physical fatigability and lower skeletal muscle energetics are predictors of mobility decline. Characterizing associations between muscle energetics and perceived fatigability may provide insight into potential targets to prevent mobility decline. We examined associations of in vivo (maximal ATP production, ATPmax) and ex vivo (maximal carbohydrate supported oxidative phosphorylation [max OXPHOS] and maximal fatty acid supported OXPHOS [max FAO OXPHOS]) measures of mitochondrial energetics with two measures of perceived physical fatigability, Pittsburgh Fatigability Scale (PFS, 0-50, higher=greater) and Rating of Perceived Exertion (RPE Fatigability, 6-20, higher=greater) after a slow treadmill walk. Participants from the Study of Muscle, Mobility and Aging (N=873) were 76.3±5.0 years old, 59.2% women, and 85.3% White. Higher muscle energetics (both in vivo and ex vivo ) were associated with lower perceived physical fatigability, all p<0.03. When stratified by sex, higher ATPmax was associated with lower PFS Physical for men only; higher max OXPHOS and max FAO OXPHOS were associated with lower RPE fatigability for both sexes. Higher skeletal muscle energetics were associated with 40-55% lower odds of being in the most (PFS≥25, RPE Fatigability≥12) vs least (PFS 0-4, RPE Fatigability 6-7) severe fatigability strata, all p<0.03. Being a woman was associated with 2-3 times higher odds of being in the most severe fatigability strata when controlling for ATPmax but not the in vivo measures (p<0.05). Better mitochondrial energetics were linked to lower fatigability and less severe fatigability in older adults. Findings imply that improving skeletal muscle energetics may mitigate perceived physical fatigability and prolong healthy aging.
Collapse
|
22
|
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
|
23
|
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
|
24
|
Mau T, Barnes HN, Blackwell TL, Kramer PA, Bauer SR, Marcinek DJ, Ramos SV, Forman DE, Toledo FGS, Hepple RT, Kritchevsky SB, Cummings SR, Newman AB, Coen PM, Cawthon PM. Lower muscle mitochondrial energetics is associated with greater phenotypic frailty in older women and men: the Study of Muscle, Mobility and Aging. GeroScience 2024; 46:2409-2424. [PMID: 37987886 PMCID: PMC10828481 DOI: 10.1007/s11357-023-01002-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Accepted: 10/29/2023] [Indexed: 11/22/2023] Open
Abstract
BACKGROUND Phenotypic frailty syndrome identifies older adults at greater risk for adverse health outcomes. Despite the critical role of mitochondria in maintaining cellular function, including energy production, the associations between muscle mitochondrial energetics and frailty have not been widely explored in a large, well-phenotyped, older population. METHODS The Study of Muscle, Mobility and Aging (SOMMA) assessed muscle energetics in older adults (N = 879, mean age = 76.3 years, 59.2% women). 31Phosporous magnetic resonance spectroscopy measured maximal production of adenosine triphosphate (ATPmax) in vivo, while ex vivo high-resolution respirometry of permeabilized muscle fibers from the vastus lateralis measured maximal oxygen consumption supported by fatty acids and complex I- and II-linked carbohydrates (e.g., Max OXPHOSCI+CII). Five frailty criteria, shrinking, weakness, exhaustion, slowness, and low activity, were used to classify participants as robust (0, N = 397), intermediate (1-2, N = 410), or frail (≥ 3, N = 66). We estimated the proportional odds ratio (POR) for greater frailty, adjusted for multiple potential confounders. RESULTS One-SD decrements of most respirometry measures (e.g., Max OXPHOSCI+CII, adjusted POR = 1.5, 95%CI [1.2,1.8], p = 0.0001) were significantly associated with greater frailty classification. The associations of ATPmax with frailty were weaker than those between Max OXPHOSCI+CII and frailty. Muscle energetics was most strongly associated with slowness and low physical activity components. CONCLUSIONS Our data suggest that deficits in muscle mitochondrial energetics may be a biological driver of frailty in older adults. On the other hand, we did observe differential relationships between measures of muscle mitochondrial energetics and the individual components of frailty.
Collapse
Affiliation(s)
- Theresa Mau
- San Francisco Coordinating Center, California Pacific Medical Center Research Institute, San Francisco, CA, USA.
- Department of Epidemiology and Biostatistics, University of California, San Francisco, CA, USA.
| | - Haley N Barnes
- San Francisco Coordinating Center, California Pacific Medical Center Research Institute, San Francisco, CA, USA
| | - Terri L Blackwell
- San Francisco Coordinating Center, California Pacific Medical Center Research Institute, San Francisco, CA, USA
| | - Philip A Kramer
- Department of Internal Medicine-Gerontology and Geriatric Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Scott R Bauer
- Department of Epidemiology and Biostatistics, University of California, San Francisco, CA, USA
- Department of Medicine and Urology, University of California, San Francisco, CA, USA
- Division of General Internal Medicine, San Francisco VA Healthcare System, San Francisco, CA, USA
| | - David J Marcinek
- Department of Radiology, University of Washington, Seattle, WA, USA
| | - Sofhia V Ramos
- AdventHealth, Translational Research Institute, Orlando, FL, USA
| | - Daniel E Forman
- Department of Medicine-Division of Geriatrics and Cardiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Geriatrics Research, Education, and Clinical Care (GRECC), VA Pittsburgh Healthcare System, Pittsburgh, PA, USA
| | - Frederico G S Toledo
- Department of Medicine-Division of Endocrinology and Metabolism, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Russell T Hepple
- Department of Physical Therapy, Department of Physiology and Aging, University of Florida, Gainesville, FL, USA
| | - Stephen B Kritchevsky
- Department of Internal Medicine-Gerontology and Geriatric Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Steven R Cummings
- San Francisco Coordinating Center, California Pacific Medical Center Research Institute, San Francisco, CA, USA
- Department of Epidemiology and Biostatistics, University of California, San Francisco, CA, USA
| | - Anne B Newman
- Department of Epidemiology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Paul M Coen
- AdventHealth, Translational Research Institute, Orlando, FL, USA
| | - Peggy M Cawthon
- San Francisco Coordinating Center, California Pacific Medical Center Research Institute, San Francisco, CA, USA
- Department of Epidemiology and Biostatistics, University of California, San Francisco, CA, USA
| |
Collapse
|
25
|
Kramer PA, Coen PM, Cawthon PM, Distefano G, Cummings SR, Goodpaster BH, Hepple RT, Kritchevsky SB, Shankland EG, Marcinek DJ, Toledo FGS, Duchowny KA, Ramos SV, Harrison S, Newman AB, Molina AJA. Skeletal Muscle Energetics Explain the Sex Disparity in Mobility Impairment in the Study of Muscle, Mobility and Aging. J Gerontol A Biol Sci Med Sci 2024; 79:glad283. [PMID: 38150179 PMCID: PMC10960628 DOI: 10.1093/gerona/glad283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Indexed: 12/28/2023] Open
Abstract
The age-related decline in muscle mitochondrial energetics contributes to the loss of mobility in older adults. Women experience a higher prevalence of mobility impairment compared to men, but it is unknown whether sex-specific differences in muscle energetics underlie this disparity. In the Study of Muscle, Mobility and Aging (SOMMA), muscle energetics were characterized using in vivo phosphorus-31 magnetic resonance spectroscopy and high-resolution respirometry of vastus lateralis biopsies in 773 participants (56.4% women, age 70-94 years). A Short Physical Performance Battery (SPPB) score ≤8 was used to define lower-extremity mobility impairment. Muscle mitochondrial energetics were lower in women compared to men (eg, Maximal Complex I&II OXPHOS: Women = 55.06 ± 15.95; Men = 65.80 ± 19.74; p < .001) and in individuals with mobility impairment compared to those without (eg, Maximal Complex I&II OXPHOS in women: SPPB ≥ 9 = 56.59 ± 16.22; SPPB ≤ 8 = 47.37 ± 11.85; p < .001). Muscle energetics were negatively associated with age only in men (eg, Maximal ETS capacity: R = -0.15, p = .02; age/sex interaction, p = .04), resulting in muscle energetics measures that were significantly lower in women than men in the 70-79 age group but not the 80+ age group. Similarly, the odds of mobility impairment were greater in women than men only in the 70-79 age group (70-79 age group, odds ratio [OR]age-adjusted = 1.78, 95% confidence interval [CI] = 1.03, 3.08, p = .038; 80+ age group, ORage-adjusted = 1.05, 95% CI = 0.52, 2.15, p = .89). Accounting for muscle energetics attenuated up to 75% of the greater odds of mobility impairment in women. Women had lower muscle mitochondrial energetics compared to men, which largely explain their greater odds of lower-extremity mobility impairment.
Collapse
Affiliation(s)
- Philip A Kramer
- Department of Internal Medicine-Gerontology and Geriatric Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
| | - Paul M Coen
- AdventHealth, Translational Research Institute, Orlando, Florida, USA
| | - Peggy M Cawthon
- Department of Epidemiology and Biostatistics, University of California San Francisco, San Francisco, California, USA
- San Francisco Coordinating Center, California Pacific Medical Center Research Institute, San Francisco, California, USA
| | | | - Steven R Cummings
- Department of Epidemiology and Biostatistics, University of California San Francisco, San Francisco, California, USA
- San Francisco Coordinating Center, California Pacific Medical Center Research Institute, San Francisco, California, USA
| | - Bret H Goodpaster
- AdventHealth, Translational Research Institute, Orlando, Florida, USA
| | - Russell T Hepple
- Department of Physical Therapy, University of Florida, Gainesville, Florida, USA
| | - Stephen B Kritchevsky
- Department of Internal Medicine-Gerontology and Geriatric Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
| | - Eric G Shankland
- Department of Radiology, University of Washington, Seattle, Washington, USA
| | - David J Marcinek
- Department of Radiology, University of Washington, Seattle, Washington, USA
| | - Frederico G S Toledo
- Department of Medicine-Division of Endocrinology and Metabolism, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Kate A Duchowny
- Institute for Social Research, University of Michigan, Ann Arbor, Michigan, USA
| | - Sofhia V Ramos
- AdventHealth, Translational Research Institute, Orlando, Florida, USA
| | - Stephanie Harrison
- San Francisco Coordinating Center, California Pacific Medical Center Research Institute, San Francisco, California, USA
| | - Anne B Newman
- Department of Epidemiology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Anthony J A Molina
- Department of Medicine-Division of Geriatrics, Gerontology, and Palliative Care, University of California San Diego School of Medicine, La Jolla, California, USA
| |
Collapse
|
26
|
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
|
27
|
Duchowny KA, Marcinek DJ, Mau T, Diaz-Ramierz LG, Lui LY, Toledo FGS, Cawthon PM, Hepple RT, Kramer PA, Newman AB, Kritchevsky SB, Cummings SR, Coen PM, Molina AJA. Childhood adverse life events and skeletal muscle mitochondrial function. SCIENCE ADVANCES 2024; 10:eadj6411. [PMID: 38446898 PMCID: PMC10917337 DOI: 10.1126/sciadv.adj6411] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Accepted: 02/01/2024] [Indexed: 03/08/2024]
Abstract
Social stress experienced in childhood is associated with adverse health later in life. Mitochondrial function has been implicated as a mechanism for how stressful life events "get under the skin" to influence physical well-being. Using data from the Study of Muscle, Mobility, and Aging (n = 879, 59% women), linear models examined whether adverse childhood events (i.e., physical abuse) were associated with two measures of skeletal muscle mitochondrial energetics in older adults: (i) maximal adenosine triphosphate production (ATPmax) and (ii) maximal state 3 respiration (Max OXPHOS). Forty-five percent of the sample reported experiencing one or more adverse childhood events. After adjustment, each additional event was associated with -0.08 SD (95% confidence interval = -0.13, -0.02) lower ATPmax. No association was observed with Max OXPHOS. Adverse childhood events are associated with lower ATP production in later life. Findings indicate that mitochondrial function may be a mechanism for understanding how early social stress influences health in later life.
Collapse
Affiliation(s)
- Kate A. Duchowny
- Institute for Social Research, University of Michigan, Ann Arbor, MI, USA
| | | | - Theresa Mau
- California Pacific Medical Center Research Institute, San Francisco, CA, USA
- Department of Epidemiology and Biostatistics, University of California, San Francisco, CA, USA
| | - L. Grisell Diaz-Ramierz
- Division of Geriatrics, Department of Medicine, UCSF School of Medicine, San Francisco, CA, USA
| | - Li-Yung Lui
- California Pacific Medical Center Research Institute, San Francisco, CA, USA
| | - Frederico G. S. Toledo
- Department of Medicine, Division of Endocrinology and Metabolism, University of Pittsburgh, Pittsburgh, PA, USA
| | - Peggy M. Cawthon
- California Pacific Medical Center Research Institute, San Francisco, CA, USA
- Department of Epidemiology and Biostatistics, University of California, San Francisco, CA, USA
| | - Russell T. Hepple
- Department of Physical Therapy, University of Florida, Gainesville, FL, USA
| | - Philip A. Kramer
- Department of Internal Medicine-Gerontology and Geriatric Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Anne B. Newman
- Department of Epidemiology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Stephen B. Kritchevsky
- Department of Internal Medicine-Gerontology and Geriatric Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Steven R. Cummings
- California Pacific Medical Center Research Institute, San Francisco, CA, USA
- Department of Epidemiology and Biostatistics, University of California, San Francisco, CA, USA
| | - Paul M. Coen
- AdventHealth, Translational Research Institute, Orlando, FL, USA
| | - Anthony J. A. Molina
- Department of Medicine-Division of Geriatrics, Gerontology, and Palliative Care, University of California San Diego School of Medicine, La Jolla, CA, USA
| |
Collapse
|
28
|
Affourtit C, Carré JE. Mitochondrial involvement in sarcopenia. Acta Physiol (Oxf) 2024; 240:e14107. [PMID: 38304924 DOI: 10.1111/apha.14107] [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: 11/30/2023] [Revised: 01/11/2024] [Accepted: 01/22/2024] [Indexed: 02/03/2024]
Abstract
Sarcopenia lowers the quality-of-life for millions of people across the world, as accelerated loss of skeletal muscle mass and function contributes to both age- and disease-related frailty. Physical activity remains the only proven therapy for sarcopenia to date, but alternatives are much sought after to manage this progressive muscle disorder in individuals who are unable to exercise. Mitochondria have been widely implicated in the etiology of sarcopenia and are increasingly suggested as attractive therapeutic targets to help restore the perturbed balance between protein synthesis and breakdown that underpins skeletal muscle atrophy. Reviewing current literature, we note that mitochondrial bioenergetic changes in sarcopenia are generally interpreted as intrinsic dysfunction that renders muscle cells incapable of making sufficient ATP to fuel protein synthesis. Based on the reported mitochondrial effects of therapeutic interventions, however, we argue that the observed bioenergetic changes may instead reflect an adaptation to pathologically decreased energy expenditure in sarcopenic muscle. Discrimination between these mechanistic possibilities will be crucial for improving the management of sarcopenia.
Collapse
Affiliation(s)
| | - Jane E Carré
- School of Biomedical Sciences, University of Plymouth, Plymouth, UK
| |
Collapse
|
29
|
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
|
30
|
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
|
31
|
Tian Q, Lee PR, Yang Q, Moore AZ, Landman BA, Resnick SM, Ferrucci L. The mediation roles of intermuscular fat and inflammation in muscle mitochondrial associations with cognition and mobility. J Cachexia Sarcopenia Muscle 2024; 15:138-148. [PMID: 38116708 PMCID: PMC10834332 DOI: 10.1002/jcsm.13413] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 10/24/2023] [Accepted: 11/20/2023] [Indexed: 12/21/2023] Open
Abstract
BACKGROUND Mitochondrial dysfunction may contribute to brain and muscle health through inflammation or fat infiltration in the muscle, both of which are associated with cognitive function and mobility. We aimed to examine the association between skeletal muscle mitochondrial function and cognitive and mobility outcomes and tested the mediation effect of inflammation or fat infiltration. METHODS We analysed data from 596 Baltimore Longitudinal Study of Aging participants who had concurrent data on skeletal muscle oxidative capacity and cognitive and mobility measures of interest (mean age: 66.1, 55% women, 24% Black). Skeletal muscle oxidative capacity was assessed as post-exercise recovery rate (kPCr) via P31 MR spectroscopy. Fat infiltration was measured as intermuscular fat (IMF) via CT scan and was available for 541 participants. Inflammation markers [IL-6, C-reactive protein (CRP), total white blood cell (WBC), neutrophil count, erythrocyte sedimentation rate (ESR), or albumin] were available in 594 participants. We examined the association of kPCr and cognitive and mobility measures using linear regression and tested the mediation effect of IMF or inflammation using the mediation package in R. Models were adjusted for demographics and PCr depletion. RESULTS kPCr and IMF were both significantly associated with specific cognitive domains (DSST, TMA-A, and pegboard dominant hand performance) and mobility (usual gait speed, HABCPPB, 400 m walk time) (all P < 0.05). IMF significantly mediated the relationship between kPCr and these cognitive and mobility measures (all P < 0.05, proportion mediated 13.1% to 27%). Total WBC, neutrophil count, and ESR, but not IL-6 or CRP, also mediated at least one of the cognitive and mobility outcomes (all P < 0.05, proportion mediated 9.4% to 15.3%). CONCLUSIONS Skeletal muscle mitochondrial function is associated with cognitive performance involving psychomotor speed. Muscle fat infiltration and specific inflammation markers mediate the relationship between muscle mitochondrial function and cognitive and mobility outcomes. Future studies are needed to confirm these associations longitudinally and to understand their mechanistic underpinnings.
Collapse
Affiliation(s)
- Qu Tian
- Longitudinal Studies Section, Translational Gerontology Branch, National Institute on Aging, Baltimore, MD, USA
| | - Philip R Lee
- Longitudinal Studies Section, Translational Gerontology Branch, National Institute on Aging, Baltimore, MD, USA
| | - Qi Yang
- Department of Computer Science, Vanderbilt University, Nashville, TN, USA
| | - Anne Z Moore
- Longitudinal Studies Section, Translational Gerontology Branch, National Institute on Aging, Baltimore, MD, 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
- 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
|
32
|
Brisendine MH, Nichenko AS, Bandara AB, Willoughby OS, Amiri N, Weingrad Z, Specht KS, Bond JM, Addington A, Jones RG, Murach KA, Poelzing S, Craige SM, Grange RW, Drake JC. Neuromuscular Dysfunction Precedes Cognitive Impairment in a Mouse Model of Alzheimer's Disease. FUNCTION 2023; 5:zqad066. [PMID: 38111538 PMCID: PMC10727840 DOI: 10.1093/function/zqad066] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 11/17/2023] [Accepted: 11/26/2023] [Indexed: 12/20/2023] Open
Abstract
Alzheimer's disease (AD) develops along a continuum that spans years prior to diagnosis. Decreased muscle function and mitochondrial respiration occur years earlier in those that develop AD; however, it is unknown what causes these peripheral phenotypes in a disease of the brain. Exercise promotes muscle, mitochondria, and cognitive health and is proposed to be a potential therapeutic for AD, but no study has investigated how skeletal muscle adapts to exercise training in an AD-like context. Utilizing 5xFAD mice, an AD model that develops ad-like pathology and cognitive impairments around 6 mo of age, we examined in vivo neuromuscular function and exercise adapations (mitochondrial respiration and RNA sequencing) before the manifestation of overt cognitive impairment. We found 5xFAD mice develop neuromuscular dysfunction beginning as early as 4 mo of age, characterized by impaired nerve-stimulated muscle torque production and compound nerve action potential of the sciatic nerve. Furthermore, skeletal muscle in 5xFAD mice had altered, sex-dependent, adaptive responses (mitochondrial respiration and gene expression) to exercise training in the absence of overt cognitive impairment. Changes in peripheral systems, specifically neural communication to skeletal muscle, may be harbingers for AD and have implications for lifestyle interventions, like exercise, in AD.
Collapse
Affiliation(s)
- Matthew H Brisendine
- Department of Human Nutrition, Foods, and Exercise, Virginia Tech, Blacksburg, VA 24061, USA
| | - Anna S Nichenko
- Department of Human Nutrition, Foods, and Exercise, Virginia Tech, Blacksburg, VA 24061, USA
| | - Aloka B Bandara
- Department of Human Nutrition, Foods, and Exercise, Virginia Tech, Blacksburg, VA 24061, USA
| | - Orion S Willoughby
- Department of Human Nutrition, Foods, and Exercise, Virginia Tech, Blacksburg, VA 24061, USA
| | - Niloufar Amiri
- Department of Human Nutrition, Foods, and Exercise, Virginia Tech, Blacksburg, VA 24061, USA
| | - Zach Weingrad
- Department of Biological Sciences, Virginia Tech, Blacksburg, VA 24061, USA
| | - Kalyn S Specht
- Department of Human Nutrition, Foods, and Exercise, Virginia Tech, Blacksburg, VA 24061, USA
| | - Jacob M Bond
- Translational Biology, Medicine, and Health Program, Virginia Tech, Roanoke, VA 24016, USA
| | - Adele Addington
- Department of Human Nutrition, Foods, and Exercise, Virginia Tech, Blacksburg, VA 24061, USA
| | - Ronald G Jones
- Department of Health, Human Performance, and Recreation, University of Arkansas, Fayetteville, AR 72701, USA
| | - Kevin A Murach
- Department of Health, Human Performance, and Recreation, University of Arkansas, Fayetteville, AR 72701, USA
| | - Steven Poelzing
- Translational Biology, Medicine, and Health Program, Virginia Tech, Roanoke, VA 24016, USA
| | - Siobhan M Craige
- Department of Human Nutrition, Foods, and Exercise, Virginia Tech, Blacksburg, VA 24061, USA
- Translational Biology, Medicine, and Health Program, Virginia Tech, Roanoke, VA 24016, USA
| | - Robert W Grange
- Department of Human Nutrition, Foods, and Exercise, Virginia Tech, Blacksburg, VA 24061, USA
| | - Joshua C Drake
- Department of Human Nutrition, Foods, and Exercise, Virginia Tech, Blacksburg, VA 24061, USA
- Translational Biology, Medicine, and Health Program, Virginia Tech, Roanoke, VA 24016, USA
| |
Collapse
|
33
|
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
|
34
|
Granic A, Suetterlin K, Shavlakadze T, Grounds M, Sayer A. Hallmarks of ageing in human skeletal muscle and implications for understanding the pathophysiology of sarcopenia in women and men. Clin Sci (Lond) 2023; 137:1721-1751. [PMID: 37986616 PMCID: PMC10665130 DOI: 10.1042/cs20230319] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 11/01/2023] [Accepted: 11/14/2023] [Indexed: 11/22/2023]
Abstract
Ageing is a complex biological process associated with increased morbidity and mortality. Nine classic, interdependent hallmarks of ageing have been proposed involving genetic and biochemical pathways that collectively influence ageing trajectories and susceptibility to pathology in humans. Ageing skeletal muscle undergoes profound morphological and physiological changes associated with loss of strength, mass, and function, a condition known as sarcopenia. The aetiology of sarcopenia is complex and whilst research in this area is growing rapidly, there is a relative paucity of human studies, particularly in older women. Here, we evaluate how the nine classic hallmarks of ageing: genomic instability, telomere attrition, epigenetic alterations, loss of proteostasis, deregulated nutrient sensing, mitochondrial dysfunction, cellular senescence, stem cell exhaustion, and altered intercellular communication contribute to skeletal muscle ageing and the pathophysiology of sarcopenia. We also highlight five novel hallmarks of particular significance to skeletal muscle ageing: inflammation, neural dysfunction, extracellular matrix dysfunction, reduced vascular perfusion, and ionic dyshomeostasis, and discuss how the classic and novel hallmarks are interconnected. Their clinical relevance and translational potential are also considered.
Collapse
Affiliation(s)
- Antoneta Granic
- AGE Research Group, Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, U.K
- NIHR Newcastle Biomedical Research Centre, Newcastle University and Newcastle Upon Tyne Hospitals NHS Foundation Trust, Newcastle Upon Tyne, U.K
| | - Karen Suetterlin
- AGE Research Group, Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, U.K
- NIHR Newcastle Biomedical Research Centre, Newcastle University and Newcastle Upon Tyne Hospitals NHS Foundation Trust, Newcastle Upon Tyne, U.K
- John Walton Muscular Dystrophy Research Centre, Institute of Genetic Medicine, Newcastle University, Centre for Life, Newcastle upon Tyne, U.K
| | - Tea Shavlakadze
- Regeneron Pharmaceuticals Inc., Tarrytown, New York, NY, U.S.A
| | - Miranda D. Grounds
- Department of Anatomy, Physiology and Human Biology, School of Human Sciences, the University of Western Australia, Perth, WA 6009, Australia
| | - Avan A. Sayer
- AGE Research Group, Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, U.K
- NIHR Newcastle Biomedical Research Centre, Newcastle University and Newcastle Upon Tyne Hospitals NHS Foundation Trust, Newcastle Upon Tyne, U.K
| |
Collapse
|
35
|
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
|
36
|
Kramer PA, Coen PM, Cawthon PM, Distefano G, Cummings SR, Goodpaster BH, Hepple RT, Kritchevsky SB, Shankland EG, Marcinek DJ, Toledo FGS, Duchowny KA, Ramos SV, Harrison S, Newman AB, Molina AJA. Skeletal muscle energetics explain the sex disparity in mobility impairment in the Study of Muscle, Mobility and Aging (SOMMA). MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.11.08.23298271. [PMID: 37987007 PMCID: PMC10659490 DOI: 10.1101/2023.11.08.23298271] [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
The age-related decline in muscle mitochondrial energetics contributes to the loss of mobility in older adults. Women experience a higher prevalence of mobility impairment compared to men, but it is unknown whether sex-specific differences in muscle energetics underlie this disparity. In the Study of Muscle, Mobility and Aging (SOMMA), muscle energetics were characterized using in vivo phosphorus-31 magnetic resonance spectroscopy and high-resolution respirometry of vastus lateralis biopsies in 773 participants (56.4% women, age 70-94 years). A Short Physical Performance Battery score ≤ 8 was used to define lower-extremity mobility impairment. Muscle mitochondrial energetics were lower in women compared to men (e.g. Maximal Complex I&II OXPHOS: Women=55.06 +/- 15.95; Men=65.80 +/- 19.74; p<0.001) and in individuals with mobility impairment compared to those without (e.g., Maximal Complex I&II OXPHOS in women: SPPB≥9=56.59 +/- 16.22; SPPB≤8=47.37 +/- 11.85; p<0.001). Muscle energetics were negatively associated with age only in men (e.g., Maximal ETS capacity: R=-0.15, p=0.02; age/sex interaction, p=0.04), resulting in muscle energetics measures that were significantly lower in women than men in the 70-79 age group but not the 80+ age group. Similarly, the odds of mobility impairment were greater in women than men only in the 70-79 age group (70-79 age group, OR age-adjusted =1.78, 95% CI=1.03, 3.08, p=0.038; 80+ age group, OR age-adjusted =1.05, 95% CI=0.52, 2.15, p=0.89). Accounting for muscle energetics attenuated up to 75% of the greater odds of mobility impairment in women. Women had lower muscle mitochondrial energetics compared to men, which largely explain their greater odds of lower-extremity mobility impairment.
Collapse
|
37
|
Mau T, Blackwell TL, Cawthon PM, Molina AJA, Coen PM, Distefano G, Kramer PA, Ramos SV, Forman DE, Goodpaster BH, Toledo FGS, Duchowny KA, Sparks LM, Newman AB, Kritchevsky SB, Cummings SR. Muscle mitochondrial bioenergetic capacities is associated with multimorbidity burden in older adults: the Study of Muscle, Mobility and Aging (SOMMA). MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.11.06.23298175. [PMID: 39711735 PMCID: PMC11661392 DOI: 10.1101/2023.11.06.23298175] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2024]
Abstract
Background The geroscience hypothesis posits that aging biological processes contribute to many age-related deficits, including the accumulation of multiple chronic diseases. Though only one facet of mitochondrial function, declines in muscle mitochondrial bioenergetic capacities may contribute to this increased susceptibility to multimorbidity. Methods The Study of Muscle, Mobility and Aging (SOMMA) assessed ex vivo muscle mitochondrial energetics in 764 older adults (mean age =76.4, 56.5% women, 85.9% non-Hispanic white) by high-resolution respirometry of permeabilized muscle fibers. We estimated the proportional odds ratio (POR [95%CI]) for the likelihood of greater multimorbidity (four levels: 0 conditions, N=332; 1 condition, N=299; 2 conditions, N=98; or 3+ conditions, N=35) from an index of 11 conditions, per SD decrement in muscle mitochondrial energetic parameters. Distribution of conditions allowed for testing the associations of maximal muscle energetics with some individual conditions. Results Lower oxidative phosphorylation supported by fatty acids and/or complex-I and -II linked carbohydrates (e.g., Max OXPHOSCI+CII) was associated with a greater multimorbidity index score (POR=1.32[1.13,1.54]) and separately with diabetes mellitus (OR=1.62[1.26,2.09]), depressive symptoms (OR=1.45[1.04,2.00]) and possibly chronic kidney disease (OR=1.57[0.98,2.52]) but not significantly with other conditions (e.g., cardiac arrhythmia, chronic obstructive pulmonary disease). Conclusions Lower muscle mitochondrial bioenergetic capacities was associated with a worse composite multimorbidity index score. Our results suggest that decrements in muscle mitochondrial energetics may contribute to a greater global burden of disease and is more strongly related to some conditions than others.
Collapse
Affiliation(s)
- Theresa Mau
- San Francisco Coordinating Center, California Pacific Medical Center Research Institute, San Francisco, California
- Department of Epidemiology and Biostatistics, University of California, San Francisco, California
| | - Terri L Blackwell
- San Francisco Coordinating Center, California Pacific Medical Center Research Institute, San Francisco, California
| | - Peggy M Cawthon
- San Francisco Coordinating Center, California Pacific Medical Center Research Institute, San Francisco, California
- Department of Epidemiology and Biostatistics, University of California, San Francisco, California
| | - Anthony J A Molina
- Department of Medicine-Division of Geriatrics, Gerontology, and Palliative Care, University of California San Diego School of Medicine, La Jolla, California
| | - Paul M Coen
- Translational Research Institute, AdventHealth, Orlando, Florida
| | | | - Philip A Kramer
- Department of Internal Medicine-Gerontology and Geriatric Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina
| | - Sofhia V Ramos
- Translational Research Institute, AdventHealth, Orlando, Florida
| | - Daniel E Forman
- Department of Medicine-Division of Geriatrics and Cardiology, University of Pittsburgh, Geriatrics Research, Education, and Clinical Care (GRECC), VA Pittsburgh Healthcare System, Pittsburgh, Pennsylvania
| | | | - Frederico G S Toledo
- Department of Medicine-Division of Endocrinology and Metabolism, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Kate A Duchowny
- Social Environment and Health, Survey Research Center, Institute for Social Research, University of Michigan, Ann Arbor, Michigan
| | - Lauren M Sparks
- Translational Research Institute, AdventHealth, Orlando, Florida
| | - Anne B Newman
- Department of Epidemiology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Stephen B Kritchevsky
- Department of Internal Medicine-Gerontology and Geriatric Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina
| | - Steven R Cummings
- San Francisco Coordinating Center, California Pacific Medical Center Research Institute, San Francisco, California
- Department of Epidemiology and Biostatistics, University of California, San Francisco, California
| |
Collapse
|
38
|
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
|
39
|
Smith JAB, Murach KA, Dyar KA, Zierath JR. Exercise metabolism and adaptation in skeletal muscle. Nat Rev Mol Cell Biol 2023; 24:607-632. [PMID: 37225892 PMCID: PMC10527431 DOI: 10.1038/s41580-023-00606-x] [Citation(s) in RCA: 98] [Impact Index Per Article: 49.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/30/2023] [Indexed: 05/26/2023]
Abstract
Viewing metabolism through the lens of exercise biology has proven an accessible and practical strategy to gain new insights into local and systemic metabolic regulation. Recent methodological developments have advanced understanding of the central role of skeletal muscle in many exercise-associated health benefits and have uncovered the molecular underpinnings driving adaptive responses to training regimens. In this Review, we provide a contemporary view of the metabolic flexibility and functional plasticity of skeletal muscle in response to exercise. First, we provide background on the macrostructure and ultrastructure of skeletal muscle fibres, highlighting the current understanding of sarcomeric networks and mitochondrial subpopulations. Next, we discuss acute exercise skeletal muscle metabolism and the signalling, transcriptional and epigenetic regulation of adaptations to exercise training. We address knowledge gaps throughout and propose future directions for the field. This Review contextualizes recent research of skeletal muscle exercise metabolism, framing further advances and translation into practice.
Collapse
Affiliation(s)
- Jonathon A B Smith
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Kevin A Murach
- Molecular Mass Regulation Laboratory, Exercise Science Research Center, Department of Health, Human Performance and Recreation, University of Arkansas, Fayetteville, AR, USA
| | - Kenneth A Dyar
- Metabolic Physiology, Institute for Diabetes and Cancer, Helmholtz Diabetes Center, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Juleen R Zierath
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden.
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden.
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
40
|
Campbell MD, Samuelson AT, Chiao YA, Sweetwyne MT, Ladiges WC, Rabinovitch PS, Marcinek DJ. Intermittent treatment with elamipretide preserves exercise tolerance in aged female mice. GeroScience 2023; 45:2245-2255. [PMID: 36840897 PMCID: PMC10651577 DOI: 10.1007/s11357-023-00754-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Accepted: 02/09/2023] [Indexed: 02/26/2023] Open
Abstract
The pathology of aging impacts multiple organ systems, including the kidney and skeletal and cardiac muscles. Long-term treatment with the mitochondrial-targeted peptide elamipretide has previously been shown to improve in vivo mitochondrial function in aged mice, which is associated with increased fatigue resistance and treadmill performance, improved cardiovascular diastolic function, and glomerular architecture of the kidney. However, elamipretide is a short tetrameric peptide that is not orally bioavailable, limiting its routes of administration. This study tested whether twice weekly intermittent injections of elamipretide could recapitulate the same functional improvements as continuous long-term infusion. We found that intermittent treatment with elamipretide for 8 months preserved exercise tolerance and left ventricular mass in mice with modest protection of diastolic function and skeletal muscle force production but did not affect kidney function as previously reported using continuous treatment.
Collapse
Affiliation(s)
- Matthew D Campbell
- Department of Radiology, University of Washington, Seattle, WA, 98109, USA
| | - Ashton T Samuelson
- Department of Radiology, University of Washington, Seattle, WA, 98109, USA
- Oregon Health Sciences Surgical Residency Program, Portland, OR, 97239, USA
| | - Ying Ann Chiao
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
- Aging & Metabolism Research Program MS21, Oklahoma Medical Research Foundation, Oklahoma City, OK, 73104, USA
| | - Mariya T Sweetwyne
- Department of Laboratory Medicine, University of Washington, Seattle, WA, 98109, USA
| | - Warren C Ladiges
- Department of Comparative Medicine, University of Washington, Seattle, WA, 98109, USA
| | - Peter S Rabinovitch
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
| | - David J Marcinek
- Department of Radiology, University of Washington, Seattle, WA, 98109, USA.
- South Lake Union Campus, 850 Republican St., Brotman D142, Box 358050, Seattle, WA, 98109, USA.
| |
Collapse
|
41
|
Tian Q, Shardell MD, Kuo PL, Tanaka T, Simonsick EM, Moaddel R, Resnick SM, Ferrucci L. Plasma metabolomic signatures of dual decline in memory and gait in older adults. GeroScience 2023; 45:2659-2667. [PMID: 37052768 PMCID: PMC10651620 DOI: 10.1007/s11357-023-00792-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2023] [Accepted: 04/04/2023] [Indexed: 04/14/2023] Open
Abstract
Older adults experiencing dual decline in memory and gait have greater dementia risk than those with memory or gait decline only, but mechanisms are unknown. Dual decline may indicate specific pathophysiological pathways to dementia which can be reflected by circulating metabolites. We compared longitudinal changes in plasma metabolite biomarkers of older adults with and without dual decline in the Baltimore Longitudinal Study of Aging (BLSA). Participants were grouped into 4 phenotypes based on annual rates of decline in verbal memory and gait speed: no decline in memory or gait, memory decline only, gait decline only, and dual decline. Repeated measures of plasma metabolomics were measured by biocrates p500 kit during the same time of memory and gait assessments. In BLSA, 18 metabolites differed across groups (q-value < 0.05). Metabolites differentially abundant were enriched for lysophosphatidylcholines (lysoPC C18:0,C16:0,C17:0,C18:1,C18:2), ceramides (d18:2/24:0,d16:1/24:0,d16:1/23:0), and amino acids (glycine) classes. Compared to no decline, the dual decline group showed greater declines in lysoPC C18:0, homoarginine synthesis, and the metabolite module containing mostly triglycerides, and showed a greater increase in indoleamine 2,3-dioxygenase (IDO) activity. Metabolites distinguishing dual decline and no decline groups were implicated in metabolic pathways of the aminoacyl-tRNA biosynthesis, valine, leucine and isoleucine biosynthesis, histidine metabolism, and sphingolipid metabolism. Older adults with dual decline exhibit the most extensive alterations in metabolic profiling of lysoPCs, ceramides, IDO activity, and homoarginine synthesis. Alterations in these metabolites may indicate mitochondrial dysfunction, compromised immunity, and elevated burden of cardiovascular and kidney pathology.
Collapse
Affiliation(s)
- Qu Tian
- Translational Gerontology Branch, National Institute On Aging, 251 Bayview Blvd., Suite 100, Room 04B316, Baltimore, MD, 21224, USA.
| | | | - Pei-Lun Kuo
- Translational Gerontology Branch, National Institute On Aging, 251 Bayview Blvd., Suite 100, Room 04B316, Baltimore, MD, 21224, USA
| | - Toshiko Tanaka
- Translational Gerontology Branch, National Institute On Aging, 251 Bayview Blvd., Suite 100, Room 04B316, Baltimore, MD, 21224, USA
| | - Eleanor M Simonsick
- Translational Gerontology Branch, National Institute On Aging, 251 Bayview Blvd., Suite 100, Room 04B316, Baltimore, MD, 21224, USA
| | - Ruin Moaddel
- Laboratory of Clinical Investigation, National Institute on Aging, Baltimore, MD, USA
| | - Susan M Resnick
- Laboratory of Behavioral Neuroscience, National Institute on Aging, Baltimore, MD, USA
| | - Luigi Ferrucci
- Translational Gerontology Branch, National Institute On Aging, 251 Bayview Blvd., Suite 100, Room 04B316, Baltimore, MD, 21224, USA
| |
Collapse
|
42
|
Baechle JJ, Chen N, Makhijani P, Winer S, Furman D, Winer DA. Chronic inflammation and the hallmarks of aging. Mol Metab 2023; 74:101755. [PMID: 37329949 PMCID: PMC10359950 DOI: 10.1016/j.molmet.2023.101755] [Citation(s) in RCA: 122] [Impact Index Per Article: 61.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 05/30/2023] [Accepted: 06/13/2023] [Indexed: 06/19/2023] Open
Abstract
BACKGROUND Recently, the hallmarks of aging were updated to include dysbiosis, disabled macroautophagy, and chronic inflammation. In particular, the low-grade chronic inflammation during aging, without overt infection, is defined as "inflammaging," which is associated with increased morbidity and mortality in the aging population. Emerging evidence suggests a bidirectional and cyclical relationship between chronic inflammation and the development of age-related conditions, such as cardiovascular diseases, neurodegeneration, cancer, and frailty. How the crosstalk between chronic inflammation and other hallmarks of aging underlies biological mechanisms of aging and age-related disease is thus of particular interest to the current geroscience research. SCOPE OF REVIEW This review integrates the cellular and molecular mechanisms of age-associated chronic inflammation with the other eleven hallmarks of aging. Extra discussion is dedicated to the hallmark of "altered nutrient sensing," given the scope of Molecular Metabolism. The deregulation of hallmark processes during aging disrupts the delicate balance between pro-inflammatory and anti-inflammatory signaling, leading to a persistent inflammatory state. The resultant chronic inflammation, in turn, further aggravates the dysfunction of each hallmark, thereby driving the progression of aging and age-related diseases. MAIN CONCLUSIONS The crosstalk between chronic inflammation and other hallmarks of aging results in a vicious cycle that exacerbates the decline in cellular functions and promotes aging. Understanding this complex interplay will provide new insights into the mechanisms of aging and the development of potential anti-aging interventions. Given their interconnectedness and ability to accentuate the primary elements of aging, drivers of chronic inflammation may be an ideal target with high translational potential to address the pathological conditions associated with aging.
Collapse
Affiliation(s)
- Jordan J Baechle
- Buck Artificial Intelligence Platform, the Buck Institute for Research on Aging, Novato, CA, USA
| | - Nan Chen
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada; Division of Cellular & Molecular Biology, Diabetes Research Group, Toronto General Hospital Research Institute (TGHRI), University Health Network, Toronto, ON, Canada
| | - Priya Makhijani
- Buck Artificial Intelligence Platform, the Buck Institute for Research on Aging, Novato, CA, USA; Department of Immunology, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Shawn Winer
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - David Furman
- Buck Artificial Intelligence Platform, the Buck Institute for Research on Aging, Novato, CA, USA; Stanford 1000 Immunomes Project, Stanford University School of Medicine, Stanford, CA, USA; Instituto de Investigaciones en Medicina Traslacional (IIMT), Universidad Austral, CONICET, Pilar, Argentina.
| | - Daniel A Winer
- Buck Artificial Intelligence Platform, the Buck Institute for Research on Aging, Novato, CA, USA; Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada; Department of Immunology, Faculty of Medicine, University of Toronto, Toronto, ON, Canada; Division of Cellular & Molecular Biology, Diabetes Research Group, Toronto General Hospital Research Institute (TGHRI), University Health Network, Toronto, ON, Canada; Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA.
| |
Collapse
|
43
|
Tian Q, Ferrucci L. Beyond the relationship between mitochondria and mobility in aging. Aging (Albany NY) 2023; 15:4574-4575. [PMID: 37315298 PMCID: PMC10292870 DOI: 10.18632/aging.204649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Accepted: 04/04/2023] [Indexed: 06/16/2023]
Affiliation(s)
- Qu Tian
- Longitudinal Studies Section, Translational Gerontology Branch, National Institute on Aging, Baltimore, Maryland 21224, US
| | - Luigi Ferrucci
- Longitudinal Studies Section, Translational Gerontology Branch, National Institute on Aging, Baltimore, Maryland 21224, US
| |
Collapse
|
44
|
Turkel I, Ozerklig B, Yılmaz M, Ulger O, Kubat GB, Tuncer M. Mitochondrial transplantation as a possible therapeutic option for sarcopenia. J Mol Med (Berl) 2023:10.1007/s00109-023-02326-3. [PMID: 37209146 DOI: 10.1007/s00109-023-02326-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 04/09/2023] [Accepted: 04/20/2023] [Indexed: 05/22/2023]
Abstract
With advancing age, the skeletal muscle phenotype is characterized by a progressive loss of mass, strength, and quality. This phenomenon, known as sarcopenia, has a negative impact on quality of life and increases the risk of morbidity and mortality in older adults. Accumulating evidence suggests that damaged and dysfunctional mitochondria play a critical role in the pathogenesis of sarcopenia. Lifestyle modifications, such as physical activity, exercise, and nutrition, as well as medical interventions with therapeutic agents, are effective in the management of sarcopenia and offer solutions to maintain and improve skeletal muscle health. Although a great deal of effort has been devoted to the identification of the best treatment option, these strategies are not sufficient to overcome sarcopenia. Recently, it has been reported that mitochondrial transplantation may be a possible therapeutic approach for the treatment of mitochondria-related pathological conditions such as ischemia, liver toxicity, kidney injury, cancer, and non-alcoholic fatty liver disease. Given the role of mitochondria in the function and metabolism of skeletal muscle, mitochondrial transplantation may be a possible option for the treatment of sarcopenia. In this review, we summarize the definition and characteristics of sarcopenia and molecular mechanisms associated with mitochondria that are known to contribute to sarcopenia. We also discuss mitochondrial transplantation as a possible option. Despite the progress made in the field of mitochondrial transplantation, further studies are needed to elucidate the role of mitochondrial transplantation in sarcopenia. KEY MESSAGES: Sarcopenia is the progressive loss of skeletal muscle mass, strength, and quality. Although the specific mechanisms that lead to sarcopenia are not fully understood, mitochondria have been identified as a key factor in the development of sarcopenia. Damaged and dysfunctional mitochondria initiate various cellular mediators and signaling pathways, which largely contribute to the age-related loss of skeletal muscle mass and strength. Mitochondrial transplantation has been reported to be a possible option for the treatment/prevention of several diseases. Mitochondrial transplantation may be a possible therapeutic option for improving skeletal muscle health and treating sarcopenia. Mitochondrial transplantation as a possible treatment option for sarcopenia.
Collapse
Affiliation(s)
- Ibrahim Turkel
- Department of Exercise and Sport Sciences, Faculty of Sport Sciences, Hacettepe University, Ankara, Turkey
- Division of Sport Sciences and Technology, Institute of Health Sciences, Hacettepe University, Ankara, Turkey
| | - Berkay Ozerklig
- Department of Exercise and Sport Sciences, Faculty of Sport Sciences, Hacettepe University, Ankara, Turkey
- Division of Sport Sciences and Technology, Institute of Health Sciences, Hacettepe University, Ankara, Turkey
| | - Merve Yılmaz
- Department of Medical Biochemistry, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| | - Oner Ulger
- Department of Mitochondria and Cellular Research, Health Sciences Institute, Health Sciences University, Ankara, Turkey
| | - Gokhan Burcin Kubat
- Division of Sport Sciences and Technology, Institute of Health Sciences, Hacettepe University, Ankara, Turkey.
- Department of Mitochondria and Cellular Research, Health Sciences Institute, Health Sciences University, Ankara, Turkey.
| | - Meltem Tuncer
- Department of Physiology, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| |
Collapse
|
45
|
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
|
46
|
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
|
47
|
Tian Q, An Y, Kitner-Triolo MH, Davatzikos C, Studenski SA, Ferrucci L, Resnick SM. Associations of Olfaction With Longitudinal Trajectories of Brain Volumes and Neuropsychological Function in Older Adults. Neurology 2023; 100:e964-e974. [PMID: 36460474 PMCID: PMC9990434 DOI: 10.1212/wnl.0000000000201646] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Accepted: 10/18/2022] [Indexed: 12/04/2022] Open
Abstract
BACKGROUND AND OBJECTIVES Olfactory function declines with aging, and olfactory deficits are one of the earliest features of neurodegenerative diseases, such as Parkinson disease and Alzheimer disease. Previous studies have shown that olfaction is associated with brain volumes and cognitive function, but data are exclusively cross-sectional. We aimed to examine longitudinal associations of olfaction with changes in brain volumes and neuropsychological function. METHODS In the Baltimore Longitudinal Study of Aging, we chose the first assessment of olfaction to examine the associations with retrospective and prospective changes in neuropsychological performance and brain volumes in participants aged 50 years or older using linear mixed-effects models, adjusted for demographic variables and cardiovascular disease. Olfaction was measured as odor identification scores through the 16-item Sniffin' Sticks. RESULTS We analyzed data from 567 (58% women, 42% men, 27% Black, 66% White, and 7% others) participants who had data on odor identification scores and brain volumetric MRI (n = 420 with retrospective repeats over a mean of 3.7 years, n = 280 with prospective repeats over a mean of 1.2 years). We also analyzed data from 754 participants (56% women, 44% men, 29% Black, 65% White, and 6% others) with neuropsychological assessments (n = 630 with retrospective repeats over a mean of 6.6 years, n = 280 with prospective repeats over a mean of 1.5 years). After adjustment, higher odor identification scores were associated with prior and subsequent slower brain atrophy in the entorhinal cortex (β ± SE = 0.0093 ± 0.0031, p = 0.0028 and β ± SE = 0.0176 ± 0.0073, p = 0.0169, respectively), hippocampus (β ± SE = 0.0070 ± 0.0030, p = 0.0192 and β ± SE = 0.0173 ± 0.0066, p = 0.0089, respectively), and additional frontal and temporal areas (all p < 0.05). Higher odor identification scores were also associated with prior slower decline in memory, attention, processing speed, and manual dexterity and subsequent slower decline in attention (all p < 0.05). Some associations were attenuated after exclusion of data points at and after symptom onset of cognitive impairment or dementia. DISCUSSION In older adults, olfaction is related to brain atrophy of specific brain regions and neuropsychological changes in specific domains over time. The observed associations are driven, in part, by those who developed cognitive impairment or dementia. Future longitudinal studies with longer follow-ups are needed to understand whether olfactory decline precedes cognitive decline and whether it is mediated through regionally specific brain atrophy.
Collapse
Affiliation(s)
- Qu Tian
- From the Translational Gerontology Branch (Q.T., S.A.S., L.F.), National Institute on Aging, Baltimore, MD; Laboratory of Behavioral Neuroscience (Y.A., M.H.K.T., S.M.R.), National Institute on Aging, Baltimore, MD; and Department of Radiology (C.D.), University of Pennsylvania School of Medicine, Philadelphia.
| | - Yang An
- From the Translational Gerontology Branch (Q.T., S.A.S., L.F.), National Institute on Aging, Baltimore, MD; Laboratory of Behavioral Neuroscience (Y.A., M.H.K.T., S.M.R.), National Institute on Aging, Baltimore, MD; and Department of Radiology (C.D.), University of Pennsylvania School of Medicine, Philadelphia.
| | - Melissa H Kitner-Triolo
- From the Translational Gerontology Branch (Q.T., S.A.S., L.F.), National Institute on Aging, Baltimore, MD; Laboratory of Behavioral Neuroscience (Y.A., M.H.K.T., S.M.R.), National Institute on Aging, Baltimore, MD; and Department of Radiology (C.D.), University of Pennsylvania School of Medicine, Philadelphia
| | - Christos Davatzikos
- From the Translational Gerontology Branch (Q.T., S.A.S., L.F.), National Institute on Aging, Baltimore, MD; Laboratory of Behavioral Neuroscience (Y.A., M.H.K.T., S.M.R.), National Institute on Aging, Baltimore, MD; and Department of Radiology (C.D.), University of Pennsylvania School of Medicine, Philadelphia
| | - Stephanie A Studenski
- From the Translational Gerontology Branch (Q.T., S.A.S., L.F.), National Institute on Aging, Baltimore, MD; Laboratory of Behavioral Neuroscience (Y.A., M.H.K.T., S.M.R.), National Institute on Aging, Baltimore, MD; and Department of Radiology (C.D.), University of Pennsylvania School of Medicine, Philadelphia
| | - Luigi Ferrucci
- From the Translational Gerontology Branch (Q.T., S.A.S., L.F.), National Institute on Aging, Baltimore, MD; Laboratory of Behavioral Neuroscience (Y.A., M.H.K.T., S.M.R.), National Institute on Aging, Baltimore, MD; and Department of Radiology (C.D.), University of Pennsylvania School of Medicine, Philadelphia
| | - Susan M Resnick
- From the Translational Gerontology Branch (Q.T., S.A.S., L.F.), National Institute on Aging, Baltimore, MD; Laboratory of Behavioral Neuroscience (Y.A., M.H.K.T., S.M.R.), National Institute on Aging, Baltimore, MD; and Department of Radiology (C.D.), University of Pennsylvania School of Medicine, Philadelphia
| |
Collapse
|
48
|
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
|