1
|
Oyabu M, Ohira Y, Fujita M, Yoshioka K, Kawaguchi R, Kubo A, Hatazawa Y, Yukitoshi H, Ortuste Quiroga HP, Horii N, Miura F, Araki H, Okano M, Hatada I, Gotoh H, Yoshizawa T, Fukada SI, Ogawa Y, Ito T, Ishihara K, Ono Y, Kamei Y. Dnmt3a overexpression disrupts skeletal muscle homeostasis, promotes an aging-like phenotype, and reduces metabolic elasticity. iScience 2025; 28:112144. [PMID: 40151644 PMCID: PMC11937683 DOI: 10.1016/j.isci.2025.112144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 05/10/2024] [Accepted: 02/27/2025] [Indexed: 03/29/2025] Open
Abstract
Mammalian aging is reportedly driven by the loss of epigenetic information; however, its impact on skeletal muscle aging remains unclear. This study shows that aging mouse skeletal muscle exhibits increased DNA methylation, and overexpression of DNA methyltransferase 3a (Dnmt3a) induces an aging-like phenotype. Muscle-specific Dnmt3a overexpression leads to an increase in central nucleus-positive myofibers, predominantly in fast-twitch fibers, a shift toward slow-twitch fibers, elevated inflammatory and senescence markers, mitochondrial OXPHOS complex I reduction, and decreased basal autophagy. Dnmt3a overexpression resulted in reduced muscle mass and strength and impaired endurance exercise capacity with age, accompanied by an enhanced inflammatory signature. In addition, Dnmt3a overexpression reduced not only sensitivity to starvation-induced muscle atrophy but also the restorability from muscle atrophy. These findings suggest that increased DNA methylation disrupts skeletal muscle homeostasis, promotes an aging-like phenotype, and reduces muscle metabolic elasticity.
Collapse
Affiliation(s)
- Mamoru Oyabu
- Graduate School of Life and Environmental Sciences, Kyoto Prefectural University, Kyoto 606-8522, Japan
| | - Yuto Ohira
- Graduate School of Life and Environmental Sciences, Kyoto Prefectural University, Kyoto 606-8522, Japan
| | - Mariko Fujita
- Graduate School of Life and Environmental Sciences, Kyoto Prefectural University, Kyoto 606-8522, Japan
| | - Kiyoshi Yoshioka
- Graduate School of Life and Environmental Sciences, Kyoto Prefectural University, Kyoto 606-8522, Japan
- Institute for Research on Productive Aging (IRPA), Tokyo, Japan
| | - Runa Kawaguchi
- Graduate School of Life and Environmental Sciences, Kyoto Prefectural University, Kyoto 606-8522, Japan
| | - Atsushi Kubo
- Laboratory of Stem Cell Regeneration and Adaptation, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Yukino Hatazawa
- Graduate School of Life and Environmental Sciences, Kyoto Prefectural University, Kyoto 606-8522, Japan
| | - Hinako Yukitoshi
- Graduate School of Life and Environmental Sciences, Kyoto Prefectural University, Kyoto 606-8522, Japan
| | - Huascar Pedro Ortuste Quiroga
- Department of Muscle Development and Regeneration, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto 860-0811, Japan
| | - Naoki Horii
- Department of Muscle Development and Regeneration, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto 860-0811, Japan
| | - Fumihito Miura
- Department of Biochemistry, Kyushu University Graduate School of Medical Sciences, Fukuoka 812-8582, Japan
| | - Hiromitsu Araki
- Department of Biochemistry, Kyushu University Graduate School of Medical Sciences, Fukuoka 812-8582, Japan
| | - Masaki Okano
- Department of Pluripotent Stem Cell Biology, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto 860-0811, Japan
| | - Izuho Hatada
- Laboratory of Genome Science, Biosignal Genome Resource Center, Institute for Molecular and Cellular Regulation, Gunma University, Gunma 371-8512, Japan
- Viral Vector Core, Gunma University Initiative for Advanced Research (GIAR), Maebashi 371-8511, Japan
| | - Hitoshi Gotoh
- Cell Biology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto 606-0823, Japan
| | - Tatsuya Yoshizawa
- Cell Biology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto 606-0823, Japan
| | - So-ichiro Fukada
- Laboratory of Stem Cell Regeneration and Adaptation, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Yoshihiro Ogawa
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan
| | - Takashi Ito
- Department of Biochemistry, Kyushu University Graduate School of Medical Sciences, Fukuoka 812-8582, Japan
| | - Kengo Ishihara
- Department of Food Science and Human Nutrition, Faculty of Agriculture, Ryukoku University, Shiga 520-2194, Japan
| | - Yusuke Ono
- Department of Muscle Development and Regeneration, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto 860-0811, Japan
- Tokyo Metropolitan Institute for Geriatrics and Gerontology, Tokyo 173-0015, Japan
| | - Yasutomi Kamei
- Graduate School of Life and Environmental Sciences, Kyoto Prefectural University, Kyoto 606-8522, Japan
| |
Collapse
|
2
|
Hardman D, Hennig K, Martins IB, Roman W, Gomes ER, Bernabeu MO. Quantitative measurement of morphometric indicators of skeletal muscle cell behaviour and quality. J R Soc Interface 2025; 22:20240634. [PMID: 40233801 PMCID: PMC11999735 DOI: 10.1098/rsif.2024.0634] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 01/24/2025] [Accepted: 03/10/2025] [Indexed: 04/17/2025] Open
Abstract
In vitro culturing of effective human-induced pluripotent stem cell-derived skeletal muscle cells (hiPSC-SMCs) has proven to be challenging. Progress is hindered by the limited range of metrics applied to assess experimental success. We present a semi-automated workflow for segmenting, tracking and quantifying migration and fusion behaviour in live and static images of myoblast and myotube cells. Workflow outputs are validated against manually labelled images and the metrics applied to images from case studies of in vitro cultures of primary mouse muscle cells under varying culture media conditions, mouse primary cells undergoing optogenetic stimulation and hiPSC-SMC. We show culture media-dependent differences in cell fusion dynamics and increased acetylcholine receptors in myonuclei under optogenetic stimulation. We show that myoblasts have greater persistence and proliferation in primary mouse cells than hiPSC, and cell-cell fusion occurred earlier but at a steadier rate in primary mouse cells.
Collapse
Affiliation(s)
- David Hardman
- Centre for Medical Informatics, The University of Edinburgh Usher Institute of Population Health Sciences and Informatics, Edinburgh, UK
| | - Katharina Hennig
- Universidade de Lisboa Instituto de Medicina Molecular João Lobo Antunes, Lisbon, Portugal
| | - Inês Belo Martins
- Universidade de Lisboa Instituto de Medicina Molecular João Lobo Antunes, Lisbon, Portugal
- Gulbenkian Institute for Molecular Medicine, Lisbon, Portugal
| | - William Roman
- Australian Regenerative Medicine Institute, Clayton, Victoria, Australia
- Victoria Node, European Molecular Biology Laboratory, Clayton, Victoria, Australia
| | - Edgar R. Gomes
- Universidade de Lisboa Instituto de Medicina Molecular João Lobo Antunes, Lisbon, Portugal
- Gulbenkian Institute for Molecular Medicine, Lisbon, Portugal
| | - Miguel O. Bernabeu
- Centre for Medical Informatics, The University of Edinburgh Usher Institute of Population Health Sciences and Informatics, Edinburgh, UK
| |
Collapse
|
3
|
Sullivan BP, Larson AA, Shams AS, McMillin SL, Ebeling MC, Peng S, Kyba M, Lowe DA. Estradiol deficiency as a consequence of aging contributes to the depletion of the satellite cell pool in female mice. Aging Cell 2025; 24:e14441. [PMID: 39641290 PMCID: PMC11984698 DOI: 10.1111/acel.14441] [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: 08/07/2024] [Revised: 10/29/2024] [Accepted: 11/17/2024] [Indexed: 12/07/2024] Open
Abstract
The effects of aging on the satellite cell pool have primarily been studied in male mice, where the role of cell-intrinsic versus environmental changes on satellite cell function remains contentious. Estradiol is necessary for maintenance of satellite cell pool size in adult female mice-here we investigate the hypothesis that in females, estradiol is a major environmental driver of age-associated effects on satellite cells. In 24-26 month-old ovarian senescent mice, we find the satellite cell pool size is severely diminished in certain muscles (TA and EDL) but only marginally affected in others (soleus and gastrocnemius). Supplementation with 17-beta estradiol significantly increases satellite cell pool size in the TA and EDL. To assess cell-intrinsic versus environmental regulation, we perform two transplantation experiments, Adult or Aged satellite cells transplanted into Adult recipients, and Adult satellite cells transplanted into Adult or Aged mice. These results demonstrate that the aged environment dominates over cell-autonomous age in terms of the specification of satellite cell pool size. Transcriptional profiling on satellite cells from Adult, Aged and ovariectomized mice revealed commonalities across the two estradiol-deficient conditions, Aged and ovariectomized, in GO terms from differentially expressed genes. Our findings support the hypothesis that the lack of estradiol contributes to reductions in satellite cell number in Aged female muscle, yet cells that remain are functional in terms of proliferative potential and self-renewal capacity. These findings have implications for sex hormone treatment of menopausal women and highlight the vital role of estradiol in the maintenance of the satellite cell pool.
Collapse
Affiliation(s)
- Brian P. Sullivan
- Division of Physical Therapy and Rehabilitation Science, Department of Family Medicine and Community Health, Medical SchoolUniversity of MinnesotaMinneapolisMinnesotaUSA
| | - Alexie A. Larson
- Division of Physical Therapy and Rehabilitation Science, Department of Family Medicine and Community Health, Medical SchoolUniversity of MinnesotaMinneapolisMinnesotaUSA
| | - Ahmed S. Shams
- Lillehei Heart Institute and Department of Pediatrics, Medical SchoolUniversity of MinnesotaMinneapolisMinnesotaUSA
- Human Anatomy and Embryology Department, Faculty of MedicineSuez Canal UniversityIsmailiaEgypt
| | - Shawna L. McMillin
- Division of Physical Therapy and Rehabilitation Science, Department of Family Medicine and Community Health, Medical SchoolUniversity of MinnesotaMinneapolisMinnesotaUSA
| | - Mara C. Ebeling
- Division of Physical Therapy and Rehabilitation Science, Department of Family Medicine and Community Health, Medical SchoolUniversity of MinnesotaMinneapolisMinnesotaUSA
| | - Sydney Peng
- Lillehei Heart Institute and Department of Pediatrics, Medical SchoolUniversity of MinnesotaMinneapolisMinnesotaUSA
| | - Michael Kyba
- Lillehei Heart Institute and Department of Pediatrics, Medical SchoolUniversity of MinnesotaMinneapolisMinnesotaUSA
| | - Dawn A. Lowe
- Division of Physical Therapy and Rehabilitation Science, Department of Family Medicine and Community Health, Medical SchoolUniversity of MinnesotaMinneapolisMinnesotaUSA
| |
Collapse
|
4
|
Matsuzaki R, Matsuoka T, Nakanishi K, Tani A, Kakimoto S, Kato Y, Kawatani T, Nakagawa S, Baba Y, Kobayashi M, Takihara T, Sakakima H. Effects of green tea catechins and exercise on age-related muscle atrophy and satellite cell functions in a mouse model of sarcopenia. Exp Gerontol 2025; 202:112720. [PMID: 40015490 DOI: 10.1016/j.exger.2025.112720] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Revised: 02/14/2025] [Accepted: 02/24/2025] [Indexed: 03/01/2025]
Abstract
Sarcopenia negatively affects the quality of life and health of older individuals. Physical exercise is a standard treatment for sarcopenia. Recently, the potential benefits of green tea catechins (GTCs) in sarcopenia have gained considerable attention. In this study, we investigated the effects of a combination of GTCs and physical exercise on the symptoms and pathologies of sarcopenia using male senescence-accelerated mouse prone 8 (SAMP8). These mice were divided into four groups: control, GTCs, exercise (Ex), and GTCs + Ex. GTC-fed mice were fed a diet containing 0.33 % GTCs. The mice were subjected to exercise training (voluntary wheel running) for 12 weeks, from 5 to 8 months of age, and grip strength and gastrocnemius muscle alterations were investigated. SAMP8 mice exhibited symptoms and pathologies of sarcopenia, including loss of muscle mass, decreased grip strength, reduced mitochondrial capacity, increased oxidative stress, reduced number of satellite cells, and an increased ratio of 5-bromo-2'-deoxyuridine (BrdU)-positive nuclei located within the muscle cells in the aged muscle. The GTCs and/or Ex groups showed improved symptoms and pathologies of sarcopenia. In addition, the GTCs + Ex group exhibited enhanced mitochondrial capacity, myogenic differentiation, and maturation in aged skeletal muscle than that observed in the GTCs group. Our findings suggested that GTCs and/or Ex are effective in ameliorating several age-related changes in muscle morphology and function. Notably, GTCs intake, together with habitual exercise, may enhance the beneficial effects on the symptoms and pathologies of sarcopenia in aged muscle.
Collapse
MESH Headings
- Animals
- Sarcopenia/pathology
- Sarcopenia/physiopathology
- Sarcopenia/therapy
- Sarcopenia/metabolism
- Satellite Cells, Skeletal Muscle/drug effects
- Satellite Cells, Skeletal Muscle/physiology
- Satellite Cells, Skeletal Muscle/pathology
- Male
- Catechin/pharmacology
- Disease Models, Animal
- Physical Conditioning, Animal/physiology
- Mice
- Muscle, Skeletal/drug effects
- Muscle, Skeletal/pathology
- Muscle, Skeletal/metabolism
- Muscle, Skeletal/physiopathology
- Aging/pathology
- Tea/chemistry
- Oxidative Stress/drug effects
- Muscular Atrophy/pathology
- Muscular Atrophy/physiopathology
- Muscle Strength/drug effects
- Hand Strength
- Mitochondria, Muscle/drug effects
Collapse
Affiliation(s)
- Ryoma Matsuzaki
- Department of Physical Therapy, School of Health Sciences, Faculty of Medicine, Kagoshima University, Kagoshima, Japan
| | - Teruki Matsuoka
- Department of Physical Therapy, School of Health Sciences, Faculty of Medicine, Kagoshima University, Kagoshima, Japan
| | - Kazuki Nakanishi
- Department of Physical Therapy, School of Health Sciences, Faculty of Medicine, Kagoshima University, Kagoshima, Japan
| | - Akira Tani
- Department of Physical Therapy, School of Health Sciences, Faculty of Medicine, Kagoshima University, Kagoshima, Japan
| | - Shogo Kakimoto
- Department of Physical Therapy, School of Health Sciences, Faculty of Medicine, Kagoshima University, Kagoshima, Japan
| | - Yuki Kato
- Department of Physical Therapy, School of Health Sciences, Faculty of Medicine, Kagoshima University, Kagoshima, Japan
| | - Takuya Kawatani
- Department of Physical Therapy, School of Health Sciences, Faculty of Medicine, Kagoshima University, Kagoshima, Japan
| | - Sae Nakagawa
- Central Research Institute, ITO EN, Ltd., 21 Mekami, Makinohara, Shizuoka 421-0516, Japan
| | - Yoshitake Baba
- Central Research Institute, ITO EN, Ltd., 21 Mekami, Makinohara, Shizuoka 421-0516, Japan
| | - Makoto Kobayashi
- Central Research Institute, ITO EN, Ltd., 21 Mekami, Makinohara, Shizuoka 421-0516, Japan
| | - Takanobu Takihara
- Central Research Institute, ITO EN, Ltd., 21 Mekami, Makinohara, Shizuoka 421-0516, Japan
| | - Harutoshi Sakakima
- Department of Physical Therapy, School of Health Sciences, Faculty of Medicine, Kagoshima University, Kagoshima, Japan.
| |
Collapse
|
5
|
Conning-Rowland M, Cheng CW, Brown O, Giannoudi M, Levelt E, Roberts LD, Griffin KJ, Cubbon RM. Application of CIBERSORTx and BayesPrism to deconvolution of bulk RNA-seq data from human myocardium and skeletal muscle. Heliyon 2025; 11:e42499. [PMID: 40034311 PMCID: PMC11872574 DOI: 10.1016/j.heliyon.2025.e42499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 02/04/2025] [Accepted: 02/05/2025] [Indexed: 03/05/2025] Open
Abstract
RNA-sequencing (RNA-seq) is an important tool to explore molecular mechanisms of disease. Technological advances mean this can be performed at the single-cell level, but the large sample sizes needed in clinical studies are currently prohibitively expensive and complex. Deconvolution of bulk RNA-seq offers an opportunity to bridge this gap by defining the cell lineage composition of samples. This approach is widely used in immunology studies, but currently there are no validated pipelines for researchers analysing human myocardium or skeletal muscle. Here, we describe the application and in silico validation of two pipelines to deconvolute human right atrium, left ventricle and skeletal muscle bulk RNA-seq data. Specifically, we have defined the major cell lineages of these tissues using single cell/nucleus RNA-seq data from the Heart Cell Atlas, which are then applied during deconvolution using the CIBERSORTx or BayesPrism deconvolution packages. Both pipelines gave robust estimates of the proportion of all major cell lineages in these tissues. We demonstrate their value in defining age- and sex-differences in tissue composition using bulk RNA-seq data from the GTEx consortium. Our validated pipelines can be rapidly applied by researchers working with existing or novel bulk RNA-seq of myocardium or skeletal muscle to gain novel insights.
Collapse
Affiliation(s)
- Marcella Conning-Rowland
- Leeds Institute of Cardiovascular and Metabolic Medicine, The University of Leeds, Leeds, United Kingdom
| | - Chew W. Cheng
- Leeds Institute of Cardiovascular and Metabolic Medicine, The University of Leeds, Leeds, United Kingdom
| | - Oliver Brown
- Leeds Institute of Cardiovascular and Metabolic Medicine, The University of Leeds, Leeds, United Kingdom
| | - Marilena Giannoudi
- Leeds Institute of Cardiovascular and Metabolic Medicine, The University of Leeds, Leeds, United Kingdom
| | - Eylem Levelt
- Leeds Institute of Cardiovascular and Metabolic Medicine, The University of Leeds, Leeds, United Kingdom
| | - Lee D. Roberts
- Leeds Institute of Cardiovascular and Metabolic Medicine, The University of Leeds, Leeds, United Kingdom
| | - Kathryn J. Griffin
- Leeds Institute of Cardiovascular and Metabolic Medicine, The University of Leeds, Leeds, United Kingdom
| | - Richard M. Cubbon
- Leeds Institute of Cardiovascular and Metabolic Medicine, The University of Leeds, Leeds, United Kingdom
| |
Collapse
|
6
|
Altab G, Merry BJ, Beckett CW, Raina P, Lopes I, Goljanek-Whysall K, de Magalhães JP. Unravelling the transcriptomic symphony of muscle ageing: key pathways and hub genes altered by ageing and caloric restriction in rat muscle revealed by RNA sequencing. BMC Genomics 2025; 26:29. [PMID: 39800693 PMCID: PMC11727704 DOI: 10.1186/s12864-024-11051-1] [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: 05/13/2024] [Accepted: 11/14/2024] [Indexed: 01/16/2025] Open
Abstract
Age-related muscle wasting, sarcopenia is an extensive loss of muscle mass and strength with age and a major cause of disability and accidents in the elderly. Mechanisms purported to be involved in muscle ageing and sarcopenia are numerous but poorly understood, necessitating deeper study. Hence, we employed high-throughput RNA sequencing to survey the global changes in protein-coding gene expression occurring in skeletal muscle with age. Caloric restriction (CR) is a known prophylactic intervention against sarcopenia. Therefore, total RNA was isolated from the muscle tissue of both rats fed ad libitum and CR rats. RNA-seq data were subjected to Gene Ontology, pathway, co-expression, and interaction network analyses. This revealed the functional pathways most activated by both ageing and CR, as well as the key "hub" proteins involved in their activation.RNA-seq revealed 442 protein-coding genes to be upregulated and 377 to be downregulated in aged muscle, compared to young muscle. Upregulated genes were commonly involved in protein folding and immune responses; meanwhile, downregulated genes were often related to developmental biology. CR was found to suppress 69.7% and rescue 57.8% of the genes found to be upregulated and downregulated in aged muscle, respectively. In addition, CR uniquely upregulated 291 and downregulated 304 protein-coding genes. Hub genes implicated in both ageing and CR included Gc, Plg, Irf7, Ifit3, Usp18, Rsad2, Blm and RT1-A2, whilst those exclusively implicated in CR responses included Alb, Apoa1, Ambp, F2, Apoh, Orm1, Mx1, Oasl2 and Rtp4. Hub genes involved in ageing but unaffected by CR included Fgg, Fga, Fgb and Serpinc1. In conclusion, this comprehensive RNA sequencing study highlights gene expression patterns, hub genes and signalling pathways most affected by ageing in skeletal muscle. This data may provide the initial evidence for several targets for potential future therapeutic interventions against sarcopenia.
Collapse
Affiliation(s)
- Gulam Altab
- Integrative Genomics of Ageing Group, Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool, L7 8TX, UK
| | - Brian J Merry
- Institute of Integrative Biology, University of Liverpool, Liverpool, L69 3BX, UK
| | - Charles W Beckett
- Integrative Genomics of Ageing Group, Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool, L7 8TX, UK
| | - Priyanka Raina
- Integrative Genomics of Ageing Group, Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool, L7 8TX, UK
| | - Inês Lopes
- Integrative Genomics of Ageing Group, Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool, L7 8TX, UK
| | - Katarzyna Goljanek-Whysall
- Integrative Genomics of Ageing Group, Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool, L7 8TX, UK
- College of Medicine, Nursing and Health Sciences, University of Galway, Galway, H91 TK33, Ireland
| | - João Pedro de Magalhães
- Integrative Genomics of Ageing Group, Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool, L7 8TX, UK.
- Institute of Inflammation and Ageing, University of Birmingham, Queen Elizabeth Hospital, Mindelsohn Way, Birmingham, B15 2WB, UK.
| |
Collapse
|
7
|
Wang T, Zhou D, Hong Z. Sarcopenia and cachexia: molecular mechanisms and therapeutic interventions. MedComm (Beijing) 2025; 6:e70030. [PMID: 39764565 PMCID: PMC11702502 DOI: 10.1002/mco2.70030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 11/11/2024] [Accepted: 11/12/2024] [Indexed: 03/17/2025] Open
Abstract
Sarcopenia is defined as a muscle-wasting syndrome that occurs with accelerated aging, while cachexia is a severe wasting syndrome associated with conditions such as cancer and immunodeficiency disorders, which cannot be fully addressed through conventional nutritional supplementation. Sarcopenia can be considered a component of cachexia, with the bidirectional interplay between adipose tissue and skeletal muscle potentially serving as a molecular mechanism for both conditions. However, the underlying mechanisms differ. Recognizing the interplay and distinctions between these disorders is essential for advancing both basic and translational research in this area, enhancing diagnostic accuracy and ultimately achieving effective therapeutic solutions for affected patients. This review discusses the muscle microenvironment's changes contributing to these conditions, recent therapeutic approaches like lifestyle modifications, small molecules, and nutritional interventions, and emerging strategies such as gene editing, stem cell therapy, and gut microbiome modulation. We also address the challenges and opportunities of multimodal interventions, aiming to provide insights into the pathogenesis and molecular mechanisms of sarcopenia and cachexia, ultimately aiding in innovative strategy development and improved treatments.
Collapse
Affiliation(s)
- Tiantian Wang
- Department of NeurologyWest China Hospital of Sichuan UniversityChengduSichuanChina
- Institute of Brain Science and Brain‐Inspired Technology of West China HospitalSichuan UniversityChengduSichuanChina
- Department of NeurologyChengdu Shangjin Nanfu HospitalChengduSichuanChina
| | - Dong Zhou
- Department of NeurologyWest China Hospital of Sichuan UniversityChengduSichuanChina
- Institute of Brain Science and Brain‐Inspired Technology of West China HospitalSichuan UniversityChengduSichuanChina
- Department of NeurologyChengdu Shangjin Nanfu HospitalChengduSichuanChina
| | - Zhen Hong
- Department of NeurologyWest China Hospital of Sichuan UniversityChengduSichuanChina
- Institute of Brain Science and Brain‐Inspired Technology of West China HospitalSichuan UniversityChengduSichuanChina
- Department of NeurologyChengdu Shangjin Nanfu HospitalChengduSichuanChina
| |
Collapse
|
8
|
Lin WH, Tzeng CY, Kao FC, Tsao CW, Li N, Wu CC, Lee SH, Huang KF, Hu WW, Chen SL. The proliferation and differentiation of skeletal muscle stem cells are enhanced in a bioreactor. Biotechnol Bioeng 2025; 122:95-109. [PMID: 39369338 DOI: 10.1002/bit.28857] [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: 04/03/2024] [Revised: 09/18/2024] [Accepted: 09/24/2024] [Indexed: 10/07/2024]
Abstract
Skeletal muscle (SKM) is the largest organ in mammalian body and it can repair damages by using the residential myogenic stem cells (MuSC), but this repairing capacity reduces with age and in some genetic muscular dystrophy. Under these circumstances, artificial amplification of autologous MuSC in vitro might be necessary to repair the damaged SKM. The amplification of MuSC is highly dependent on myogenic signals, such as sonic hedgehog (Shh), Wnt3a, and fibroblast growth factors, so formulating an optimum myogenic kit composed of specific myogenic signals might increase the proliferation and differentiation of MuSC efficiently. In this study, various myogenic signals have been tested on C2C12 myoblasts and primary MuSC, and a myogenic kit consists of insulin, lithium chloride, T3, and retinoic acid has been formulated, and we found it significantly increased the fusion index and MHC expression level of both C2C12 and MuSC myotubes. A novel bioreactor providing cyclic stretching (CS) and electrical stimulation (ES) has been fabricated to enhance the myogenic differentiation of both C2C12 and MuSC. We further found that coating the bioreactor substratum with collagen gave the best effect on proliferation and differentiation of MuSC. Furthermore, combining the collagen coating and physical stimuli (CS + ES) in the bioreactor can generate more proliferative primary MuSC cells. Our results have demonstrated that the combination of myogenic kit and bioreactor can provide environment for efficient MuSC proliferation and differentiation. These MuSC and mature myotubes amplified in the bioreactor might be useful for clinical grafting into damaged SKM in the future.
Collapse
Affiliation(s)
- Wei-Hsuan Lin
- Department of Life Sciences, National Central University, Taoyuan, Taiwan
| | - Chung-Yuh Tzeng
- Department of Orthopedics, Taichung Veterans General Hospital, Taichung, Taiwan
- Department of Rehabilitation, Jen-Teh Junior College of Medicine, Nursing, and Management, Miaoli, Taiwan
- Institute of Biomedical Sciences, National Chung Hsing University, Taichung, Taiwan
| | - Fan-Che Kao
- Department of Life Sciences, National Central University, Taoyuan, Taiwan
| | - Chia-Wen Tsao
- Department of Mechanical Engineering, National Central University, Taoyuan, Taiwan
| | - Ning Li
- Department of Life Sciences, National Central University, Taoyuan, Taiwan
| | - Chuan-Che Wu
- Department of Life Sciences, National Central University, Taoyuan, Taiwan
| | - Sheng-Huei Lee
- Department of Electric Engineering, Chien Hsin University of Science and Technology, Taoyuan, Taiwan
| | - Kai-Fan Huang
- Department of Life Sciences, National Central University, Taoyuan, Taiwan
| | - Wei-Wen Hu
- Department of Chemical and Materials Engineering, National Central University, Taoyuan, Taiwan
| | - Shen-Liang Chen
- Department of Life Sciences, National Central University, Taoyuan, Taiwan
| |
Collapse
|
9
|
Janssen TA, Lowisz CV, Phillips S. From molecular to physical function: The aging trajectory. Curr Res Physiol 2024; 8:100138. [PMID: 39811024 PMCID: PMC11732118 DOI: 10.1016/j.crphys.2024.100138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 10/18/2024] [Accepted: 12/15/2024] [Indexed: 01/16/2025] Open
Abstract
Aging is accompanied by a decline in muscle mass, strength, and physical function, a condition known as sarcopenia. Muscle disuse attributed to decreased physical activity, hospitalization, or illness (e.g. sarcopenia) results in a rapid decline in muscle mass in aging individuals and effectively accelerates sarcopenia. Consuming protein at levels above (at least 50-100% higher) the current recommended intakes of ∼0.8 g protein/kg bodyweight/d, along with participating in both resistance and aerobic exercise, will aid in the preservation of muscle mass. Physiological muscle adaptations often accompany the observable changes in physical independence an older adult undergoes. Muscle fibre adaptations include a reduction in type 2 fibre size and number, a loss of motor units, reduced sensitivity to calcium, reduced elasticity, and weak cross-bridges. Mitochondrial function and structure are impaired in relation to aging and are worsened with inactivity and disease states but could be overcome by engaging in exercise. Intramuscular connective tissue adaptations with age are evident in animal models; however, the adaptations in collagenous tissue within human aging are less clear. We know that the satellite muscle cell pool decreases with age, and there is a reduced capacity for muscle repair/regeneration. Finally, a pro-inflammatory state associated with age has detrimental impacts on the muscle. The purpose of this review is to highlight the physiological adaptations driving muscle aging and their potential mitigation with exercise/physical activity and nutrition.
Collapse
Affiliation(s)
- Tom A.H. Janssen
- Department of Kinesiology, McMaster University, Hamilton, Ontario, Canada
| | - Caroline V. Lowisz
- Department of Kinesiology, McMaster University, Hamilton, Ontario, Canada
| | - Stuart Phillips
- Department of Kinesiology, McMaster University, Hamilton, Ontario, Canada
- Department of Sport and Exercise Science, Manchester Metropolitan University Institute of Sport, Manchester, UK
| |
Collapse
|
10
|
Wang K, Wang X, Wang Y. Factors, mechanisms and improvement methods of muscle strength loss. Front Cell Dev Biol 2024; 12:1509519. [PMID: 39698495 PMCID: PMC11653071 DOI: 10.3389/fcell.2024.1509519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Accepted: 11/21/2024] [Indexed: 12/20/2024] Open
Abstract
Muscle strength is a crucial aspect of muscle function, essential for maintaining normal physical activity and quality of life. The global aging population coupled with the increasing prevalence of muscle disorders and strength loss, poses a remarkable public health challenge. Understanding the mechanisms behind muscle strength decline is vital for improving public health outcomes. This review discusses recent research advancements on muscle strength loss from various perspectives, including factors contributing to muscle strength decline, the signaling pathways involved in the deterioration of muscle function, and the methods for assessing muscle strength. The final section explores the influence of exercise stimulation and nutrition on muscle strength.
Collapse
Affiliation(s)
- Kaiyong Wang
- Department of Physical Education, Guangdong University of Finance and Economics, Guangzhou, Guangdong, China
| | - Xuyu Wang
- Master program under the Graduate School of Education, Graduate University of Mongolia, Ulaanbaatar, Mongolia
| | - Yanqiu Wang
- School of Physical Education, Central China Normal University, Wuhan, Hubei, China
| |
Collapse
|
11
|
Zhu GZ, Zhao K, Li HZ, Wu DZ, Chen YB, Han D, Gao JW, Chen XY, Yu YP, Huang ZW, Tu C, Zhong ZM. Melatonin ameliorates age-related sarcopenia by inhibiting fibrogenic conversion of satellite cell. Mol Med 2024; 30:238. [PMID: 39614149 DOI: 10.1186/s10020-024-00998-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Accepted: 11/13/2024] [Indexed: 12/01/2024] Open
Abstract
The fibrogenic conversion of satellite cells contributes to the atrophy and fibrosis of skeletal muscle, playing a significant role in the pathogenesis of age-related sarcopenia. Melatonin, a hormone secreted by the pineal gland, exhibits anti-aging and anti-fibrotic effects in various conditions. However, the effect of melatonin on satellite cell fate and age-related sarcopenia remains under-explored. Here, we report that melatonin treatment mitigated the loss of muscle mass and strength in aged mice, replenished the satellite cell pool and curtailed muscle fibrosis. When primary SCs were cultured in vitro and subjected to aging induction via D-galactose, they exhibited a diminished myogenic potential and a conversion from myogenic to fibrogenic lineage. Notably, melatonin treatment effectively restored the myogenic potential and inhibited this lineage conversion. Furthermore, melatonin attenuated the expression of the fibrogenic cytokine, transforming growth factor-β1, and reduced the phosphorylation of its downstream targets Smad2/3 both in vivo and in vitro. In summary, our findings show melatonin's capacity to counteract muscle decline and inhibit fibrogenic conversion in aging SCs and highlight its potential therapeutic value for age-related sarcopenia.
Collapse
Affiliation(s)
- Guo-Zheng Zhu
- Division of Spine Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, 1838 North Guangzhou Ave, Guangzhou, 510515, People's Republic of China
- Department of Orthopaedics, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Kai Zhao
- Department of Orthopaedics, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Hong-Zhou Li
- Division of Spine Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, 1838 North Guangzhou Ave, Guangzhou, 510515, People's Republic of China
| | - Di-Zheng Wu
- Division of Spine Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, 1838 North Guangzhou Ave, Guangzhou, 510515, People's Republic of China
| | - Yun-Biao Chen
- Division of Spine Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, 1838 North Guangzhou Ave, Guangzhou, 510515, People's Republic of China
| | - Dong Han
- Division of Spine Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, 1838 North Guangzhou Ave, Guangzhou, 510515, People's Republic of China
| | - Jia-Wen Gao
- Division of Spine Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, 1838 North Guangzhou Ave, Guangzhou, 510515, People's Republic of China
| | - Xing-Yu Chen
- Division of Spine Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, 1838 North Guangzhou Ave, Guangzhou, 510515, People's Republic of China
| | - Yong-Peng Yu
- Division of Spine Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, 1838 North Guangzhou Ave, Guangzhou, 510515, People's Republic of China
| | - Zhi-Wei Huang
- Division of Spine Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, 1838 North Guangzhou Ave, Guangzhou, 510515, People's Republic of China
| | - Chen Tu
- Division of Spine Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, 1838 North Guangzhou Ave, Guangzhou, 510515, People's Republic of China
| | - Zhao-Ming Zhong
- Division of Spine Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, 1838 North Guangzhou Ave, Guangzhou, 510515, People's Republic of China.
| |
Collapse
|
12
|
Amornsuradech S, Yamaguchi K, Nakagawa K, Yoshimi K, Moritoyo R, Tohara H. Aging-associated differences between perioral and trunk muscle characteristics. Sci Rep 2024; 14:27745. [PMID: 39532993 PMCID: PMC11557850 DOI: 10.1038/s41598-024-79268-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Accepted: 11/07/2024] [Indexed: 11/16/2024] Open
Abstract
The physiological and functional changes from aging affect the systemic and swallowing muscles. While ultrasound is used to examine muscle characteristics, no previous studies have examined the relationship between age and perioral and trunk muscle characteristics, including stiffness. This study aimed to investigate the relationship between age and the characteristics of perioral and trunk muscles, including quality, quantity, and stiffness, using ultrasound. A total of 215 participants were recruited for this cross-sectional study: 98 younger adults (20-64 years) and 117 older adults (≥ 65 years). Muscle characteristics were assessed along with data on age, sex, body mass index, and number of teeth. The correlation between age and muscle characteristics was examined. Multiple linear regression was performed with muscle characteristics as dependent variables and age, body mass index, and number of teeth as explanatory variables. We found that only trunk muscle stiffness was associated with age, with a stronger correlation in quality and quantity than perioral muscles. Masseter muscle quality and quantity showed a higher correlation with age than those of the geniohyoid muscle. In conclusion, characteristics of the trunk and perioral muscles exhibit differences in age-related changes, highlighting the importance of maintaining both trunk and perioral muscle activity in older adults to prevent functional decline.
Collapse
Affiliation(s)
- Sirinthip Amornsuradech
- Department of Dysphagia Rehabilitation, Graduate School of Medical and Dental Sciences, Institute of Science Tokyo, Tokyo, Japan
- Department of Community Dentistry, Faculty of Dentistry, Mahidol University, Bangkok, Thailand
| | - Kohei Yamaguchi
- Department of Dysphagia Rehabilitation, Graduate School of Medical and Dental Sciences, Institute of Science Tokyo, Tokyo, Japan.
| | - Kazuharu Nakagawa
- Department of Dysphagia Rehabilitation, Graduate School of Medical and Dental Sciences, Institute of Science Tokyo, Tokyo, Japan
| | - Kanako Yoshimi
- Department of Dysphagia Rehabilitation, Graduate School of Medical and Dental Sciences, Institute of Science Tokyo, Tokyo, Japan
| | - Rieko Moritoyo
- Department of Dysphagia Rehabilitation, Graduate School of Medical and Dental Sciences, Institute of Science Tokyo, Tokyo, Japan
| | - Haruka Tohara
- Department of Dysphagia Rehabilitation, Graduate School of Medical and Dental Sciences, Institute of Science Tokyo, Tokyo, Japan
| |
Collapse
|
13
|
Loreti M, Cecchini A, Kaufman CD, Stamenkovic C, Renero A, Nicoletti C, Kervadec A, Guarnaccia G, Mayer D, Colas A, Lorenzo Puri P, Sacco A. Tenascin-C from the tissue microenvironment promotes muscle stem cell self-renewal through Annexin A2. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.29.620732. [PMID: 39554125 PMCID: PMC11565721 DOI: 10.1101/2024.10.29.620732] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/19/2024]
Abstract
Skeletal muscle tissue self-repair occurs through the finely timed activation of resident muscle stem cells (MuSC). Following perturbation, MuSC exit quiescence, undergo myogenic commitment, and differentiate to regenerate the injured muscle. This process is coordinated by signals present in the tissue microenvironment, however the precise mechanisms by which the microenvironment regulates MuSC activation are still poorly understood. Here, we identified Tenascin-C (TnC), an extracellular matrix (ECM) glycoprotein, as a key player in promoting of MuSC self-renewal and function. We show that fibro-adipogenic progenitors (FAPs) are the primary cellular source of TnC during muscle repair, and that MuSC sense TnC signaling through cell the surface receptor Annexin A2. We provide in vivo evidence that TnC is required for efficient muscle repair, as mice lacking TnC exhibit a regeneration phenotype of premature aging. We propose that the decline of TnC in physiological aging contributes to inefficient muscle regeneration in aged muscle. Taken together, our results highlight the pivotal role of TnC signaling during muscle repair in healthy and aging skeletal muscle.
Collapse
Affiliation(s)
- Mafalda Loreti
- Development, Aging and Regeneration Program, Sanford Burnham Prebys Medical Discovery Institute, 10901 N Torrey Pines Rd, La Jolla, CA 92037, USA
- Current affiliation: J&J, 3880 Murphy Canyon Rd, San Diego, CA 92123, USA
| | - Alessandra Cecchini
- Development, Aging and Regeneration Program, Sanford Burnham Prebys Medical Discovery Institute, 10901 N Torrey Pines Rd, La Jolla, CA 92037, USA
| | - Collin D. Kaufman
- Development, Aging and Regeneration Program, Sanford Burnham Prebys Medical Discovery Institute, 10901 N Torrey Pines Rd, La Jolla, CA 92037, USA
| | - Cedomir Stamenkovic
- Development, Aging and Regeneration Program, Sanford Burnham Prebys Medical Discovery Institute, 10901 N Torrey Pines Rd, La Jolla, CA 92037, USA
| | - Alma Renero
- Development, Aging and Regeneration Program, Sanford Burnham Prebys Medical Discovery Institute, 10901 N Torrey Pines Rd, La Jolla, CA 92037, USA
- Current affiliation: University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Chiara Nicoletti
- Development, Aging and Regeneration Program, Sanford Burnham Prebys Medical Discovery Institute, 10901 N Torrey Pines Rd, La Jolla, CA 92037, USA
| | - Anais Kervadec
- Development, Aging and Regeneration Program, Sanford Burnham Prebys Medical Discovery Institute, 10901 N Torrey Pines Rd, La Jolla, CA 92037, USA
- Current affiliation: Avidity Biosciences, Inc., 10578 Science Center Drive Suite 125, San Diego, CA 92121, USA
| | - Gabriele Guarnaccia
- Development, Aging and Regeneration Program, Sanford Burnham Prebys Medical Discovery Institute, 10901 N Torrey Pines Rd, La Jolla, CA 92037, USA
| | - Daphne Mayer
- Rice University, 6100 Main St, Huston, TX 77005, USA
| | - Alexandre Colas
- Development, Aging and Regeneration Program, Sanford Burnham Prebys Medical Discovery Institute, 10901 N Torrey Pines Rd, La Jolla, CA 92037, USA
| | - Pier Lorenzo Puri
- Development, Aging and Regeneration Program, Sanford Burnham Prebys Medical Discovery Institute, 10901 N Torrey Pines Rd, La Jolla, CA 92037, USA
| | - Alessandra Sacco
- Development, Aging and Regeneration Program, Sanford Burnham Prebys Medical Discovery Institute, 10901 N Torrey Pines Rd, La Jolla, CA 92037, USA
| |
Collapse
|
14
|
Liu J, Chen M, Xia X, Wang Z, Wang Y, Xi L. Causal associations between the insulin-like growth factor family and sarcopenia: a bidirectional Mendelian randomization study. Front Endocrinol (Lausanne) 2024; 15:1422472. [PMID: 39507055 PMCID: PMC11537870 DOI: 10.3389/fendo.2024.1422472] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Accepted: 10/08/2024] [Indexed: 11/08/2024] Open
Abstract
Objective Insulin-like growth factor (IGF) is closely associated with sarcopenia, yet the causal relationship of this association remains unclear. This study aims to explore the potential causal relationship between members of the IGF family and sarcopenia from a genetic perspective through bidirectional Mendelian randomization (MR) analysis using two-sample datasets. Methods Five genetically predicted factors of the IGF family (IGF-1, IGF-1R, IGF-2R, IGFBP-3, IGFBP-7) as one sample, while four relevant features of sarcopenia (low hand grip strength, appendicular lean mass, whole body fat-free mass, and walking pace) as another sample, in conducting a two-sample MR analysis. Results The forward MR results of the relationship between IGF and sarcopenia showed that elevated levels of IGF-1 reduced the risk of low hand grip strength (OR = 0.936, 95% CI=0.892-0.983, P = 0.008) and increased appendicular lean mass of the extremities and whole body fat-free mass (OR = 1.125, 95% CI=1.070-1.182,P = 0.000; OR =1.076, 95% CI=1.047-1.106, P=0.000), reduced the risk of sarcopenia. Elevated IGF-1R also favored an increase in whole body fat-free mass (OR=1.023, 95% CI=1.008-1.038, P =0.002), and the appendicular lean mass trait was more pronounced with elevated IGFBP-3 and IGFBP-7 (OR=1.034, 95% CI=1.024-1.044, P =0.000; OR=1.020, 95% CI=1.010-1.030, P=0.000). Inverse MR results of the effect of sarcopenia on IGF showed that decreased hand grip strength may elevate IGF-1 levels (OR=1.243, 95% CI=1.026-1.505,P =0.027), whereas improvements in appendicular lean mass, whole body fat-free mass traits, and increased walking pace decreased IGF-1 levels (OR=0.902, 95% CI: 0.877-0.927, P = 0.000; OR=0.903, 95% CI=0.859-0.949,P = 0.000; OR=0.209, 95% CI=0.051-0.862,P = 0.045). Also decreased hand grip strength may elevate IGF-1R levels (OR=1.454, 95% CI=1.108-1.909, P =0.007), and appendicular lean mass stimulated high expression of IGFBP-1 (OR=1.314, 95% CI=1.003-1.722, P =0.047). Heterogeneity and pleiotropy were not detected in all results, and the results were stable and reliable. Conclusion There is a bi-directional causal association between IGF family members and the risk of sarcopenia, which provides a more adequate basis for early biological monitoring of sarcopenia and may provide new targets for early intervention and treatment of sarcopenia.
Collapse
Affiliation(s)
- Jili Liu
- Department of Geriatrics, The First Hospital, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Meng Chen
- Department of Geriatrics and Special Needs Medicine, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, China
| | - Xin Xia
- The Center of Gerontology and Geriatrics and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Zhaolin Wang
- Department of Traditional Chinese Medicine, The Second Hospital, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Yanqin Wang
- Department of Hematology, Shanxi Hospital of Traditional Chinese Medicine, Taiyuan, Shanxi, China
| | - Ling Xi
- Department of Geriatrics, The First Hospital, Shanxi Medical University, Taiyuan, Shanxi, China
| |
Collapse
|
15
|
Moriggi M, Torretta E, Cescon M, Russo L, Gregorio I, Braghetta P, Sabatelli P, Faldini C, Merlini L, Gargioli C, Bonaldo P, Gelfi C, Capitanio D. Characterization of Proteome Changes in Aged and Collagen VI-Deficient Human Pericyte Cultures. Int J Mol Sci 2024; 25:7118. [PMID: 39000224 PMCID: PMC11241165 DOI: 10.3390/ijms25137118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 06/17/2024] [Accepted: 06/25/2024] [Indexed: 07/16/2024] Open
Abstract
Pericytes are a distinct type of cells interacting with endothelial cells in blood vessels and contributing to endothelial barrier integrity. Furthermore, pericytes show mesenchymal stem cell properties. Muscle-derived pericytes can demonstrate both angiogenic and myogenic capabilities. It is well known that regenerative abilities and muscle stem cell potential decline during aging, leading to sarcopenia. Therefore, this study aimed to investigate the potential of pericytes in supporting muscle differentiation and angiogenesis in elderly individuals and in patients affected by Ullrich congenital muscular dystrophy or by Bethlem myopathy, two inherited conditions caused by mutations in collagen VI genes and sharing similarities with the progressive skeletal muscle changes observed during aging. The study characterized pericytes from different age groups and from individuals with collagen VI deficiency by mass spectrometry-based proteomic and bioinformatic analyses. The findings revealed that aged pericytes display metabolic changes comparable to those seen in aging skeletal muscle, as well as a decline in their stem potential, reduced protein synthesis, and alterations in focal adhesion and contractility, pointing to a decrease in their ability to form blood vessels. Strikingly, pericytes from young patients with collagen VI deficiency showed similar characteristics to aged pericytes, but were found to still handle oxidative stress effectively together with an enhanced angiogenic capacity.
Collapse
Affiliation(s)
- Manuela Moriggi
- Department of Biomedical Sciences for Health, University of Milano, 20133 Milano, Italy; (M.M.); (C.G.)
| | - Enrica Torretta
- Laboratory of Proteomics and Lipidomics, IRCCS Orthopedic Institute Galeazzi, 20161 Milano, Italy;
| | - Matilde Cescon
- Department of Molecular Medicine, University of Padova, 35121 Padova, Italy; (M.C.); (L.R.); (I.G.); (P.B.); (P.B.)
| | - Loris Russo
- Department of Molecular Medicine, University of Padova, 35121 Padova, Italy; (M.C.); (L.R.); (I.G.); (P.B.); (P.B.)
| | - Ilaria Gregorio
- Department of Molecular Medicine, University of Padova, 35121 Padova, Italy; (M.C.); (L.R.); (I.G.); (P.B.); (P.B.)
| | - Paola Braghetta
- Department of Molecular Medicine, University of Padova, 35121 Padova, Italy; (M.C.); (L.R.); (I.G.); (P.B.); (P.B.)
| | - Patrizia Sabatelli
- CNR-Institute of Molecular Genetics, 40136 Bologna, Italy;
- IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy
| | - Cesare Faldini
- 1st Orthopedics and Traumatology Department, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy;
- Department of Biomedical and Neuromotor Science, DIBINEM, University of Bologna, 40136 Bologna, Italy;
| | - Luciano Merlini
- Department of Biomedical and Neuromotor Science, DIBINEM, University of Bologna, 40136 Bologna, Italy;
| | - Cesare Gargioli
- Department of Biology, University of Rome Tor Vergata, 00133 Rome, Italy;
| | - Paolo Bonaldo
- Department of Molecular Medicine, University of Padova, 35121 Padova, Italy; (M.C.); (L.R.); (I.G.); (P.B.); (P.B.)
| | - Cecilia Gelfi
- Department of Biomedical Sciences for Health, University of Milano, 20133 Milano, Italy; (M.M.); (C.G.)
- Laboratory of Proteomics and Lipidomics, IRCCS Orthopedic Institute Galeazzi, 20161 Milano, Italy;
| | - Daniele Capitanio
- Department of Biomedical Sciences for Health, University of Milano, 20133 Milano, Italy; (M.M.); (C.G.)
| |
Collapse
|
16
|
Tam LM, Rand MD. Review: myogenic and muscle toxicity targets of environmental methylmercury exposure. Arch Toxicol 2024; 98:1645-1658. [PMID: 38546836 PMCID: PMC11105986 DOI: 10.1007/s00204-024-03724-3] [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: 12/12/2023] [Accepted: 02/29/2024] [Indexed: 05/01/2024]
Abstract
A number of environmental toxicants are noted for their activity that leads to declined motor function. However, the role of muscle as a proximal toxicity target organ for environmental agents has received considerably less attention than the toxicity targets in the nervous system. Nonetheless, the effects of conventional neurotoxicants on processes of myogenesis and muscle maintenance are beginning to resolve a concerted role of muscle as a susceptible toxicity target. A large body of evidence from epidemiological, animal, and in vitro studies has established that methylmercury (MeHg) is a potent developmental toxicant, with the nervous system being a preferred target. Despite its well-recognized status as a neurotoxicant, there is accumulating evidence that MeHg also targets muscle and neuromuscular development as well as contributes to the etiology of motor defects with prenatal MeHg exposure. Here, we summarize evidence for targets of MeHg in the morphogenesis and maintenance of skeletal muscle that reveal effects on MeHg distribution, myogenesis, myotube formation, myotendinous junction formation, neuromuscular junction formation, and satellite cell-mediated muscle repair. We briefly recapitulate the molecular and cellular mechanisms of skeletal muscle development and highlight the pragmatic role of alternative model organisms, Drosophila and zebrafish, in delineating the molecular underpinnings of muscle development and MeHg-mediated myotoxicity. Finally, we discuss how toxicity targets in muscle development may inform the developmental origins of health and disease theory to explain the etiology of environmentally induced adult motor deficits and accelerated decline in muscle fitness with aging.
Collapse
Affiliation(s)
- Lok Ming Tam
- Department of Environmental Medicine, University of Rochester School of Medicine and Dentistry, 601 Elmwood Ave, Rochester, NY, 14642, USA.
- Clinical and Translational Science Institute, University of Rochester Medical Center, 601 Elmwood Ave, Rochester, NY, 14642, USA.
| | - Matthew D Rand
- Department of Environmental Medicine, University of Rochester School of Medicine and Dentistry, 601 Elmwood Ave, Rochester, NY, 14642, USA
| |
Collapse
|
17
|
Nolt GL, Keeble AR, Wen Y, Strong AC, Thomas NT, Valentino TR, Brightwell CR, Murach KA, Patrizia S, Weinstabl H, Gollner A, McCarthy JJ, Fry CS, Franti M, Filareto A, Peterson CA, Dungan CM. Inhibition of p53-MDM2 binding reduces senescent cell abundance and improves the adaptive responses of skeletal muscle from aged mice. GeroScience 2024; 46:2153-2176. [PMID: 37872294 PMCID: PMC10828311 DOI: 10.1007/s11357-023-00976-2] [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: 07/24/2023] [Accepted: 10/09/2023] [Indexed: 10/25/2023] Open
Abstract
Skeletal muscle adaptation to external stimuli, such as regeneration following injury and hypertrophy in response to resistance exercise, are blunted with advanced age. The accumulation of senescent cells, along with defects in myogenic progenitor cell (MPC) proliferation, have been strongly linked as contributing factors to age-associated impairment in muscle adaptation. p53 plays an integral role in all these processes, as upregulation of p53 causes apoptosis in senescent cells and prevents mitotic catastrophe in MPCs from old mice. The goal of this study was to determine if a novel pharmaceutical agent (BI01), which functions by upregulating p53 through inhibition of binding to MDM2, the primary p53 regulatory protein, improves muscle regeneration and hypertrophy in old mice. BI01 effectively reduced the number of senescent cells in vitro but had no effect on MPC survival or proliferation at a comparable dose. Following repeated oral gavage with 2 mg/kg of BI01 (OS) or vehicle (OV), old mice (24 months) underwent unilateral BaCl2 injury in the tibialis anterior (TA) muscle, with PBS injections serving as controls. After 7 days, satellite cell number was higher in the TA of OS compared to OV mice, as was the expression of genes involved in ATP production. By 35 days, old mice treated with BI01 displayed reduced senescent cell burden, enhanced regeneration (higher muscle mass and fiber cross-sectional area) and restoration of muscle function relative to OV mice. To examine the impact of 2 mg/kg BI01 on muscle hypertrophy, the plantaris muscle was subjected to 28 days of mechanical overload (MOV) in OS and OV mice. In response to MOV, OS mice had larger plantaris muscles and muscle fibers than OV mice, particularly type 2b + x fibers, associated with reduced senescent cells. Together our data show that BI01 is an effective senolytic agent that may also augment muscle metabolism to enhance muscle regeneration and hypertrophy in old mice.
Collapse
Affiliation(s)
- Georgia L Nolt
- Department of Physiology, University of Kentucky, Lexington, KY, USA
- The Center for Muscle Biology, University of Kentucky, Lexington, KY, USA
| | - Alexander R Keeble
- Department of Physiology, University of Kentucky, Lexington, KY, USA
- The Center for Muscle Biology, University of Kentucky, Lexington, KY, USA
| | - Yuan Wen
- The Center for Muscle Biology, University of Kentucky, Lexington, KY, USA
- Department of Physical Therapy, University of Kentucky, Lexington, KY, USA
| | - Aubrey C Strong
- The Center for Muscle Biology, University of Kentucky, Lexington, KY, USA
| | - Nicholas T Thomas
- The Center for Muscle Biology, University of Kentucky, Lexington, KY, USA
- Department of Athletic Training and Clinical Nutrition, University of Kentucky, Lexington, KY, USA
| | - Taylor R Valentino
- Department of Physiology, University of Kentucky, Lexington, KY, USA
- The Center for Muscle Biology, University of Kentucky, Lexington, KY, USA
| | - Camille R Brightwell
- The Center for Muscle Biology, University of Kentucky, Lexington, KY, USA
- Department of Athletic Training and Clinical Nutrition, University of Kentucky, Lexington, KY, USA
| | - Kevin A Murach
- Department of Health, Human Performance, and Recreation, University of Arkansas, Fayetteville, AR, USA
| | - Sini Patrizia
- Regenerative Medicine, Boehringer Ingelheim Pharmaceuticals Inc., 900 Ridgebury Road, Ridgefield, CT, 06877, USA
| | - Harald Weinstabl
- Boehringer Ingelheim RCV, Boehringer Ingelheim Pharmaceuticals Inc., Vienna, Austria
| | - Andreas Gollner
- Boehringer Ingelheim RCV, Boehringer Ingelheim Pharmaceuticals Inc., Vienna, Austria
| | - John J McCarthy
- Department of Physiology, University of Kentucky, Lexington, KY, USA
- The Center for Muscle Biology, University of Kentucky, Lexington, KY, USA
| | - Christopher S Fry
- The Center for Muscle Biology, University of Kentucky, Lexington, KY, USA
- Department of Athletic Training and Clinical Nutrition, University of Kentucky, Lexington, KY, USA
| | - Michael Franti
- Regenerative Medicine, Boehringer Ingelheim Pharmaceuticals Inc., 900 Ridgebury Road, Ridgefield, CT, 06877, USA
| | - Antonio Filareto
- Regenerative Medicine, Boehringer Ingelheim Pharmaceuticals Inc., 900 Ridgebury Road, Ridgefield, CT, 06877, USA.
| | - Charlotte A Peterson
- The Center for Muscle Biology, University of Kentucky, Lexington, KY, USA
- Department of Physical Therapy, University of Kentucky, Lexington, KY, USA
| | - Cory M Dungan
- The Center for Muscle Biology, University of Kentucky, Lexington, KY, USA.
- Department of Physical Therapy, University of Kentucky, Lexington, KY, USA.
- Department of Health, Human Performance, and Recreation, Baylor University, One Bear Place #97313, Waco, TX, 76706, USA.
| |
Collapse
|
18
|
Chen S, Huang L, Liu B, Duan H, Li Z, Liu Y, Li H, Fu X, Lin J, Xu Y, Liu L, Wan D, Yin Y, Xie L. Dynamic changes in butyrate levels regulate satellite cell homeostasis by preventing spontaneous activation during aging. SCIENCE CHINA. LIFE SCIENCES 2024; 67:745-764. [PMID: 38157106 DOI: 10.1007/s11427-023-2400-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Accepted: 06/15/2023] [Indexed: 01/03/2024]
Abstract
The gut microbiota plays a pivotal role in systemic metabolic processes and in particular functions, such as developing and preserving the skeletal muscle system. However, the interplay between gut microbiota/metabolites and the regulation of satellite cell (SC) homeostasis, particularly during aging, remains elusive. We propose that gut microbiota and its metabolites modulate SC physiology and homeostasis throughout skeletal muscle development, regeneration, and aging process. Our investigation reveals that microbial dysbiosis manipulated by either antibiotic treatment or fecal microbiota transplantation from aged to adult mice, leads to the activation of SCs or a significant reduction in the total number. Furthermore, employing multi-omics (e.g., RNA-seq, 16S rRNA gene sequencing, and metabolomics) and bioinformatic analysis, we demonstrate that the reduced butyrate levels, alongside the gut microbial dysbiosis, could be the primary factor contributing to the reduction in the number of SCs and subsequent impairments during skeletal muscle aging. Meanwhile, butyrate supplementation can mitigate the antibiotics-induced SC activation irrespective of gut microbiota, potentially by inhibiting the proliferation and differentiation of SCs/myoblasts. The butyrate effect is likely facilitated through the monocarboxylate transporter 1 (Mct1), a lactate transporter enriched on membranes of SCs and myoblasts. As a result, butyrate could serve as an alternative strategy to enhance SC homeostasis and function during skeletal muscle aging. Our findings shed light on the potential application of microbial metabolites in maintaining SC homeostasis and preventing skeletal muscle aging.
Collapse
Affiliation(s)
- Shujie Chen
- Department of Endocrinology and Metabolism, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China
- State Key Laboratory of Applied Microbiology Southern China, Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, Guangdong Open Laboratory of Applied Microbiology, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, 510070, China
- Department of Rehabilitation Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510000, China
| | - Liujing Huang
- Department of Endocrinology and Metabolism, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China
- State Key Laboratory of Applied Microbiology Southern China, Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, Guangdong Open Laboratory of Applied Microbiology, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, 510070, China
| | - Bingdong Liu
- Department of Endocrinology and Metabolism, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China
- State Key Laboratory of Applied Microbiology Southern China, Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, Guangdong Open Laboratory of Applied Microbiology, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, 510070, China
| | - Huimin Duan
- State Key Laboratory of Applied Microbiology Southern China, Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, Guangdong Open Laboratory of Applied Microbiology, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, 510070, China
- Department of Rehabilitation Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510000, China
| | - Ze Li
- State Key Laboratory of Applied Microbiology Southern China, Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, Guangdong Open Laboratory of Applied Microbiology, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, 510070, China
- School of Public Health, Xinxiang Medical University, Xinxiang, 453003, China
| | - Yifan Liu
- State Key Laboratory of Applied Microbiology Southern China, Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, Guangdong Open Laboratory of Applied Microbiology, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, 510070, China
- Institute of Aging Research, Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, School of Medical Technology, Guangdong Medical University, Dongguan, 524023, China
| | - Hu Li
- Bioland Laboratory, Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou, 510005, China
| | - Xiang Fu
- Bioland Laboratory, Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou, 510005, China
| | - Jingchao Lin
- Metabo-Profile Biotechnology (Shanghai) Co. Ltd., Shanghai, 201315, China
| | - Yinlan Xu
- School of Public Health, Xinxiang Medical University, Xinxiang, 453003, China
| | - Li Liu
- Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Dan Wan
- Institute of Aging Research, Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, School of Medical Technology, Guangdong Medical University, Dongguan, 524023, China.
- Laboratory of Animal Nutritional Physiology and Metabolic Process, Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, 410125, China.
| | - Yulong Yin
- Institute of Aging Research, Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, School of Medical Technology, Guangdong Medical University, Dongguan, 524023, China.
- Laboratory of Animal Nutritional Physiology and Metabolic Process, Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, 410125, China.
| | - Liwei Xie
- Department of Endocrinology and Metabolism, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China.
- State Key Laboratory of Applied Microbiology Southern China, Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, Guangdong Open Laboratory of Applied Microbiology, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, 510070, China.
- School of Public Health, Xinxiang Medical University, Xinxiang, 453003, China.
- Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China.
| |
Collapse
|
19
|
Fard D, Barbiera A, Dobrowolny G, Tamagnone L, Scicchitano BM. Semaphorins: Missing Signals in Age-dependent Alteration of Neuromuscular Junctions and Skeletal Muscle Regeneration. Aging Dis 2024; 15:517-534. [PMID: 37728580 PMCID: PMC10917540 DOI: 10.14336/ad.2023.0801] [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: 05/03/2023] [Accepted: 08/01/2023] [Indexed: 09/21/2023] Open
Abstract
Skeletal muscle is characterized by a remarkable capacity to rearrange after physiological changes and efficiently regenerate. However, during aging, extensive injury, or pathological conditions, the complete regenerative program is severely affected, with a progressive loss of muscle mass and function, a condition known as sarcopenia. The compromised tissue repair program is attributable to the gradual depletion of stem cells and to altered regulatory signals. Defective muscle regeneration can severely affect re-innervation by motor axons, and neuromuscular junctions (NMJs) development, ultimately leading to skeletal muscle atrophy. Defects in NMJ formation and maintenance occur physiologically during aging and are responsible for the pathogenesis of several neuromuscular disorders. However, it is still largely unknown how neuromuscular connections are restored on regenerating fibers. It has been suggested that attractive and repelling signals used for axon guidance could be implicated in this process; in particular, guidance molecules called semaphorins play a key role. Semaphorins are a wide family of extracellular regulatory signals with a multifaceted role in cell-cell communication. Originally discovered as axon guidance factors, they have been implicated in cancer progression, embryonal organogenesis, skeletal muscle innervation, and other physiological and developmental functions in different tissues. In particular, in skeletal muscle, specific semaphorin molecules are involved in the restoration and remodeling of the nerve-muscle connections, thus emphasizing their plausible role to ensure the success of muscle regeneration. This review article aims to discuss the impact of aging on skeletal muscle regeneration and NMJs remodeling and will highlight the most recent insights about the role of semaphorins in this context.
Collapse
Affiliation(s)
- Damon Fard
- Sezione di Istologia ed Embriologia, Dipartimento di Scienze della Vita e Sanità Pubblica,Università Cattolica del Sacro Cuore, 00168 Roma, Italy.
| | - Alessandra Barbiera
- Sezione di Istologia ed Embriologia, Dipartimento di Scienze della Vita e Sanità Pubblica,Università Cattolica del Sacro Cuore, 00168 Roma, Italy.
| | - Gabriella Dobrowolny
- DAHFMO-Unità di Istologia ed Embriologia Medica, Sapienza Università di Roma, 00161 Roma, Italy.
| | - Luca Tamagnone
- Sezione di Istologia ed Embriologia, Dipartimento di Scienze della Vita e Sanità Pubblica,Università Cattolica del Sacro Cuore, 00168 Roma, Italy.
- Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Roma, Italy.
| | - Bianca Maria Scicchitano
- Sezione di Istologia ed Embriologia, Dipartimento di Scienze della Vita e Sanità Pubblica,Università Cattolica del Sacro Cuore, 00168 Roma, Italy.
- Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Roma, Italy.
| |
Collapse
|
20
|
Cisterna B, Malatesta M. Molecular and Structural Alterations of Skeletal Muscle Tissue Nuclei during Aging. Int J Mol Sci 2024; 25:1833. [PMID: 38339110 PMCID: PMC10855217 DOI: 10.3390/ijms25031833] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Revised: 01/30/2024] [Accepted: 01/31/2024] [Indexed: 02/12/2024] Open
Abstract
Aging is accompanied by a progressive loss of skeletal muscle mass and strength. The mechanisms underlying this phenomenon are certainly multifactorial and still remain to be fully elucidated. Changes in the cell nucleus structure and function have been considered among the possible contributing causes. This review offers an overview of the current knowledge on skeletal muscle nuclei in aging, focusing on the impairment of nuclear pathways potentially involved in age-related muscle decline. In skeletal muscle two types of cells are present: fiber cells, constituting the contractile muscle mass and containing hundreds of myonuclei, and the satellite cells, i.e., the myogenic mononuclear stem cells occurring at the periphery of the fibers and responsible for muscle growth and repair. Research conducted on different experimental models and with different methodological approaches demonstrated that both the myonuclei and satellite cell nuclei of aged skeletal muscles undergo several structural and molecular alterations, affecting chromatin organization, gene expression, and transcriptional and post-transcriptional activities. These alterations play a key role in the impairment of muscle fiber homeostasis and regeneration, thus contributing to the age-related decrease in skeletal muscle mass and function.
Collapse
Affiliation(s)
| | - Manuela Malatesta
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Strada Le Grazie 8, 37134 Verona, Italy;
| |
Collapse
|
21
|
Thompson SD, Barrett KL, Rugel CL, Redmond R, Rudofski A, Kurian J, Curtin JL, Dayanidhi S, Lavasani M. Sex-specific preservation of neuromuscular function and metabolism following systemic transplantation of multipotent adult stem cells in a murine model of progeria. GeroScience 2024; 46:1285-1302. [PMID: 37535205 PMCID: PMC10828301 DOI: 10.1007/s11357-023-00892-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Accepted: 07/25/2023] [Indexed: 08/04/2023] Open
Abstract
Onset and rates of sarcopenia, a disease characterized by a loss of muscle mass and function with age, vary greatly between sexes. Currently, no clinical interventions successfully arrest age-related muscle impairments since the decline is frequently multifactorial. Previously, we found that systemic transplantation of our unique adult multipotent muscle-derived stem/progenitor cells (MDSPCs) isolated from young mice-but not old-extends the health-span in DNA damage mouse models of progeria, a disease of accelerated aging. Additionally, induced neovascularization in the muscles and brain-where no transplanted cells were detected-strongly suggests a systemic therapeutic mechanism, possibly activated through circulating secreted factors. Herein, we used ZMPSTE24-deficient mice, a lamin A defect progeria model, to investigate the ability of young MDSPCs to preserve neuromuscular tissue structure and function. We show that progeroid ZMPST24-deficient mice faithfully exhibit sarcopenia and age-related metabolic dysfunction. However, systemic transplantation of young MDSPCs into ZMPSTE24-deficient progeroid mice sustained healthy function and histopathology of muscular tissues throughout their 6-month life span in a sex-specific manner. Indeed, female-but not male-mice systemically transplanted with young MDSPCs demonstrated significant preservation of muscle endurance, muscle fiber size, mitochondrial respirometry, and neuromuscular junction morphometrics. These novel findings strongly suggest that young MDSPCs modulate the systemic environment of aged animals by secreted rejuvenating factors to maintain a healthy homeostasis in a sex-specific manner and that the female muscle microenvironment remains responsive to exogenous regenerative cues in older age. This work highlights the age- and sex-related differences in neuromuscular tissue degeneration and the future prospect of preserving health in older adults with systemic regenerative treatments.
Collapse
Affiliation(s)
- Seth D Thompson
- Shirley Ryan AbilityLab, 355 E. Erie St, Chicago, IL, 60611, USA.
- Department of Physical Medicine and Rehabilitation, Northwestern University, Chicago, IL, 60611, USA.
- Northwestern University Interdepartmental Neuroscience (NUIN) Graduate Program, Northwestern University, Chicago, IL, 60611, USA.
| | - Kelsey L Barrett
- Shirley Ryan AbilityLab, 355 E. Erie St, Chicago, IL, 60611, USA
| | - Chelsea L Rugel
- Shirley Ryan AbilityLab, 355 E. Erie St, Chicago, IL, 60611, USA
- Department of Physical Medicine and Rehabilitation, Northwestern University, Chicago, IL, 60611, USA
- Northwestern University Interdepartmental Neuroscience (NUIN) Graduate Program, Northwestern University, Chicago, IL, 60611, USA
| | - Robin Redmond
- Shirley Ryan AbilityLab, 355 E. Erie St, Chicago, IL, 60611, USA
| | - Alexia Rudofski
- Shirley Ryan AbilityLab, 355 E. Erie St, Chicago, IL, 60611, USA
| | - Jacob Kurian
- Department of Biomedical Engineering, Northwestern University, Chicago, IL, 60611, USA
| | - Jodi L Curtin
- Shirley Ryan AbilityLab, 355 E. Erie St, Chicago, IL, 60611, USA
| | - Sudarshan Dayanidhi
- Shirley Ryan AbilityLab, 355 E. Erie St, Chicago, IL, 60611, USA
- Department of Physical Medicine and Rehabilitation, Northwestern University, Chicago, IL, 60611, USA
| | - Mitra Lavasani
- Shirley Ryan AbilityLab, 355 E. Erie St, Chicago, IL, 60611, USA.
- Department of Physical Medicine and Rehabilitation, Northwestern University, Chicago, IL, 60611, USA.
- Northwestern University Interdepartmental Neuroscience (NUIN) Graduate Program, Northwestern University, Chicago, IL, 60611, USA.
| |
Collapse
|
22
|
Li DCW, Rudloff S, Langer HT, Norman K, Herpich C. Age-Associated Differences in Recovery from Exercise-Induced Muscle Damage. Cells 2024; 13:255. [PMID: 38334647 PMCID: PMC10854791 DOI: 10.3390/cells13030255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2023] [Revised: 01/23/2024] [Accepted: 01/24/2024] [Indexed: 02/10/2024] Open
Abstract
Understanding the intricate mechanisms governing the cellular response to resistance exercise is paramount for promoting healthy aging. This narrative review explored the age-related alterations in recovery from resistance exercise, focusing on the nuanced aspects of exercise-induced muscle damage in older adults. Due to the limited number of studies in older adults that attempt to delineate age differences in muscle discovery, we delve into the multifaceted cellular influences of chronic low-grade inflammation, modifications in the extracellular matrix, and the role of lipid mediators in shaping the recovery landscape in aging skeletal muscle. From our literature search, it is evident that aged muscle displays delayed, prolonged, and inefficient recovery. These changes can be attributed to anabolic resistance, the stiffening of the extracellular matrix, mitochondrial dysfunction, and unresolved inflammation as well as alterations in satellite cell function. Collectively, these age-related impairments may impact subsequent adaptations to resistance exercise. Insights gleaned from this exploration may inform targeted interventions aimed at enhancing the efficacy of resistance training programs tailored to the specific needs of older adults, ultimately fostering healthy aging and preserving functional independence.
Collapse
Affiliation(s)
- Donna Ching Wah Li
- Department of Nutrition and Gerontology, German Institute of Human Nutrition Potsdam-Rehbrücke, 14558 Nuthetal, Germany
- Institute of Nutritional Science, University of Potsdam, 14558 Nuthetal, Germany
| | - Stefan Rudloff
- Department of Geriatrics and Medical Gerontology, Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 13347 Berlin, Germany
| | | | - Kristina Norman
- Department of Nutrition and Gerontology, German Institute of Human Nutrition Potsdam-Rehbrücke, 14558 Nuthetal, Germany
- Institute of Nutritional Science, University of Potsdam, 14558 Nuthetal, Germany
- Department of Geriatrics and Medical Gerontology, Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 13347 Berlin, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Berlin, 10785 Berlin, Germany
| | - Catrin Herpich
- Department of Nutrition and Gerontology, German Institute of Human Nutrition Potsdam-Rehbrücke, 14558 Nuthetal, Germany
- Institute of Nutritional Science, University of Potsdam, 14558 Nuthetal, Germany
- Department of Geriatrics and Medical Gerontology, Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 13347 Berlin, Germany
| |
Collapse
|
23
|
Hidalgo-Alvarez V, Madl CM. Leveraging Biomaterial Platforms to Study Aging-Related Neural and Muscular Degeneration. Biomolecules 2024; 14:69. [PMID: 38254669 PMCID: PMC10813704 DOI: 10.3390/biom14010069] [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: 12/11/2023] [Revised: 12/28/2023] [Accepted: 12/30/2023] [Indexed: 01/24/2024] Open
Abstract
Aging is a complex multifactorial process that results in tissue function impairment across the whole organism. One of the common consequences of this process is the loss of muscle mass and the associated decline in muscle function, known as sarcopenia. Aging also presents with an increased risk of developing other pathological conditions such as neurodegeneration. Muscular and neuronal degeneration cause mobility issues and cognitive impairment, hence having a major impact on the quality of life of the older population. The development of novel therapies that can ameliorate the effects of aging is currently hindered by our limited knowledge of the underlying mechanisms and the use of models that fail to recapitulate the structure and composition of the cell microenvironment. The emergence of bioengineering techniques based on the use of biomimetic materials and biofabrication methods has opened the possibility of generating 3D models of muscular and nervous tissues that better mimic the native extracellular matrix. These platforms are particularly advantageous for drug testing and mechanistic studies. In this review, we discuss the developments made in the creation of 3D models of aging-related neuronal and muscular degeneration and we provide a perspective on the future directions for the field.
Collapse
Affiliation(s)
| | - Christopher M. Madl
- Department of Materials Science and Engineering, University of Pennsylvania, Philadelphia, PA 19104, USA;
| |
Collapse
|
24
|
Yılmaz D, Mathavan N, Wehrle E, Kuhn GA, Müller R. Mouse models of accelerated aging in musculoskeletal research for assessing frailty, sarcopenia, and osteoporosis - A review. Ageing Res Rev 2024; 93:102118. [PMID: 37935249 DOI: 10.1016/j.arr.2023.102118] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 10/01/2023] [Accepted: 11/03/2023] [Indexed: 11/09/2023]
Abstract
Musculoskeletal aging encompasses the decline in bone and muscle function, leading to conditions such as frailty, osteoporosis, and sarcopenia. Unraveling the underlying molecular mechanisms and developing effective treatments are crucial for improving the quality of life for those affected. In this context, accelerated aging models offer valuable insights into these conditions by displaying the hallmarks of human aging. Herein, this review focuses on relevant mouse models of musculoskeletal aging with particular emphasis on frailty, osteoporosis, and sarcopenia. Among the discussed models, PolgA mice in particular exhibit hallmarks of musculoskeletal aging, presenting early-onset frailty, as well as reduced bone and muscle mass that closely resemble human musculoskeletal aging. Ultimately, findings from these models hold promise for advancing interventions targeted at age-related musculoskeletal disorders, effectively addressing the challenges posed by musculoskeletal aging and associated conditions in humans.
Collapse
Affiliation(s)
- Dilara Yılmaz
- Institute for Biomechanics, ETH Zurich, Zurich, Switzerland
| | | | - Esther Wehrle
- Institute for Biomechanics, ETH Zurich, Zurich, Switzerland; AO Research Institute Davos, Davos Platz, Switzerland
| | - Gisela A Kuhn
- Institute for Biomechanics, ETH Zurich, Zurich, Switzerland
| | - Ralph Müller
- Institute for Biomechanics, ETH Zurich, Zurich, Switzerland.
| |
Collapse
|
25
|
Careccia G, Mangiavini L, Cirillo F. Regulation of Satellite Cells Functions during Skeletal Muscle Regeneration: A Critical Step in Physiological and Pathological Conditions. Int J Mol Sci 2023; 25:512. [PMID: 38203683 PMCID: PMC10778731 DOI: 10.3390/ijms25010512] [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: 10/26/2023] [Revised: 12/20/2023] [Accepted: 12/28/2023] [Indexed: 01/12/2024] Open
Abstract
Skeletal muscle regeneration is a complex process involving the generation of new myofibers after trauma, competitive physical activity, or disease. In this context, adult skeletal muscle stem cells, also known as satellite cells (SCs), play a crucial role in regulating muscle tissue homeostasis and activating regeneration. Alterations in their number or function have been associated with various pathological conditions. The main factors involved in the dysregulation of SCs' activity are inflammation, oxidative stress, and fibrosis. This review critically summarizes the current knowledge on the role of SCs in skeletal muscle regeneration. It examines the changes in the activity of SCs in three of the most common and severe muscle disorders: sarcopenia, muscular dystrophy, and cancer cachexia. Understanding the molecular mechanisms involved in their dysregulations is essential for improving current treatments, such as exercise, and developing personalized approaches to reactivate SCs.
Collapse
Affiliation(s)
- Giorgia Careccia
- Department of Biosciences, University of Milan, 20133 Milan, Italy;
| | - Laura Mangiavini
- IRCCS Istituto Ortopedico Galeazzi, 20161 Milan, Italy;
- Department of Biomedical Sciences for Health, University of Milan, 20133 Milan, Italy
| | - Federica Cirillo
- IRCCS Policlinico San Donato, 20097 San Donato Milanese, Italy
- Institute for Molecular and Translational Cardiology (IMTC), 20097 San Donato Milanese, Italy
| |
Collapse
|
26
|
Motohashi N, Minegishi K, Aoki Y. Inherited myogenic abilities in muscle precursor cells defined by the mitochondrial complex I-encoding protein. Cell Death Dis 2023; 14:689. [PMID: 37857600 PMCID: PMC10587152 DOI: 10.1038/s41419-023-06192-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 09/18/2023] [Accepted: 09/28/2023] [Indexed: 10/21/2023]
Abstract
Skeletal muscle comprises different muscle fibers, including slow- and fast-type muscles, and satellite cells (SCs), which exist in individual muscle fibers and possess different myogenic properties. Previously, we reported that myoblasts (MBs) from slow-type enriched soleus (SOL) had a high potential to self-renew compared with cells derived from fast-type enriched tibialis anterior (TA). However, whether the functionality of myogenic cells in adult muscles is attributed to the muscle fiber in which they reside and whether the characteristics of myogenic cells derived from slow- and fast-type fibers can be distinguished at the genetic level remain unknown. Global gene expression analysis revealed that the myogenic potential of MBs was independent of the muscle fiber type they reside in but dependent on the region of muscles they are derived from. Thus, in this study, proteomic analysis was conducted to clarify the molecular differences between MBs derived from TA and SOL. NADH dehydrogenase (ubiquinone) iron-sulfur protein 8 (Ndufs8), a subunit of NADH dehydrogenase in mitochondrial complex I, significantly increased in SOL-derived MBs compared with that in TA-derived cells. Moreover, the expression level of Ndufs8 in MBs significantly decreased with age. Gain- and loss-of-function experiments revealed that Ndufs8 expression in MBs promoted differentiation, self-renewal, and apoptosis resistance. In particular, Ndufs8 suppression in MBs increased p53 acetylation, followed by a decline in NAD/NADH ratio. Nicotinamide mononucleotide treatment, which restores the intracellular NAD+ level, could decrease p53 acetylation and increase myogenic cell self-renewal ability in vivo. These results suggested that the functional differences in MBs derived from SOL and TA governed by the mitochondrial complex I-encoding gene reflect the magnitude of the decline in SC number observed with aging, indicating that the replenishment of NAD+ is a possible approach for improving impaired cellular functions caused by aging or diseases.
Collapse
Affiliation(s)
- Norio Motohashi
- Department of Molecular Therapy, National Institute of Neuroscience, National Center of Neurology and Psychiatry (NCNP), Tokyo, 187-8502, Japan.
| | - Katsura Minegishi
- Department of Molecular Therapy, National Institute of Neuroscience, National Center of Neurology and Psychiatry (NCNP), Tokyo, 187-8502, Japan
| | - Yoshitsugu Aoki
- Department of Molecular Therapy, National Institute of Neuroscience, National Center of Neurology and Psychiatry (NCNP), Tokyo, 187-8502, Japan.
| |
Collapse
|
27
|
Barsky ST, Monks DA. Androgen action on myogenesis throughout the lifespan; comparison with neurogenesis. Front Neuroendocrinol 2023; 71:101101. [PMID: 37669703 DOI: 10.1016/j.yfrne.2023.101101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 08/31/2023] [Accepted: 09/02/2023] [Indexed: 09/07/2023]
Abstract
Androgens' pleiotropic actions in promoting sex differences present not only a challenge to providing a comprehensive account of their function, but also an opportunity to gain insights by comparing androgenic actions across organ systems. Although often overlooked by neuroscientists, skeletal muscle is another androgen-responsive organ system which shares with the nervous system properties of electrochemical excitability, behavioral relevance, and remarkable capacity for adaptive plasticity. Here we review androgenic regulation of mitogenic plasticity in skeletal muscle with the goal of identifying areas of interest to those researching androgenic mechanisms mediating sexual differentiation of neurogenesis. We use an organizational-activational framework to relate broad areas of similarity and difference between androgen effects on mitogenesis in muscle and brain throughout the lifespan, from early organogenesis, through pubertal organization, adult activation, and aging. The focus of the review is androgenic regulation of muscle-specific stem cells (satellite cells), which share with neural stem cells essential functions in development, plasticity, and repair, albeit with distinct, muscle-specific features. Also considered are areas of paracrine and endocrine interaction between androgen action on muscle and nervous system, including mediation of neural plasticity of innervating and distal neural populations by muscle-produced trophic factors.
Collapse
Affiliation(s)
- Sabrina Tzivia Barsky
- Department of Cell & Systems Biology, Faculty of Arts & Science, University of Toronto, Toronto, Ontario, Canada.
| | - Douglas Ashley Monks
- Department of Cell & Systems Biology, Faculty of Arts & Science, University of Toronto, Toronto, Ontario, Canada; Department of Psychology, Faculty of Arts & Science, University of Toronto Mississauga, Mississauga, Ontario, Canada.
| |
Collapse
|
28
|
Wajda A, Bogucka D, Stypińska B, Radkowski MJ, Targowski T, Dudek E, Kmiołek T, Modzelewska E, Paradowska-Gorycka A. Expression of Prostaglandin Genes and β-Catenin in Whole Blood as Potential Markers of Muscle Degeneration. Int J Mol Sci 2023; 24:12885. [PMID: 37629065 PMCID: PMC10454559 DOI: 10.3390/ijms241612885] [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: 08/02/2023] [Revised: 08/09/2023] [Accepted: 08/12/2023] [Indexed: 08/27/2023] Open
Abstract
Prostaglandin signaling pathways are closely related to inflammation, but also muscle regeneration and processes associated with frailty and sarcopenia, whereas β-catenin (CTNNB1 gene) as a part of Wnt signaling is also involved in the differentiation of muscle cells and fibrosis. The present study analyzed the association between selected prostaglandin pathway genes and clinical parameters in patients with sarcopenia and frailty syndrome. The present study was conducted on patients with sarcopenia, frailty syndrome, and control older patients (N = 25). Additionally, two healthy controls at the age of 25-30 years (N = 51) and above 50 years old (N = 42) were included. The expression of the PTRGER4, PTGES2 (COX2), PTGS2, and CTNNB1 genes in whole blood was checked by the qPCR method. The serum cytokine levels (IL-10, TNFα, IFN-y, IL-1α, IL-1β) in patients and controls were checked by the Q-Plex Human Cytokine Panel. The results showed a significant effect of age on PTGER4 gene expression (p = 0.01). A negative trend between the appendicular skeletal muscle mass parameter (ASSM) and the expression of PTGER4 has been noted (r = -0.224, p = 0.484). PTGES2 and PTGS2 expressions negatively correlated with creatine phosphokinase (r = -0.71, p = 0.009; r = -0.58, p = 0.047) and positively with the functional mobility test timed up and go scale (TUG) (r = 0.61, p = 0.04; r = 0.63, p = 0.032). In the older control group, a negative association between iron levels and the expression of PTGS2 (r = -0.47, p = 0.017) was observed. A similar tendency was noted in patients with sarcopenia (r = -0.112, p = 0.729). A negative trend between appendicular skeletal muscle mass (ASMM) and PTGER4 seems to confirm the impairment of muscle regeneration associated with sarcopenia. The expression of the studied genes revealed a trend in associations with the clinical picture of muscular dystrophy and weakening patients. Perhaps PTGS2 and PTGES2 is in opposition to the role of the PTGER4 receptor in muscle physiology. Nevertheless, further, including functional studies is needed.
Collapse
Affiliation(s)
- Anna Wajda
- Department of Molecular Biology, National Institute of Geriatrics, Rheumatology and Rehabilitation, 02-637 Warsaw, Poland; (D.B.); (B.S.); (E.D.); (T.K.); (E.M.); (A.P.-G.)
| | - Diana Bogucka
- Department of Molecular Biology, National Institute of Geriatrics, Rheumatology and Rehabilitation, 02-637 Warsaw, Poland; (D.B.); (B.S.); (E.D.); (T.K.); (E.M.); (A.P.-G.)
| | - Barbara Stypińska
- Department of Molecular Biology, National Institute of Geriatrics, Rheumatology and Rehabilitation, 02-637 Warsaw, Poland; (D.B.); (B.S.); (E.D.); (T.K.); (E.M.); (A.P.-G.)
| | - Marcin Jerzy Radkowski
- Department of Geriatrics, National Institute of Geriatrics, Rheumatology and Rehabilitation, 02-637 Warsaw, Poland; (M.J.R.); (T.T.)
| | - Tomasz Targowski
- Department of Geriatrics, National Institute of Geriatrics, Rheumatology and Rehabilitation, 02-637 Warsaw, Poland; (M.J.R.); (T.T.)
| | - Ewa Dudek
- Department of Molecular Biology, National Institute of Geriatrics, Rheumatology and Rehabilitation, 02-637 Warsaw, Poland; (D.B.); (B.S.); (E.D.); (T.K.); (E.M.); (A.P.-G.)
| | - Tomasz Kmiołek
- Department of Molecular Biology, National Institute of Geriatrics, Rheumatology and Rehabilitation, 02-637 Warsaw, Poland; (D.B.); (B.S.); (E.D.); (T.K.); (E.M.); (A.P.-G.)
| | - Ewa Modzelewska
- Department of Molecular Biology, National Institute of Geriatrics, Rheumatology and Rehabilitation, 02-637 Warsaw, Poland; (D.B.); (B.S.); (E.D.); (T.K.); (E.M.); (A.P.-G.)
| | - Agnieszka Paradowska-Gorycka
- Department of Molecular Biology, National Institute of Geriatrics, Rheumatology and Rehabilitation, 02-637 Warsaw, Poland; (D.B.); (B.S.); (E.D.); (T.K.); (E.M.); (A.P.-G.)
| |
Collapse
|
29
|
Nùñez-Lisboa M, Valero-Breton M, Dewolf AH. Unraveling age-related impairment of the neuromuscular system: exploring biomechanical and neurophysiological perspectives. Front Physiol 2023; 14:1194889. [PMID: 37427405 PMCID: PMC10323685 DOI: 10.3389/fphys.2023.1194889] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Accepted: 06/14/2023] [Indexed: 07/11/2023] Open
Abstract
With extended life expectancy, the quality of life of elders is a priority. Loss of mobility, increased morbidity and risks of falls have dramatic individual and societal impacts. Here we consider the age-related modifications of gait, from a biomechanical and neurophysiological perspective. Among the many factors of frailty involved (e.g., metabolic, hormonal, immunological), loss of muscle strength and neurodegenerative changes inducing slower muscle contraction may play a key role. We highlight that the impact of the multifactorial age-related changes in the neuromuscular systems results in common features of gait in the immature gait of infants and older adults. Besides, we also consider the reversibility of age-related neuromuscular deterioration by, on the one hand, exercise training, and the other hand, novel techniques such as direct spinal stimulation (tsDCS).
Collapse
Affiliation(s)
- M. Nùñez-Lisboa
- Laboratoire de Biomécanique et Physiologie et la Locomotion, Institute of Neuroscience, Louvain-la-Neuve, Belgium
- Exercise Science Laboratory, School of Kinesiology, Faculty of Medicine, Universidad Finis Terrae, Santiago, Chile
| | - M. Valero-Breton
- Exercise Science Laboratory, School of Kinesiology, Faculty of Medicine, Universidad Finis Terrae, Santiago, Chile
| | - A. H. Dewolf
- Laboratoire de Biomécanique et Physiologie et la Locomotion, Institute of Neuroscience, Louvain-la-Neuve, Belgium
| |
Collapse
|
30
|
Kurland JV, Cutler AA, Stanley JT, Betta ND, Van Deusen A, Pawlikowski B, Hall M, Antwine T, Russell A, Allen MA, Dowell R, Olwin B. Aging disrupts gene expression timing during muscle regeneration. Stem Cell Reports 2023; 18:1325-1339. [PMID: 37315524 PMCID: PMC10277839 DOI: 10.1016/j.stemcr.2023.05.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 05/08/2023] [Accepted: 05/09/2023] [Indexed: 06/16/2023] Open
Abstract
Skeletal muscle function and regenerative capacity decline during aging, yet factors driving these changes are incompletely understood. Muscle regeneration requires temporally coordinated transcriptional programs to drive myogenic stem cells to activate, proliferate, fuse to form myofibers, and to mature as myonuclei, restoring muscle function after injury. We assessed global changes in myogenic transcription programs distinguishing muscle regeneration in aged mice from young mice by comparing pseudotime trajectories from single-nucleus RNA sequencing of myogenic nuclei. Aging-specific differences in coordinating myogenic transcription programs necessary for restoring muscle function occur following muscle injury, likely contributing to compromised regeneration in aged mice. Differences in pseudotime alignment of myogenic nuclei when comparing aged with young mice via dynamic time warping revealed pseudotemporal differences becoming progressively more severe as regeneration proceeds. Disruptions in timing of myogenic gene expression programs may contribute to incomplete skeletal muscle regeneration and declines in muscle function as organisms age.
Collapse
Affiliation(s)
- Jesse V Kurland
- Department of Molecular, Cellular and Developmental Biology, University of Colorado, Boulder, CO 80309, USA
| | - Alicia A Cutler
- Department of Molecular, Cellular and Developmental Biology, University of Colorado, Boulder, CO 80309, USA
| | - Jacob T Stanley
- BioFrontiers Institute, University of Colorado, Boulder, 3415 Colorado Avenue, Boulder, CO 80303, USA
| | - Nicole Dalla Betta
- Department of Molecular, Cellular and Developmental Biology, University of Colorado, Boulder, CO 80309, USA
| | - Ashleigh Van Deusen
- Department of Molecular, Cellular and Developmental Biology, University of Colorado, Boulder, CO 80309, USA; Edgewise Therapeutics, 3415 Colorado Avenue, Boulder, CO 80303, USA
| | - Brad Pawlikowski
- Department of Molecular, Cellular and Developmental Biology, University of Colorado, Boulder, CO 80309, USA; Department of Pediatrics Section of Section of Hematology, Oncology, Bone Marrow Transplant, University of Colorado, Anschutz Medical Campus, Aurora, CO, USA
| | - Monica Hall
- Department of Molecular, Cellular and Developmental Biology, University of Colorado, Boulder, CO 80309, USA; Department of Microbiology and Cell Biology, Montana State University, Bozeman, MT 59718, USA
| | - Tiffany Antwine
- Department of Molecular, Cellular and Developmental Biology, University of Colorado, Boulder, CO 80309, USA
| | - Alan Russell
- Edgewise Therapeutics, 3415 Colorado Avenue, Boulder, CO 80303, USA
| | - Mary Ann Allen
- BioFrontiers Institute, University of Colorado, Boulder, 3415 Colorado Avenue, Boulder, CO 80303, USA
| | - Robin Dowell
- Department of Molecular, Cellular and Developmental Biology, University of Colorado, Boulder, CO 80309, USA; BioFrontiers Institute, University of Colorado, Boulder, 3415 Colorado Avenue, Boulder, CO 80303, USA.
| | - Bradley Olwin
- Department of Molecular, Cellular and Developmental Biology, University of Colorado, Boulder, CO 80309, USA.
| |
Collapse
|
31
|
Fahrner A, Luca E, Krützfeldt J. microRNA-501 controls myogenin +/CD74 + myogenic progenitor cells during muscle regeneration. Mol Metab 2023; 71:101704. [PMID: 36907509 PMCID: PMC10033748 DOI: 10.1016/j.molmet.2023.101704] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2022] [Revised: 03/02/2023] [Accepted: 03/05/2023] [Indexed: 03/14/2023] Open
Abstract
OBJECTIVE Skeletal muscle regeneration is markedly impaired during aging. How adult muscle stem cells contribute to this decrease in regenerative capacity is incompletely understood. We investigated mechanisms of age-related changes in myogenic progenitor cells using the tissue-specific microRNA 501. METHODS Young and old C57Bl/6 mice were used (3 months or 24 months of age, respectively) with or without global or tissue-specific genetic deletion of miR-501. Muscle regeneration was induced using intramuscular cardiotoxin injection or treadmill exercise and analysed using single cell and bulk RNA sequencing, qRT-PCR and immunofluorescence. Muscle fiber damage was assessed with Evan`s blue dye (EBD). In vitro analysis was performed in primary muscle cells obtained from mice and humans. RESULTS Single cell sequencing revealed myogenic progenitor cells in miR-501 knockout mice at day 6 after muscle injury that are characterized by high levels of myogenin and CD74. In control mice these cells were less in number and already downregulated after day 3 of muscle injury. Muscle from knockout mice had reduced myofiber size and reduced myofiber resilience to injury and exercise. miR-501 elicits this effect by regulating sarcomeric gene expression through its target gene estrogen-related receptor gamma (Esrrg). Importantly, in aged skeletal muscle where miR-501 was significantly downregulated and its target Esrrg significantly upregulated, the number of myog+/CD74+ cells during regeneration was upregulated to similar levels as observed in 501 knockout mice. Moreover, myog+/CD74+-aged skeletal muscle exhibited a similar decrease in the size of newly formed myofibers and increased number of necrotic myofibers after injury as observed in mice lacking miR-501. CONCLUSIONS miR-501 and Esrrg are regulated in muscle with decreased regenerative capacity and loss of miR-501 is permissive to the appearance of CD74+ myogenic progenitors. Our data uncover a novel link between the metabolic transcription factor Esrrg and sarcomere formation and demonstrate that stem cell heterogeneity in skeletal muscle during aging is under miRNA control. Targeting Esrrg or myog+/CD74+ progenitor cells might improve fiber size and myofiber resilience to exercise in aged skeletal muscle.
Collapse
Affiliation(s)
- Alexandra Fahrner
- Division of Endocrinology, Diabetes, and Clinical Nutrition, University Hospital Zurich, 8091, Zurich, Switzerland; Life Science Zurich Graduate School, Biomedicine, University of Zurich, 8057, Zurich, Switzerland
| | - Edlira Luca
- Division of Endocrinology, Diabetes, and Clinical Nutrition, University Hospital Zurich, 8091, Zurich, Switzerland
| | - Jan Krützfeldt
- Division of Endocrinology, Diabetes, and Clinical Nutrition, University Hospital Zurich, 8091, Zurich, Switzerland; Life Science Zurich Graduate School, Biomedicine, University of Zurich, 8057, Zurich, Switzerland.
| |
Collapse
|
32
|
Sahinyan K, Lazure F, Blackburn DM, Soleimani VD. Decline of regenerative potential of old muscle stem cells: contribution to muscle aging. FEBS J 2023; 290:1267-1289. [PMID: 35029021 DOI: 10.1111/febs.16352] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 12/23/2021] [Accepted: 01/11/2022] [Indexed: 01/01/2023]
Abstract
Muscle stem cells (MuSCs) are required for life-long muscle regeneration. In general, aging has been linked to a decline in the numbers and the regenerative potential of MuSCs. Muscle regeneration depends on the proper functioning of MuSCs, which is itself dependent on intricate interactions with its niche components. Aging is associated with both cell-intrinsic and niche-mediated changes, which can be the result of transcriptional, posttranscriptional, or posttranslational alterations in MuSCs or in the components of their niche. The interplay between cell intrinsic alterations in MuSCs and changes in the stem cell niche environment during aging and its impact on the number and the function of MuSCs is an important emerging area of research. In this review, we discuss whether the decline in the regenerative potential of MuSCs with age is the cause or the consequence of aging skeletal muscle. Understanding the effect of aging on MuSCs and the individual components of their niche is critical to develop effective therapeutic approaches to diminish or reverse the age-related defects in muscle regeneration.
Collapse
Affiliation(s)
- Korin Sahinyan
- Department of Human Genetics, McGill University, Montréal, QC, Canada.,Lady Davis Institute for Medical Research, Jewish General Hospital, Montréal, QC, Canada
| | - Felicia Lazure
- Department of Human Genetics, McGill University, Montréal, QC, Canada.,Lady Davis Institute for Medical Research, Jewish General Hospital, Montréal, QC, Canada
| | - Darren M Blackburn
- Department of Human Genetics, McGill University, Montréal, QC, Canada.,Lady Davis Institute for Medical Research, Jewish General Hospital, Montréal, QC, Canada
| | - Vahab D Soleimani
- Department of Human Genetics, McGill University, Montréal, QC, Canada.,Lady Davis Institute for Medical Research, Jewish General Hospital, Montréal, QC, Canada
| |
Collapse
|
33
|
Lazure F, Farouni R, Sahinyan K, Blackburn DM, Hernández-Corchado A, Perron G, Lu T, Osakwe A, Ragoussis J, Crist C, Perkins TJ, Jahani-Asl A, Najafabadi HS, Soleimani VD. Transcriptional reprogramming of skeletal muscle stem cells by the niche environment. Nat Commun 2023; 14:535. [PMID: 36726011 PMCID: PMC9892560 DOI: 10.1038/s41467-023-36265-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Accepted: 01/23/2023] [Indexed: 02/03/2023] Open
Abstract
Adult stem cells are indispensable for tissue regeneration, but their function declines with age. The niche environment in which the stem cells reside plays a critical role in their function. However, quantification of the niche effect on stem cell function is lacking. Using muscle stem cells (MuSC) as a model, we show that aging leads to a significant transcriptomic shift in their subpopulations accompanied by locus-specific gain and loss of chromatin accessibility and DNA methylation. By combining in vivo MuSC transplantation and computational methods, we show that the expression of approximately half of all age-altered genes in MuSCs from aged male mice can be restored by exposure to a young niche environment. While there is a correlation between gene reversibility and epigenetic alterations, restoration of gene expression occurs primarily at the level of transcription. The stem cell niche environment therefore represents an important therapeutic target to enhance tissue regeneration in aging.
Collapse
Affiliation(s)
- Felicia Lazure
- Department of Human Genetics, McGill University, 3640 rue University, Montréal, QC, H3A 0C7, Canada.,Lady Davis Institute for Medical Research, Jewish General Hospital, 3755 Chemin de la Côte-Sainte-Catherine, Montréal, QC, H3T 1E2, Canada
| | - Rick Farouni
- Department of Human Genetics, McGill University, 3640 rue University, Montréal, QC, H3A 0C7, Canada.,McGill Genome Centre, Victor Phillip Dahdaleh Institute of Genomic Medicine, 740 Dr Penfield Avenue, Montreal, QC, H3A 0G1, Canada
| | - Korin Sahinyan
- Department of Human Genetics, McGill University, 3640 rue University, Montréal, QC, H3A 0C7, Canada.,Lady Davis Institute for Medical Research, Jewish General Hospital, 3755 Chemin de la Côte-Sainte-Catherine, Montréal, QC, H3T 1E2, Canada
| | - Darren M Blackburn
- Department of Human Genetics, McGill University, 3640 rue University, Montréal, QC, H3A 0C7, Canada.,Lady Davis Institute for Medical Research, Jewish General Hospital, 3755 Chemin de la Côte-Sainte-Catherine, Montréal, QC, H3T 1E2, Canada
| | - Aldo Hernández-Corchado
- Department of Human Genetics, McGill University, 3640 rue University, Montréal, QC, H3A 0C7, Canada.,McGill Genome Centre, Victor Phillip Dahdaleh Institute of Genomic Medicine, 740 Dr Penfield Avenue, Montreal, QC, H3A 0G1, Canada
| | - Gabrielle Perron
- Department of Human Genetics, McGill University, 3640 rue University, Montréal, QC, H3A 0C7, Canada.,McGill Genome Centre, Victor Phillip Dahdaleh Institute of Genomic Medicine, 740 Dr Penfield Avenue, Montreal, QC, H3A 0G1, Canada
| | - Tianyuan Lu
- Lady Davis Institute for Medical Research, Jewish General Hospital, 3755 Chemin de la Côte-Sainte-Catherine, Montréal, QC, H3T 1E2, Canada.,Quantitative Life Sciences, McGill University, Montreal, Canada
| | - Adrien Osakwe
- Quantitative Life Sciences, McGill University, Montreal, Canada
| | - Jiannis Ragoussis
- Department of Human Genetics, McGill University, 3640 rue University, Montréal, QC, H3A 0C7, Canada.,McGill Genome Centre, Victor Phillip Dahdaleh Institute of Genomic Medicine, 740 Dr Penfield Avenue, Montreal, QC, H3A 0G1, Canada
| | - Colin Crist
- Department of Human Genetics, McGill University, 3640 rue University, Montréal, QC, H3A 0C7, Canada.,Lady Davis Institute for Medical Research, Jewish General Hospital, 3755 Chemin de la Côte-Sainte-Catherine, Montréal, QC, H3T 1E2, Canada
| | - Theodore J Perkins
- Sprott Center for Stem Cell Research, Ottawa Hospital Research Institute, 501 Smyth Road, Ottawa, ON, K1H 8L6, Canada.,Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON, K1H 8M5, Canada
| | - Arezu Jahani-Asl
- Department of Cellular and Molecular Medicine and University of Ottawa Brain and Mind Research Institute, University of Ottawa, 451 Smyth Road, Ottawa, ON, K1H 8M5, Canada
| | - Hamed S Najafabadi
- Department of Human Genetics, McGill University, 3640 rue University, Montréal, QC, H3A 0C7, Canada. .,McGill Genome Centre, Victor Phillip Dahdaleh Institute of Genomic Medicine, 740 Dr Penfield Avenue, Montreal, QC, H3A 0G1, Canada. .,Quantitative Life Sciences, McGill University, Montreal, Canada.
| | - Vahab D Soleimani
- Department of Human Genetics, McGill University, 3640 rue University, Montréal, QC, H3A 0C7, Canada. .,Lady Davis Institute for Medical Research, Jewish General Hospital, 3755 Chemin de la Côte-Sainte-Catherine, Montréal, QC, H3T 1E2, Canada.
| |
Collapse
|
34
|
Shintani-Ishida K, Tsurumi R, Ikegaya H. Decrease in the expression of muscle-specific miRNAs, miR-133a and miR-1, in myoblasts with replicative senescence. PLoS One 2023; 18:e0280527. [PMID: 36649291 PMCID: PMC9844915 DOI: 10.1371/journal.pone.0280527] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Accepted: 12/29/2022] [Indexed: 01/18/2023] Open
Abstract
Muscles that are injured or atrophied by aging undergo myogenic regeneration. Although myoblasts play a pivotal role in myogenic regeneration, their function is impaired with aging. MicroRNAs (miRNAs) are also involved in myogenic regeneration. MiRNA (miR)-1 and miR-133a are muscle-specific miRNAs that control the proliferation and differentiation of myoblasts. In this study, we determined whether miR-1 and miR-133a expression in myoblasts is altered with cellular senescence and involved in senescence-impaired myogenic differentiation. C2C12 murine skeletal myoblasts were converted to a replicative senescent state by culturing to a high passage number. Although miR-1 and miR-133a expression was largely induced during myogenic differentiation, expression was suppressed in cells at high passage numbers (passage 10 and/or passage 20). Although the senescent myoblasts exhibited a deterioration of myogenic differentiation, transfection of miR-1 or miR-133a into myoblasts ameliorated cell fusion. Treatment with the glutaminase 1 inhibitor, BPTES, removed senescent cells from C2C12 myoblasts with a high passage number, whereas myotube formation and miR-133a expression was increased. In addition, primary cultured myoblasts prepared from aged C57BL/6J male mice (20 months old) exhibited a decrease in miR-1 and miR-133a levels compared with younger mice (3 months old). The results suggest that replicative senescence suppresses muscle-specific miRNA expression in myoblasts, which contributes to the senescence-related dysfunction of myogenic regeneration.
Collapse
Affiliation(s)
- Kaori Shintani-Ishida
- Department of Forensic Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Riko Tsurumi
- Department of Forensic Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Hiroshi Ikegaya
- Department of Forensic Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| |
Collapse
|
35
|
Stevanovic M, Lazic A, Schwirtlich M, Stanisavljevic Ninkovic D. The Role of SOX Transcription Factors in Ageing and Age-Related Diseases. Int J Mol Sci 2023; 24:851. [PMID: 36614288 PMCID: PMC9821406 DOI: 10.3390/ijms24010851] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 12/27/2022] [Accepted: 12/28/2022] [Indexed: 01/05/2023] Open
Abstract
The quest for eternal youth and immortality is as old as humankind. Ageing is an inevitable physiological process accompanied by many functional declines that are driving factors for age-related diseases. Stem cell exhaustion is one of the major hallmarks of ageing. The SOX transcription factors play well-known roles in self-renewal and differentiation of both embryonic and adult stem cells. As a consequence of ageing, the repertoire of adult stem cells present in various organs steadily declines, and their dysfunction/death could lead to reduced regenerative potential and development of age-related diseases. Thus, restoring the function of aged stem cells, inducing their regenerative potential, and slowing down the ageing process are critical for improving the health span and, consequently, the lifespan of humans. Reprograming factors, including SOX family members, emerge as crucial players in rejuvenation. This review focuses on the roles of SOX transcription factors in stem cell exhaustion and age-related diseases, including neurodegenerative diseases, visual deterioration, chronic obstructive pulmonary disease, osteoporosis, and age-related cancers. A better understanding of the molecular mechanisms of ageing and the roles of SOX transcription factors in this process could open new avenues for developing novel strategies that will delay ageing and prevent age-related diseases.
Collapse
Affiliation(s)
- Milena Stevanovic
- Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Vojvode Stepe 444a, 11042 Belgrade, Serbia
- Faculty of Biology, University of Belgrade, Studentski trg 16, 11158 Belgrade, Serbia
- Serbian Academy of Sciences and Arts, Knez Mihailova 35, 11000 Belgrade, Serbia
| | - Andrijana Lazic
- Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Vojvode Stepe 444a, 11042 Belgrade, Serbia
| | - Marija Schwirtlich
- Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Vojvode Stepe 444a, 11042 Belgrade, Serbia
| | | |
Collapse
|
36
|
Skeletal Muscle Stem Cells in Aging: Asymmetric/Symmetric Division Switching. Symmetry (Basel) 2022. [DOI: 10.3390/sym14122676] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
In aged muscle, satellite cells’ symmetric and asymmetric divisions are impaired, and intrinsic and extrinsic complex mechanisms govern these processes. This review presents many updated aspects regarding muscle stem cells’ fate in normal and aging conditions. The balance between self-renewal and commitment divisions contributes to muscle regeneration, muscle homeostasis, aging, and disease. Stimulating muscle regeneration in aging could be a therapeutic target, but there is still a need to understand the many mechanisms that influence each other in satellite cells and their niche. We highlight here the general outlines regarding satellite cell divisions, the primary markers present in muscle stem cells, the aging aspects concerning signaling pathways involved in symmetric/asymmetric divisions, the regenerative capacity of satellite cells and their niche alteration in senescent muscle, genetics and epigenetics mechanisms implied in satellite cells aging and exercise effect on muscle regeneration in the elderly.
Collapse
|
37
|
Sanchez MM, Bagdasarian IA, Darch W, Morgan JT. Organotypic cultures as aging associated disease models. Aging (Albany NY) 2022; 14:9338-9383. [PMID: 36435511 PMCID: PMC9740367 DOI: 10.18632/aging.204361] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Accepted: 10/21/2022] [Indexed: 11/24/2022]
Abstract
Aging remains a primary risk factor for a host of diseases, including leading causes of death. Aging and associated diseases are inherently multifactorial, with numerous contributing factors and phenotypes at the molecular, cellular, tissue, and organismal scales. Despite the complexity of aging phenomena, models currently used in aging research possess limitations. Frequently used in vivo models often have important physiological differences, age at different rates, or are genetically engineered to match late disease phenotypes rather than early causes. Conversely, routinely used in vitro models lack the complex tissue-scale and systemic cues that are disrupted in aging. To fill in gaps between in vivo and traditional in vitro models, researchers have increasingly been turning to organotypic models, which provide increased physiological relevance with the accessibility and control of in vitro context. While powerful tools, the development of these models is a field of its own, and many aging researchers may be unaware of recent progress in organotypic models, or hesitant to include these models in their own work. In this review, we describe recent progress in tissue engineering applied to organotypic models, highlighting examples explicitly linked to aging and associated disease, as well as examples of models that are relevant to aging. We specifically highlight progress made in skin, gut, and skeletal muscle, and describe how recently demonstrated models have been used for aging studies or similar phenotypes. Throughout, this review emphasizes the accessibility of these models and aims to provide a resource for researchers seeking to leverage these powerful tools.
Collapse
Affiliation(s)
- Martina M. Sanchez
- Department of Bioengineering, University of California, Riverside, CA 92521, USA
| | | | - William Darch
- Department of Bioengineering, University of California, Riverside, CA 92521, USA
| | - Joshua T. Morgan
- Department of Bioengineering, University of California, Riverside, CA 92521, USA
| |
Collapse
|
38
|
Yedigaryan L, Gatti M, Marini V, Maraldi T, Sampaolesi M. Shared and Divergent Epigenetic Mechanisms in Cachexia and Sarcopenia. Cells 2022; 11:2293. [PMID: 35892590 PMCID: PMC9332174 DOI: 10.3390/cells11152293] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Revised: 07/19/2022] [Accepted: 07/20/2022] [Indexed: 01/27/2023] Open
Abstract
Significant loss of muscle mass may occur in cachexia and sarcopenia, which are major causes of mortality and disability. Cachexia represents a complex multi-organ syndrome associated with cancer and chronic diseases. It is often characterized by body weight loss, inflammation, and muscle and adipose wasting. Progressive muscle loss is also a hallmark of healthy aging, which is emerging worldwide as a main demographic trend. A great challenge for the health care systems is the age-related decline in functionality which threatens the independence and quality of life of elderly people. This biological decline can also be associated with functional muscle loss, known as sarcopenia. Previous studies have shown that microRNAs (miRNAs) play pivotal roles in the development and progression of muscle wasting in both cachexia and sarcopenia. These small non-coding RNAs, often carried in extracellular vesicles, inhibit translation by targeting messenger RNAs, therefore representing potent epigenetic modulators. The molecular mechanisms behind cachexia and sarcopenia, including the expression of specific miRNAs, share common and distinctive trends. The aim of the present review is to compile recent evidence about shared and divergent epigenetic mechanisms, particularly focusing on miRNAs, between cachexia and sarcopenia to understand a facet in the underlying muscle wasting associated with these morbidities and disclose potential therapeutic interventions.
Collapse
Affiliation(s)
- Laura Yedigaryan
- Translational Cardiomyology Laboratory, Stem Cell and Developmental Biology, Department of Development and Regeneration, KU Leuven, 3000 Leuven, Belgium; (L.Y.); (V.M.)
| | - Martina Gatti
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy; (M.G.); (T.M.)
| | - Vittoria Marini
- Translational Cardiomyology Laboratory, Stem Cell and Developmental Biology, Department of Development and Regeneration, KU Leuven, 3000 Leuven, Belgium; (L.Y.); (V.M.)
| | - Tullia Maraldi
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy; (M.G.); (T.M.)
| | - Maurilio Sampaolesi
- Translational Cardiomyology Laboratory, Stem Cell and Developmental Biology, Department of Development and Regeneration, KU Leuven, 3000 Leuven, Belgium; (L.Y.); (V.M.)
- Histology and Medical Embryology Unit, Department of Anatomy, Histology, Forensic Medicine and Orthopedics, Sapienza University of Rome, 00185 Rome, Italy
| |
Collapse
|
39
|
Baig MH, Ahmad K, Moon JS, Park SY, Ho Lim J, Chun HJ, Qadri AF, Hwang YC, Jan AT, Ahmad SS, Ali S, Shaikh S, Lee EJ, Choi I. Myostatin and its Regulation: A Comprehensive Review of Myostatin Inhibiting Strategies. Front Physiol 2022; 13:876078. [PMID: 35812316 PMCID: PMC9259834 DOI: 10.3389/fphys.2022.876078] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Accepted: 06/06/2022] [Indexed: 12/12/2022] Open
Abstract
Myostatin (MSTN) is a well-reported negative regulator of muscle growth and a member of the transforming growth factor (TGF) family. MSTN has important functions in skeletal muscle (SM), and its crucial involvement in several disorders has made it an important therapeutic target. Several strategies based on the use of natural compounds to inhibitory peptides are being used to inhibit the activity of MSTN. This review delivers an overview of the current state of knowledge about SM and myogenesis with particular emphasis on the structural characteristics and regulatory functions of MSTN during myogenesis and its involvements in various muscle related disorders. In addition, we review the diverse approaches used to inhibit the activity of MSTN, especially in silico approaches to the screening of natural compounds and the design of novel short peptides derived from proteins that typically interact with MSTN.
Collapse
Affiliation(s)
- Mohammad Hassan Baig
- Department of Family Medicine, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, South Korea
| | - Khurshid Ahmad
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan, South Korea
- Research Institute of Cell Culture, Yeungnam University, Gyeongsan, South Korea
| | - Jun Sung Moon
- Department of Internal Medicine, College of Medicine, Yeungnam University, Daegu, South Korea
| | - So-Young Park
- Department of Physiology, College of Medicine, Yeungnam University, Daegu, South Korea
| | - Jeong Ho Lim
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan, South Korea
- Research Institute of Cell Culture, Yeungnam University, Gyeongsan, South Korea
| | - Hee Jin Chun
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan, South Korea
- Research Institute of Cell Culture, Yeungnam University, Gyeongsan, South Korea
| | - Afsha Fatima Qadri
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan, South Korea
| | - Ye Chan Hwang
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan, South Korea
| | - Arif Tasleem Jan
- School of Biosciences and Biotechnology, Baba Ghulam Shah Badshah University, Rajouri, India
| | - Syed Sayeed Ahmad
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan, South Korea
| | - Shahid Ali
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan, South Korea
| | - Sibhghatulla Shaikh
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan, South Korea
| | - Eun Ju Lee
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan, South Korea
- Research Institute of Cell Culture, Yeungnam University, Gyeongsan, South Korea
- *Correspondence: Eun Ju Lee, ; Inho Choi,
| | - Inho Choi
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan, South Korea
- Research Institute of Cell Culture, Yeungnam University, Gyeongsan, South Korea
- *Correspondence: Eun Ju Lee, ; Inho Choi,
| |
Collapse
|
40
|
Larson AA, Shams AS, McMillin SL, Sullivan BP, Vue C, Roloff ZA, Batchelor E, Kyba M, Lowe DA. Estradiol deficiency reduces the satellite cell pool by impairing cell cycle progression. Am J Physiol Cell Physiol 2022; 322:C1123-C1137. [PMID: 35442828 PMCID: PMC9169829 DOI: 10.1152/ajpcell.00429.2021] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 03/31/2022] [Accepted: 04/17/2022] [Indexed: 12/22/2022]
Abstract
The size of the satellite cell pool is reduced in estradiol (E2)-deficient female mice and humans. Here, we use a combination of in vivo and in vitro approaches to identify mechanisms, whereby E2 deficiency impairs satellite cell maintenance. By measuring satellite cell numbers in mice at several early time points postovariectomy (Ovx), we determine that satellite cell numbers decline by 33% between 10 and 14 days post-Ovx in tibialis anterior and gastrocnemius muscles. At 14 days post-Ovx, we demonstrate that satellite cells have a reduced propensity to transition from G0/G1 to S and G2/M phases, compared with cells from ovary-intact mice, associated with changes in two key satellite cell cycle regulators, ccna2 and p16INK4a. Further, freshly isolated satellite cells treated with E2 in vitro have 62% greater cell proliferation and require less time to complete the first division. Using clonal and differentiation assays, we measured 69% larger satellite cell colonies and enhanced satellite cell-derived myoblast differentiation with E2 treatment compared with vehicle-treated cells. Together, these results identify a novel mechanism for preservation of the satellite cell pool by E2 via promotion of satellite cell cycling.
Collapse
Affiliation(s)
- Alexie A Larson
- Department of Integrative Biology and Physiology, Medical School, University of Minnesota, Minneapolis, Minnesota
| | - Ahmed S Shams
- Lillehei Heart Institute, Medical School, University of Minnesota, Minneapolis, Minnesota
- Department of Pediatrics, Medical School, University of Minnesota, Minneapolis, Minnesota
- Human Anatomy and Embryology Department, Faculty of Medicine, Suez Canal University, Ismailia, Egypt
| | - Shawna L McMillin
- Divisions of Rehabilitation Science and Physical Therapy, Department of Rehabilitation Medicine, Medical School, University of Minnesota, Minneapolis, Minnesota
| | - Brian P Sullivan
- Divisions of Rehabilitation Science and Physical Therapy, Department of Rehabilitation Medicine, Medical School, University of Minnesota, Minneapolis, Minnesota
| | - Cha Vue
- Divisions of Rehabilitation Science and Physical Therapy, Department of Rehabilitation Medicine, Medical School, University of Minnesota, Minneapolis, Minnesota
| | - Zachery A Roloff
- Divisions of Rehabilitation Science and Physical Therapy, Department of Rehabilitation Medicine, Medical School, University of Minnesota, Minneapolis, Minnesota
| | - Eric Batchelor
- Department of Integrative Biology and Physiology, Medical School, University of Minnesota, Minneapolis, Minnesota
| | - Michael Kyba
- Lillehei Heart Institute, Medical School, University of Minnesota, Minneapolis, Minnesota
- Department of Pediatrics, Medical School, University of Minnesota, Minneapolis, Minnesota
| | - Dawn A Lowe
- Divisions of Rehabilitation Science and Physical Therapy, Department of Rehabilitation Medicine, Medical School, University of Minnesota, Minneapolis, Minnesota
| |
Collapse
|
41
|
Roman W, Muñoz-Cánoves P. Muscle is a stage, and cells and factors are merely players. Trends Cell Biol 2022; 32:835-840. [DOI: 10.1016/j.tcb.2022.03.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 02/28/2022] [Accepted: 03/02/2022] [Indexed: 12/25/2022]
|
42
|
Soendenbroe C, Dahl CL, Meulengracht C, Tamáš M, Svensson RB, Schjerling P, Kjaer M, Andersen JL, Mackey AL. Preserved stem cell content and innervation profile of elderly human skeletal muscle with lifelong recreational exercise. J Physiol 2022; 600:1969-1989. [PMID: 35229299 PMCID: PMC9315046 DOI: 10.1113/jp282677] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Accepted: 02/14/2022] [Indexed: 11/21/2022] Open
Abstract
Abstract Muscle fibre denervation and declining numbers of muscle stem (satellite) cells are defining characteristics of ageing skeletal muscle. The aim of this study was to investigate the potential for lifelong recreational exercise to offset muscle fibre denervation and compromised satellite cell content and function, both at rest and under challenged conditions. Sixteen elderly lifelong recreational exercisers (LLEX) were studied alongside groups of age‐matched sedentary (SED) and young subjects. Lean body mass and maximal voluntary contraction were assessed, and a strength training bout was performed. From muscle biopsies, tissue and primary myogenic cell cultures were analysed by immunofluorescence and RT‐qPCR to assess myofibre denervation and satellite cell quantity and function. LLEX demonstrated superior muscle function under challenged conditions. When compared with SED, the muscle of LLEX was found to contain a greater content of satellite cells associated with type II myofibres specifically, along with higher mRNA levels of the beta and gamma acetylcholine receptors (AChR). No difference was observed between LLEX and SED for the proportion of denervated fibres or satellite cell function, as assessed in vitro by myogenic cell differentiation and fusion index assays. When compared with inactive counterparts, the skeletal muscle of lifelong exercisers is characterised by greater fatigue resistance under challenged conditions in vivo, together with a more youthful tissue satellite cell and AChR profile. Our data suggest a little recreational level exercise goes a long way in protecting against the emergence of classic phenotypic traits associated with the aged muscle. Key points The detrimental effects of ageing can be partially offset by lifelong self‐organized recreational exercise, as evidence by preserved type II myofibre‐associated satellite cells, a beneficial muscle innervation status and greater fatigue resistance under challenged conditions. Satellite cell function (in vitro), muscle fibre size and muscle fibre denervation determined by immunofluorescence were not affected by recreational exercise. Individuals that are recreationally active are far more abundant than master athletes, which sharply increases the translational perspective of the present study. Future studies should further investigate recreational activity in relation to muscle health, while also including female participants.
Collapse
Affiliation(s)
- Casper Soendenbroe
- Institute of Sports Medicine Copenhagen, Department of Orthopedic Surgery M, Copenhagen University Hospital - Bispebjerg and Frederiksberg, Building 8, Nielsine Nielsens vej 11, Copenhagen, NV, 2400, Denmark.,Xlab, Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3, Copenhagen N, 2200, Denmark.,Center for Healthy Aging, Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3, Copenhagen N, 2200, Denmark
| | - Christopher L Dahl
- Institute of Sports Medicine Copenhagen, Department of Orthopedic Surgery M, Copenhagen University Hospital - Bispebjerg and Frederiksberg, Building 8, Nielsine Nielsens vej 11, Copenhagen, NV, 2400, Denmark
| | - Christopher Meulengracht
- Institute of Sports Medicine Copenhagen, Department of Orthopedic Surgery M, Copenhagen University Hospital - Bispebjerg and Frederiksberg, Building 8, Nielsine Nielsens vej 11, Copenhagen, NV, 2400, Denmark
| | - Michal Tamáš
- Institute of Sports Medicine Copenhagen, Department of Orthopedic Surgery M, Copenhagen University Hospital - Bispebjerg and Frederiksberg, Building 8, Nielsine Nielsens vej 11, Copenhagen, NV, 2400, Denmark
| | - Rene B Svensson
- Institute of Sports Medicine Copenhagen, Department of Orthopedic Surgery M, Copenhagen University Hospital - Bispebjerg and Frederiksberg, Building 8, Nielsine Nielsens vej 11, Copenhagen, NV, 2400, Denmark.,Center for Healthy Aging, Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3, Copenhagen N, 2200, Denmark
| | - Peter Schjerling
- Institute of Sports Medicine Copenhagen, Department of Orthopedic Surgery M, Copenhagen University Hospital - Bispebjerg and Frederiksberg, Building 8, Nielsine Nielsens vej 11, Copenhagen, NV, 2400, Denmark.,Center for Healthy Aging, Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3, Copenhagen N, 2200, Denmark
| | - Michael Kjaer
- Institute of Sports Medicine Copenhagen, Department of Orthopedic Surgery M, Copenhagen University Hospital - Bispebjerg and Frederiksberg, Building 8, Nielsine Nielsens vej 11, Copenhagen, NV, 2400, Denmark.,Center for Healthy Aging, Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3, Copenhagen N, 2200, Denmark
| | - Jesper L Andersen
- Institute of Sports Medicine Copenhagen, Department of Orthopedic Surgery M, Copenhagen University Hospital - Bispebjerg and Frederiksberg, Building 8, Nielsine Nielsens vej 11, Copenhagen, NV, 2400, Denmark.,Center for Healthy Aging, Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3, Copenhagen N, 2200, Denmark
| | - Abigail L Mackey
- Institute of Sports Medicine Copenhagen, Department of Orthopedic Surgery M, Copenhagen University Hospital - Bispebjerg and Frederiksberg, Building 8, Nielsine Nielsens vej 11, Copenhagen, NV, 2400, Denmark.,Xlab, Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3, Copenhagen N, 2200, Denmark.,Center for Healthy Aging, Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3, Copenhagen N, 2200, Denmark
| |
Collapse
|
43
|
Staunton CA, Owen ED, Hemmings K, Vasilaki A, McArdle A, Barrett-Jolley R, Jackson MJ. Skeletal muscle transcriptomics identifies common pathways in nerve crush injury and ageing. Skelet Muscle 2022; 12:3. [PMID: 35093178 PMCID: PMC8800362 DOI: 10.1186/s13395-021-00283-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Accepted: 11/24/2021] [Indexed: 12/16/2022] Open
Abstract
Motor unit remodelling involving repeated denervation and re-innervation occurs throughout life. The efficiency of this process declines with age contributing to neuromuscular deficits. This study investigated differentially expressed genes (DEG) in muscle following peroneal nerve crush to model motor unit remodelling in C57BL/6 J mice. Muscle RNA was isolated at 3 days post-crush, RNA libraries were generated using poly-A selection, sequenced and analysed using gene ontology and pathway tools. Three hundred thirty-four DEG were found in quiescent muscle from (26mnth) old compared with (4-6mnth) adult mice and these same DEG were present in muscle from adult mice following nerve crush. Peroneal crush induced 7133 DEG in muscles of adult and 699 DEG in muscles from old mice, although only one DEG (ZCCHC17) was found when directly comparing nerve-crushed muscles from old and adult mice. This analysis revealed key differences in muscle responses which may underlie the diminished ability of old mice to repair following nerve injury.
Collapse
Affiliation(s)
- C A Staunton
- MRC- Versus Arthritis Research Centre for Integrated research into Musculoskeletal Ageing (CIMA), Department of Musculoskeletal and Ageing Science, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, L7 8TX, UK
| | - E D Owen
- MRC- Versus Arthritis Research Centre for Integrated research into Musculoskeletal Ageing (CIMA), Department of Musculoskeletal and Ageing Science, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, L7 8TX, UK
| | - K Hemmings
- MRC- Versus Arthritis Research Centre for Integrated research into Musculoskeletal Ageing (CIMA), Department of Musculoskeletal and Ageing Science, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, L7 8TX, UK
| | - A Vasilaki
- MRC- Versus Arthritis Research Centre for Integrated research into Musculoskeletal Ageing (CIMA), Department of Musculoskeletal and Ageing Science, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, L7 8TX, UK
| | - A McArdle
- MRC- Versus Arthritis Research Centre for Integrated research into Musculoskeletal Ageing (CIMA), Department of Musculoskeletal and Ageing Science, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, L7 8TX, UK
| | - R Barrett-Jolley
- MRC- Versus Arthritis Research Centre for Integrated research into Musculoskeletal Ageing (CIMA), Department of Musculoskeletal and Ageing Science, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, L7 8TX, UK
| | - M J Jackson
- MRC- Versus Arthritis Research Centre for Integrated research into Musculoskeletal Ageing (CIMA), Department of Musculoskeletal and Ageing Science, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, L7 8TX, UK.
| |
Collapse
|
44
|
Mori T, Onodera Y, Itokazu M, Takehara T, Shigi K, Iwawaki N, Akagi M, Teramura T. Depletion of NIMA-related kinase Nek2 induces aberrant self-renewal and apoptosis in stem/progenitor cells of aged muscular tissues. Mech Ageing Dev 2022; 201:111619. [PMID: 34995645 DOI: 10.1016/j.mad.2022.111619] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 12/23/2021] [Accepted: 01/03/2022] [Indexed: 11/25/2022]
Abstract
Frailty of the locomotory organs has become a widespread problem in the geriatric population. The major factor leading to frailty is an age-associated decrease in muscular mass and a reduced number of muscular cells and myofibers. In aged muscular tissues, muscular satellite cells (MuSCs) are reduced due to abnormalities in their self-renewal and the induction of apoptosis. However, the molecular mechanisms connecting aging-associated physiological changes and the reduction of MuSCs are largely unknown. NIMA-related kinase 2 (Nek2), a member of the Nek family of serine/threonine kinases, was found to be downregulated in aged MuSCs/progenitors. Further, Nek2 downregulation was found to inhibit self-renewal and apoptotic cell death by activating the p53-dependent checkpoint. Attenuated NEK2 expression was also observed in the muscular tissues of elderly donors, and its function was confirmed to be conserved in humans. Overall, this study proposes a novel mechanism for inducing muscular atrophy to understand aging-associated muscular diseases.
Collapse
Affiliation(s)
| | - Yuta Onodera
- Institute of Advanced Clinical Medicine, Kindai University Hospital, Japan
| | - Maki Itokazu
- Department of Rehabilitation Medicine, Kindai University Faculty of Medicine, Japan
| | - Toshiyuki Takehara
- Institute of Advanced Clinical Medicine, Kindai University Hospital, Japan
| | - Kanae Shigi
- Institute of Advanced Clinical Medicine, Kindai University Hospital, Japan
| | - Natsumi Iwawaki
- Institute of Advanced Clinical Medicine, Kindai University Hospital, Japan
| | - Masao Akagi
- Department of Orthopedic Surgery, Kindai University Faculty of Medicine, Japan
| | - Takeshi Teramura
- Institute of Advanced Clinical Medicine, Kindai University Hospital, Japan.
| |
Collapse
|
45
|
Arroyo E, Troutman AD, Moorthi RN, Avin KG, Coggan AR, Lim K. Klotho: An Emerging Factor With Ergogenic Potential. FRONTIERS IN REHABILITATION SCIENCES 2022; 2:807123. [PMID: 36188832 PMCID: PMC9397700 DOI: 10.3389/fresc.2021.807123] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Accepted: 12/10/2021] [Indexed: 11/13/2022]
Abstract
Sarcopenia and impaired cardiorespiratory fitness are commonly observed in older individuals and patients with chronic kidney disease (CKD). Declines in skeletal muscle function and aerobic capacity can progress into impaired physical function and inability to perform activities of daily living. Physical function is highly associated with important clinical outcomes such as hospitalization, functional independence, quality of life, and mortality. While lifestyle modifications such as exercise and dietary interventions have been shown to prevent and reverse declines in physical function, the utility of these treatment strategies is limited by poor widespread adoption and adherence due to a wide variety of both perceived and actual barriers to exercise. Therefore, identifying novel treatment targets to manage physical function decline is critically important. Klotho, a remarkable protein with powerful anti-aging properties has recently been investigated for its role in musculoskeletal health and physical function. Klotho is involved in several key processes that regulate skeletal muscle function, such as muscle regeneration, mitochondrial biogenesis, endothelial function, oxidative stress, and inflammation. This is particularly important for older adults and patients with CKD, which are known states of Klotho deficiency. Emerging data support the existence of Klotho-related benefits to exercise and for potential Klotho-based therapeutic interventions for the treatment of sarcopenia and its progression to physical disability. However, significant gaps in our understanding of Klotho must first be overcome before we can consider its potential ergogenic benefits. These advances will be critical to establish the optimal approach to future Klotho-based interventional trials and to determine if Klotho can regulate physical dysfunction.
Collapse
Affiliation(s)
- Eliott Arroyo
- Division of Nephrology & Hypertension, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Ashley D. Troutman
- Department of Physical Therapy, School of Health and Human Sciences, Indiana University Purdue University, Indianapolis, IN, United States
| | - Ranjani N. Moorthi
- Division of Nephrology & Hypertension, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Keith G. Avin
- Division of Nephrology & Hypertension, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, United States
- Department of Physical Therapy, School of Health and Human Sciences, Indiana University Purdue University, Indianapolis, IN, United States
| | - Andrew R. Coggan
- Department of Kinesiology, School of Health and Human Sciences, Indiana University Purdue University Indianapolis, Indianapolis, IN, United States
| | - Kenneth Lim
- Division of Nephrology & Hypertension, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, United States
| |
Collapse
|
46
|
Moreira-Pais A, Ferreira R, Oliveira PA, Duarte JA. A neuromuscular perspective of sarcopenia pathogenesis: deciphering the signaling pathways involved. GeroScience 2022; 44:1199-1213. [PMID: 34981273 DOI: 10.1007/s11357-021-00510-2] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 12/26/2021] [Indexed: 12/18/2022] Open
Abstract
The escalation of life expectancy is accompanied by an increase in the prevalence of age-related conditions, such as sarcopenia. Sarcopenia, a muscle condition defined by low muscle strength, muscle quality or quantity, and physical performance, has a high prevalence among the elderly and is associated to increased mortality. The neuromuscular system has been emerging as a key contributor to sarcopenia pathogenesis. Indeed, the age-related degeneration of the neuromuscular junction (NMJ) function and structure may contribute to the loss of muscle strength and ultimately to the loss of muscle mass that characterize sarcopenia. The present mini-review discusses important signaling pathways involved in the function and maintenance of the NMJ, giving emphasis to the ones that might contribute to sarcopenia pathogenesis. Some conceivable biomarkers, such as C-terminal agrin fragment (CAF) and brain-derived neurotrophic factor (BDNF), and therapeutic targets, namely acetylcholine and calcitonin gene-related peptide (CGRP), can be retrieved, making way to future studies to validate their clinical use.
Collapse
Affiliation(s)
- Alexandra Moreira-Pais
- CIAFEL, Faculty of Sport, University of Porto, Dr. Plácido da Costa 91, 4200-450, Porto, Portugal. .,LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, 3810-193, Aveiro, Portugal. .,Centre for Research and Technology of Agro Environmental and Biological Sciences (CITAB), Inov4Agro, University of Trás-Os-Montes and Alto Douro (UTAD), Quinta de Prados, 5000-801, Vila Real, Portugal.
| | - Rita Ferreira
- LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, 3810-193, Aveiro, Portugal
| | - Paula A Oliveira
- Centre for Research and Technology of Agro Environmental and Biological Sciences (CITAB), Inov4Agro, University of Trás-Os-Montes and Alto Douro (UTAD), Quinta de Prados, 5000-801, Vila Real, Portugal
| | - José A Duarte
- CIAFEL, Faculty of Sport, University of Porto, Dr. Plácido da Costa 91, 4200-450, Porto, Portugal.,TOXRUN - Toxicology Research Unit, University Institute of Health Sciences, CESPU, CRL, Gandra, Portugal
| |
Collapse
|
47
|
Dungan CM, Murach KA, Zdunek CJ, Tang ZJ, Nolt GL, Brightwell CR, Hettinger Z, Englund D, Liu Z, Fry CS, Filareto A, Franti M, Peterson CA. Deletion of SA β-Gal+ cells using senolytics improves muscle regeneration in old mice. Aging Cell 2022; 21:e13528. [PMID: 34904366 PMCID: PMC8761017 DOI: 10.1111/acel.13528] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 10/05/2021] [Accepted: 11/21/2021] [Indexed: 12/11/2022] Open
Abstract
Systemic deletion of senescent cells leads to robust improvements in cognitive, cardiovascular, and whole-body metabolism, but their role in tissue reparative processes is incompletely understood. We hypothesized that senolytic drugs would enhance regeneration in aged skeletal muscle. Young (3 months) and old (20 months) male C57Bl/6J mice were administered the senolytics dasatinib (5 mg/kg) and quercetin (50 mg/kg) or vehicle bi-weekly for 4 months. Tibialis anterior (TA) was then injected with 1.2% BaCl2 or PBS 7- or 28 days prior to euthanization. Senescence-associated β-Galactosidase positive (SA β-Gal+) cell abundance was low in muscle from both young and old mice and increased similarly 7 days following injury in both age groups, with no effect of D+Q. Most SA β-Gal+ cells were also CD11b+ in young and old mice 7- and 14 days following injury, suggesting they are infiltrating immune cells. By 14 days, SA β-Gal+/CD11b+ cells from old mice expressed senescence genes, whereas those from young mice expressed higher levels of genes characteristic of anti-inflammatory macrophages. SA β-Gal+ cells remained elevated in old compared to young mice 28 days following injury, which were reduced by D+Q only in the old mice. In D+Q-treated old mice, muscle regenerated following injury to a greater extent compared to vehicle-treated old mice, having larger fiber cross-sectional area after 28 days. Conversely, D+Q blunted regeneration in young mice. In vitro experiments suggested D+Q directly improve myogenic progenitor cell proliferation. Enhanced physical function and improved muscle regeneration demonstrate that senolytics have beneficial effects only in old mice.
Collapse
Affiliation(s)
- Cory M. Dungan
- Department of Physical TherapyCollege of Health SciencesUniversity of KentuckyLexingtonKentuckyUSA
- Sanders‐Brown Center on AgingUniversity of KentuckyLexingtonKentuckyUSA
- The Center for Muscle BiologyUniversity of KentuckyLexingtonKentuckyUSA
| | - Kevin A. Murach
- Department of Physical TherapyCollege of Health SciencesUniversity of KentuckyLexingtonKentuckyUSA
- The Center for Muscle BiologyUniversity of KentuckyLexingtonKentuckyUSA
- Present address:
Department of Health, Human Performance, and Recreation, and Cell and Molecular Biology ProgramUniversity of ArkansasFayettevilleArkansasUSA
| | | | - Zuo Jian Tang
- Computational BiologyGCBDSBoehringer Ingelheim Pharmaceuticals Inc.RidgefieldConnecticutUSA
| | - Georgia L. Nolt
- The Center for Muscle BiologyUniversity of KentuckyLexingtonKentuckyUSA
| | - Camille R. Brightwell
- The Center for Muscle BiologyUniversity of KentuckyLexingtonKentuckyUSA
- Department of Athletic Training and Clinical NutritionCollege of Health SciencesUniversity of KentuckyLexingtonKentuckyUSA
| | - Zachary Hettinger
- Department of Physical TherapyCollege of Health SciencesUniversity of KentuckyLexingtonKentuckyUSA
- The Center for Muscle BiologyUniversity of KentuckyLexingtonKentuckyUSA
| | - Davis A. Englund
- Department of Physical TherapyCollege of Health SciencesUniversity of KentuckyLexingtonKentuckyUSA
- The Center for Muscle BiologyUniversity of KentuckyLexingtonKentuckyUSA
| | - Zheng Liu
- Computational BiologyGCBDSBoehringer Ingelheim Pharmaceuticals Inc.RidgefieldConnecticutUSA
| | - Christopher S. Fry
- The Center for Muscle BiologyUniversity of KentuckyLexingtonKentuckyUSA
- Department of Athletic Training and Clinical NutritionCollege of Health SciencesUniversity of KentuckyLexingtonKentuckyUSA
| | - Antonio Filareto
- Regenerative MedicineBoehringer Ingelheim Pharmaceuticals Inc.RidgefieldConnecticutUSA
| | - Michael Franti
- Regenerative MedicineBoehringer Ingelheim Pharmaceuticals Inc.RidgefieldConnecticutUSA
| | - Charlotte A. Peterson
- Department of Physical TherapyCollege of Health SciencesUniversity of KentuckyLexingtonKentuckyUSA
- The Center for Muscle BiologyUniversity of KentuckyLexingtonKentuckyUSA
| |
Collapse
|
48
|
Jacques E, Kuang Y, Kann AP, Le Grand F, Krauss RS, Gilbert PM. The mini-IDLE 3D biomimetic culture assay enables interrogation of mechanisms governing muscle stem cell quiescence and niche repopulation. eLife 2022; 11:81738. [PMID: 36537758 PMCID: PMC9904761 DOI: 10.7554/elife.81738] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2022] [Accepted: 12/19/2022] [Indexed: 12/24/2022] Open
Abstract
Adult skeletal muscle harbours a population of muscle stem cells (MuSCs) that are required for repair after tissue injury. In youth, MuSCs return to a reversible state of cell-cycle arrest termed 'quiescence' after injury resolution. Conversely, some MuSCs in aged muscle remain semi-activated, causing a premature response to injuries that results in incomplete repair and eventual stem cell depletion. Regulating this balance between MuSC quiescence and activation may hold the key to restoring tissue homeostasis with age, but is incompletely understood. To fill this gap, we developed a simple and tractable in vitro method, to rapidly inactivate MuSCs freshly isolated from young murine skeletal muscle, and return them to a quiescent-like state for at least 1-week, which we name mini-IDLE (Inactivation and Dormancy LEveraged in vitro). This was achieved by introducing MuSCs into a 3D bioartificial niche comprised of a thin sheet of mouse myotubes, which we demonstrate provides the minimal cues necessary to induce quiescence. With different starting numbers of MuSCs, the assay revealed cellular heterogeneity and population-level adaptations that converged on a common niche repopulation density; behaviours previously observed only in vivo. Quiescence-associated hallmarks included a Pax7+CalcR+DDX6+MyoD-c-FOS- signature, quiescent-like morphologies, and polarized niche markers. Leveraging high-content bioimaging pipelines, we demonstrate a relationship between morphology and cell fate signatures for possible real-time morphology-based screening. When using MuSCs from aged muscle, they displayed aberrant proliferative activities and delayed inactivation kinetics, among other quiescence-associated defects that we show are partially rescued by wortmannin treatment. Thus, the assay offers an unprecedented opportunity to systematically investigate long-standing queries in areas such as regulation of pool size and functional heterogeneity within the MuSC population, and to uncover quiescence regulators in youth and age.
Collapse
Affiliation(s)
- Erik Jacques
- Institute of Biomedical Engineering, University of TorontoTorontoCanada,Donnelly Centre, University of TorontoTorontoCanada
| | - Yinni Kuang
- Donnelly Centre, University of TorontoTorontoCanada,Department of Cell and Systems Biology, University of TorontoTorontoCanada
| | - Allison P Kann
- Department of Cell, Developmental, and Regenerative Biology, Icahn School of Medicine at Mount SinaiNew YorkUnited States,Black Family Stem Cell Institute, Icahn School of Medicine at Mount SinaiNew YorkUnited States,Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount SinaiNew YorkUnited States
| | - Fabien Le Grand
- Université Claude Bernard Lyon 1, CNRS UMR 5261, INSERM U1315, Institut NeuroMyoGène - Pathophysiology and Genetics of Neuron and MuscleLyonFrance
| | - Robert S Krauss
- Department of Cell, Developmental, and Regenerative Biology, Icahn School of Medicine at Mount SinaiNew YorkUnited States,Black Family Stem Cell Institute, Icahn School of Medicine at Mount SinaiNew YorkUnited States,Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount SinaiNew YorkUnited States
| | - Penney M Gilbert
- Institute of Biomedical Engineering, University of TorontoTorontoCanada,Donnelly Centre, University of TorontoTorontoCanada,Department of Cell and Systems Biology, University of TorontoTorontoCanada
| |
Collapse
|
49
|
Zofkie W, Southard SM, Braun T, Lepper C. Fibroblast growth factor 6 regulates sizing of the muscle stem cell pool. Stem Cell Reports 2021; 16:2913-2927. [PMID: 34739848 PMCID: PMC8693628 DOI: 10.1016/j.stemcr.2021.10.006] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 10/06/2021] [Accepted: 10/07/2021] [Indexed: 11/25/2022] Open
Abstract
Skeletal muscle stem cells, i.e., satellite cells (SCs), are the essential source of new myonuclei for skeletal muscle regeneration following injury or chronic degenerative myopathies. Both SC number and regenerative capacity diminish during aging. However, molecular regulators that govern sizing of the initial SC pool are unknown. We demonstrate that fibroblast growth factor 6 (FGF6) is critical for SC pool scaling. Mice lacking FGF6 have reduced SCs of early postnatal origin and impaired regeneration. By contrast, increasing FGF6 during the early postnatal period is sufficient for SC expansion. Together, these data support that FGF6 is necessary and sufficient to modulate SC numbers during a critical postnatal period to establish the quiescent adult muscle stem cell pool. Our work highlights postnatal development as a time window receptive for scaling a somatic stem cell population via growth factor signaling, which might be relevant for designing new biomedical strategies to enhance tissue regeneration.
Collapse
Affiliation(s)
- William Zofkie
- Department of Physiology and Cell Biology, The Ohio State University, Columbus, OH, USA
| | | | - Thomas Braun
- Department of Cardiac Development and Remodeling, Max-Planck-Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Christoph Lepper
- Department of Physiology and Cell Biology, The Ohio State University, Columbus, OH, USA.
| |
Collapse
|
50
|
Todaka H, Arikawa M, Noguchi T, Ichikawa A, Sato T. Donepezil, an anti-Alzheimer's disease drug, promotes differentiation and regeneration in injured skeletal muscle through the elevation of the expression of myogenic regulatory factors. Eur J Pharmacol 2021; 911:174528. [PMID: 34582845 DOI: 10.1016/j.ejphar.2021.174528] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Revised: 09/18/2021] [Accepted: 09/23/2021] [Indexed: 10/20/2022]
Abstract
We previously demonstrated that donepezil, an anti-Alzheimer's disease drug, improved skeletal muscle atrophy by enhancing the angiogenesis of endothelial cells and activating the proliferation of satellite cells in a mouse model of peripheral arterial disease. However, the effect of donepezil on muscle differentiation during regeneration remains unclear. Therefore, we measured the expressions of myogenic regulatory factors and late muscle differentiation markers in donepezil-treated C2C12 myoblast cells before and after the induction of cell differentiation. The results indicate that the expressions of myogenin, troponin T (TnT) and myosin heavy chain (MyHC) were significantly increased and myotube formation was accelerated in donepezil-treated cells under the differentiation condition. However, the promotive effect of donepezil on muscle differentiation could not be reproduced by the addition of acetylcholine (ACh) and was not disrupted after treatment with ACh receptor blockers. Moreover, other kinds of acetylcholinesterase inhibitors failed to promote muscle differentiation in C2C12 cells. These results indicate that the specific characteristics of donepezil in the promotion of muscle differentiation are independent of its acetylcholinesterase-inhibitory action. We further found that donepezil induced an incremental shift of the cross-sectional area of myofibers and elevated the expressions of myogenin, TnT and MyHC in a mouse model of cardiotoxin injury. These results suggest that donepezil promotes the differentiation of muscle regeneration upon injury via the elevation of the expressions of myogenic regulatory factors and late muscle differentiation markers. Our findings suggest that donepezil can be a useful therapeutic agent for injured skeletal muscle treatment.
Collapse
Affiliation(s)
- Hiroshi Todaka
- Department of Cardiovascular Control, Kochi Medical School, Nankoku, Kochi, Japan.
| | - Mikihiko Arikawa
- Department of Biological Sciences, Faculty of Science and Technology, Kochi University, Akebono, Kochi, Japan
| | - Tatsuya Noguchi
- Department of Cardiology and Geriatrics, Kochi Medical School, Nankoku, Kochi, Japan
| | - Atsushi Ichikawa
- Department of Cardiovascular Control, Kochi Medical School, Nankoku, Kochi, Japan
| | - Takayuki Sato
- Department of Cardiovascular Control, Kochi Medical School, Nankoku, Kochi, Japan
| |
Collapse
|