1
|
Li L, Guan X, Huang Y, Qu B, Yao B, Ding H. Identification of key genes and signaling pathways based on transcriptomic studies of aerobic and resistance training interventions in sarcopenia in SAMP8 mice. SPORTS MEDICINE AND HEALTH SCIENCE 2024; 6:358-369. [PMID: 39309455 PMCID: PMC11411317 DOI: 10.1016/j.smhs.2024.01.005] [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: 11/30/2022] [Revised: 11/15/2023] [Accepted: 01/19/2024] [Indexed: 09/25/2024] Open
Abstract
We examined the effects of resistance and aerobic exercise on the gene expression and biometabolic processes of aging skeletal muscle in senescence-accelerated mouse/prone 8 mice, a model of sarcopenia, and compared them with senescence-accelerated mouse/resistant 1 mice acting as controls. We found that exercise improved muscle strength, endurance, fiber size, also modulated genes and pathways related to synaptic transmission, potassium transport, JAK-STAT signaling, and PI3K-Akt signaling. Our results suggested that BDNF, JAK2, RhoC, Myh6, Stat5a, Tnnc1, and other genes may mediate the beneficial effects of exercise on sarcopenia through these pathways.
Collapse
Affiliation(s)
- Lunyu Li
- School of Sports Medicine and Health, Chengdu Sport University, Chengdu, China
| | - Xiaotian Guan
- School of Sports Medicine and Health, Chengdu Sport University, Chengdu, China
| | - Ying Huang
- School of Sports Medicine and Health, Chengdu Sport University, Chengdu, China
| | - Bo Qu
- School of Sports Medicine and Health, Chengdu Sport University, Chengdu, China
| | - Binyu Yao
- School of Sports Medicine and Health, Chengdu Sport University, Chengdu, China
| | - Haili Ding
- School of Sports Medicine and Health, Chengdu Sport University, Chengdu, China
- Institute of Sports Medicine and Health, Chengdu Sport University, Chengdu, China
| |
Collapse
|
2
|
Rahman FA, Hian-Cheong DJ, Boonstra K, Ma A, Thoms JP, Zago AS, Quadrilatero J. Augmented mitochondrial apoptotic signaling impairs C2C12 myoblast differentiation following cellular aging through sequential passaging. J Cell Physiol 2024; 239:e31155. [PMID: 38212955 DOI: 10.1002/jcp.31155] [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: 07/12/2023] [Revised: 10/28/2023] [Accepted: 11/02/2023] [Indexed: 01/13/2024]
Abstract
Aging is associated with the steady decline of several cellular processes. The loss of skeletal muscle mass, termed sarcopenia, is one of the major hallmarks of aging. Aged skeletal muscle exhibits a robust reduction in its regenerative capacity due to dysfunction (i.e., senescence, lack of self-renewal, and impaired differentiation) of resident muscle stem cells, called satellite cells. To replicate aging in vitro, immortalized skeletal muscle cells (myoblasts) can be treated with various agents to mimic age-related dysfunction; however, these come with their own set of limitations. In the present study, we used sequential passaging of mouse myoblasts to mimic impaired differentiation that is observed in aged skeletal muscle. Further, we investigated mitochondrial apoptotic mechanisms to better understand the impaired differentiation in these "aged" cells. Our data shows that sequential passaging (>20 passages) of myoblasts is accompanied with significant reductions in differentiation and elevated cell death. Furthermore, high-passage (HP) myoblasts exhibit greater mitochondrial-mediated apoptotic signaling through mitochondrial BAX translocation, CYCS and AIFM1 release, and caspase-9 activation. Finally, we show that inhibition of mitochondrial outer membrane permeability partly recovered differentiation in HP myoblasts. Together, our findings suggests that mitochondrial apoptotic signaling is a contributing factor to the diminished differentiation that is observed in aged myoblasts.
Collapse
Affiliation(s)
- Fasih A Rahman
- Department of Kinesiology and Health Sciences, University of Waterloo, Waterloo, ON, Canada
| | - Dylan J Hian-Cheong
- Department of Kinesiology and Health Sciences, University of Waterloo, Waterloo, ON, Canada
| | - Kristen Boonstra
- Department of Kinesiology and Health Sciences, University of Waterloo, Waterloo, ON, Canada
| | - Andrew Ma
- Department of Kinesiology and Health Sciences, University of Waterloo, Waterloo, ON, Canada
| | - James P Thoms
- Department of Kinesiology and Health Sciences, University of Waterloo, Waterloo, ON, Canada
| | - Anderson S Zago
- Department of Physical Education, School of Sciences, Sao Paulo State University, Bauru, Brazil
| | - Joe Quadrilatero
- Department of Kinesiology and Health Sciences, University of Waterloo, Waterloo, ON, Canada
| |
Collapse
|
3
|
Hung YL, Sato A, Takino Y, Ishigami A, Machida S. Resistance training suppresses accumulation of senescent fibro-adipogenic progenitors and senescence-associated secretory phenotype in aging rat skeletal muscle. GeroScience 2024:10.1007/s11357-024-01338-2. [PMID: 39298108 DOI: 10.1007/s11357-024-01338-2] [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: 04/13/2024] [Accepted: 09/02/2024] [Indexed: 09/21/2024] Open
Abstract
Accumulation of senescent cells in tissues contributes to multiple aging-related pathologies. Senescent fibro-adipogenic progenitors (FAPs) contribute to aging-related muscle atrophy. Resistance training can help to maintain skeletal muscle mass, improve mobility, and reduce certain health risks commonly associated with aging. We investigated, using rat model, the impact of resistance training on FAPs in aging skeletal muscle, which remains unclear. Twenty-two-month-old female rats were divided into sedentary and training groups. The training group rodents were trained to climb a ladder while bearing a load for 20 training sessions over 2 months, after which, the flexor hallucis longus muscles were collected and analyzed. Senescent cells were identified using a senescence-associated β-galactosidase stain and p21 immunohistochemistry (IHC), and FAPs were identified using platelet-derived growth factor receptor alpha IHC. The results indicate that resistance training in rats prevented aging-associated skeletal muscle atrophy and suppressed M2 polarization of macrophages. The number of senescent cells was significantly reduced in the 24-month-old training group, with most of them being FAPs. Conversely, the number of senescent FAPs increased significantly in the 24-month-old sedentary group compared with that in the 18-month-old sedentary group. The number of senescent FAPs in the 24-month-old training group decreased significantly. Resistance training also suppressed the senescence-associated secretory phenotype (SASP). The killer T cell-specific marker, CD8α, was elevated in the skeletal muscles of the aging rats following resistance training, indicating upregulation of recognition and elimination of senescent cells. Overall, resistance training suppressed the accumulation of senescent FAPs and acquisition of SASP in aging skeletal muscles.
Collapse
Affiliation(s)
- Yung-Li Hung
- Japan Society for the Promotion of Science, 5-3-1 Kojimachi, Chiyoda-Ku, Tokyo, 102-0083, Japan
- Graduate School of Health and Sports Science, Juntendo University, 1-1 Hirakagakuendai, Inzai, Chiba, 270-1695, Japan
| | - Ayami Sato
- Molecular Regulation of Aging, Tokyo Metropolitan Institute for Geriatrics and Gerontology, 35-2 Sakae-Cho, Itabashi-Ku, Tokyo, 173-0015, Japan
| | - Yuka Takino
- Molecular Regulation of Aging, Tokyo Metropolitan Institute for Geriatrics and Gerontology, 35-2 Sakae-Cho, Itabashi-Ku, Tokyo, 173-0015, Japan
| | - Akihito Ishigami
- Molecular Regulation of Aging, Tokyo Metropolitan Institute for Geriatrics and Gerontology, 35-2 Sakae-Cho, Itabashi-Ku, Tokyo, 173-0015, Japan
| | - Shuichi Machida
- Graduate School of Health and Sports Science, Juntendo University, 1-1 Hirakagakuendai, Inzai, Chiba, 270-1695, Japan.
- Institute of Health and Sports Science & Medicine, Juntendo University, 1-1 Hirakagakuendai, Inzai, Chiba, 270-1695, Japan.
| |
Collapse
|
4
|
Falvino A, Gasperini B, Cariati I, Bonanni R, Chiavoghilefu A, Gasbarra E, Botta A, Tancredi V, Tarantino U. Cellular Senescence: The Driving Force of Musculoskeletal Diseases. Biomedicines 2024; 12:1948. [PMID: 39335461 PMCID: PMC11429507 DOI: 10.3390/biomedicines12091948] [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: 07/23/2024] [Revised: 08/20/2024] [Accepted: 08/24/2024] [Indexed: 09/30/2024] Open
Abstract
The aging of the world population is closely associated with an increased prevalence of musculoskeletal disorders, such as osteoporosis, sarcopenia, and osteoarthritis, due to common genetic, endocrine, and mechanical risk factors. These conditions are characterized by degeneration of bone, muscle, and cartilage tissue, resulting in an increased risk of fractures and reduced mobility. Importantly, a crucial role in the pathophysiology of these diseases has been proposed for cellular senescence, a state of irreversible cell cycle arrest induced by factors such as DNA damage, telomere shortening, and mitochondrial dysfunction. In addition, senescent cells secrete pro-inflammatory molecules, called senescence-associated secretory phenotype (SASP), which can alter tissue homeostasis and promote disease progression. Undoubtedly, targeting senescent cells and their secretory profiles could promote the development of integrated strategies, including regular exercise and a balanced diet or the use of senolytics and senomorphs, to improve the quality of life of the aging population. Therefore, our review aimed to highlight the role of cellular senescence in age-related musculoskeletal diseases, summarizing the main underlying mechanisms and potential anti-senescence strategies for the treatment of osteoporosis, sarcopenia, and osteoarthritis.
Collapse
Affiliation(s)
- Angela Falvino
- Department of Biomedicine and Prevention, "Tor Vergata" University of Rome, Via Montpellier 1, 00133 Rome, Italy
| | - Beatrice Gasperini
- Department of Biomedicine and Prevention, "Tor Vergata" University of Rome, Via Montpellier 1, 00133 Rome, Italy
| | - Ida Cariati
- Department of Systems Medicine, "Tor Vergata" University of Rome, Via Montpellier 1, 00133 Rome, Italy
| | - Roberto Bonanni
- Department of Biomedicine and Prevention, "Tor Vergata" University of Rome, Via Montpellier 1, 00133 Rome, Italy
| | - Angela Chiavoghilefu
- Department of Orthopaedics and Traumatology, "Policlinico Tor Vergata" Foundation, Viale Oxford 81, 00133 Rome, Italy
| | - Elena Gasbarra
- Department of Orthopaedics and Traumatology, "Policlinico Tor Vergata" Foundation, Viale Oxford 81, 00133 Rome, Italy
| | - Annalisa Botta
- Department of Biomedicine and Prevention, "Tor Vergata" University of Rome, Via Montpellier 1, 00133 Rome, Italy
| | - Virginia Tancredi
- Department of Systems Medicine, "Tor Vergata" University of Rome, Via Montpellier 1, 00133 Rome, Italy
- Centre of Space Bio-Medicine, "Tor Vergata" University of Rome, Via Montpellier 1, 00133 Rome, Italy
| | - Umberto Tarantino
- Department of Orthopaedics and Traumatology, "Policlinico Tor Vergata" Foundation, Viale Oxford 81, 00133 Rome, Italy
- Centre of Space Bio-Medicine, "Tor Vergata" University of Rome, Via Montpellier 1, 00133 Rome, Italy
- Department of Clinical Sciences and Translational Medicine, "Tor Vergata" University of Rome, Via Montpellier 1, 00133 Rome, Italy
| |
Collapse
|
5
|
Duan H, Chen S, Mai X, Fu L, Huang L, Xiao L, Liao M, Chen H, Liu G, Xie L. Low-intensity pulsed ultrasound (LIPUS) promotes skeletal muscle regeneration by regulating PGC-1α/AMPK/GLUT4 pathways in satellite cells/myoblasts. Cell Signal 2024; 117:111097. [PMID: 38355078 DOI: 10.1016/j.cellsig.2024.111097] [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: 11/09/2023] [Revised: 01/19/2024] [Accepted: 02/11/2024] [Indexed: 02/16/2024]
Abstract
Low-Intensity Pulsed Ultrasound (LIPUS) holds therapeutic potential in promoting skeletal muscle regeneration, a biological process mediated by satellite cells and myoblasts. Despite their central roles in regeneration, the detailed mechanistic of LIPUS influence on satellite cells and myoblasts are not fully underexplored. In the current investigation, we administrated LIPUS treatment to injured skeletal muscles and C2C12 myoblasts over five consecutive days. Muscle samples were collected on days 6 and 30 post-injury for an in-depth histological and molecular assessment, both in vivo and in vitro with immunofluorescence analysis. During the acute injury phase, LIPUS treatment significantly augmented the satellite cell population, concurrently enhancing the number and size of newly formed myofibers whilst reducing fibrosis levels. At 30 days post-injury, the LIPUS-treated group demonstrated a more robust satellite cell pool and a higher myofiber count, suggesting that early LIPUS intervention facilitates satellite cell proliferation and differentiation, thereby promoting long-term recovery. Additionally, LIPUS markedly accelerated C2C12 myoblast differentiation, with observed increases in AMPK phosphorylation in myoblasts, leading to elevated expression of Glut4 and PGC-1α, and subsequent glucose uptake and mitochondrial biogenesis. These findings imply that LIPUS-induced modulation of myoblasts may culminate in enhanced cellular energy availability, laying a theoretical groundwork for employing LIPUS in ameliorating skeletal muscle regeneration post-injury. NEW & NOTEWORTHY: Utilizing the cardiotoxin (CTX) muscle injury model, we investigated the influence of LIPUS on satellite cell homeostasis and skeletal muscle regeneration. Our findings indicate that LIPUS promotes satellite cell proliferation and differentiation, thereby facilitating skeletal muscle repair. Additionally, in vitro investigations lend credence to the hypothesis that the regulatory effect of LIPUS on satellite cells may be attributed to its capability to enhance cellular energy metabolism.
Collapse
Affiliation(s)
- Huimin Duan
- Department of Rehabilitation Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510000, China
| | - Shujie Chen
- 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 Anesthesiology, The Seventh Affiliated Hospital, Southern Medical University, Foshan 528244, Guangdong, China
| | - Xudong Mai
- Department of Endocrinology and Metabolism, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Liping Fu
- Department of Rehabilitation Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510000, China
| | - Liujing Huang
- Medical Affairs Department, Guangzhou Betrue Technology Co., Ltd, Guangzhou 510700, China
| | - Lanling Xiao
- 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; 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, Hunan 410125, China
| | - Miaomiao Liao
- 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
| | - Hong Chen
- Department of Endocrinology and Metabolism, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Gang Liu
- Department of Rehabilitation Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510000, China.
| | - Liwei Xie
- 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 Anesthesiology, The Seventh Affiliated Hospital, Southern Medical University, Foshan 528244, Guangdong, China; Department of Internal Medicine, Shunde Women and Children's Hospital (Maternity and Child Healthcare Hospital of Shunde Foshan), Guangdong Medical University, Foshan, Guangdong, China; Department of Endocrinology and Metabolism, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China; College of Life and Health Sciences, Guangdong Industry Polytechnic, Guangzhou, Guangdong 510300, China.
| |
Collapse
|
6
|
Ruple BA, Mattingly ML, Godwin JS, McIntosh MC, Kontos NJ, Agyin-Birikorang A, Michel JM, Plotkin DL, Chen SY, Ziegenfuss TN, Fruge AD, Gladden LB, Robinson AT, Mobley CB, Mackey AL, Roberts MD. The effects of resistance training on denervated myofibers, senescent cells, and associated protein markers in middle-aged adults. FASEB J 2024; 38:e23621. [PMID: 38651653 PMCID: PMC11047210 DOI: 10.1096/fj.202302103rrr] [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: 10/16/2023] [Revised: 04/04/2024] [Accepted: 04/08/2024] [Indexed: 04/25/2024]
Abstract
Denervated myofibers and senescent cells are hallmarks of skeletal muscle aging. However, sparse research has examined how resistance training affects these outcomes. We investigated the effects of unilateral leg extensor resistance training (2 days/week for 8 weeks) on denervated myofibers, senescent cells, and associated protein markers in apparently healthy middle-aged participants (MA, 55 ± 8 years old, 17 females, 9 males). We obtained dual-leg vastus lateralis (VL) muscle cross-sectional area (mCSA), VL biopsies, and strength assessments before and after training. Fiber cross-sectional area (fCSA), satellite cells (Pax7+), denervated myofibers (NCAM+), senescent cells (p16+ or p21+), proteins associated with denervation and senescence, and senescence-associated secretory phenotype (SASP) proteins were analyzed from biopsy specimens. Leg extensor peak torque increased after training (p < .001), while VL mCSA trended upward (interaction p = .082). No significant changes were observed for Type I/II fCSAs, NCAM+ myofibers, or senescent (p16+ or p21+) cells, albeit satellite cells increased after training (p = .037). While >90% satellite cells were not p16+ or p21+, most p16+ and p21+ cells were Pax7+ (>90% on average). Training altered 13 out of 46 proteins related to muscle-nerve communication (all upregulated, p < .05) and 10 out of 19 proteins related to cellular senescence (9 upregulated, p < .05). Only 1 out of 17 SASP protein increased with training (IGFBP-3, p = .031). In conclusion, resistance training upregulates proteins associated with muscle-nerve communication in MA participants but does not alter NCAM+ myofibers. Moreover, while training increased senescence-related proteins, this coincided with an increase in satellite cells but not alterations in senescent cell content or SASP proteins. These latter findings suggest shorter term resistance training is an unlikely inducer of cellular senescence in apparently healthy middle-aged participants. However, similar study designs are needed in older and diseased populations before definitive conclusions can be drawn.
Collapse
Affiliation(s)
| | | | | | | | | | | | - J. Max Michel
- School of Kinesiology, Auburn University, Auburn, AL, USA
| | | | | | | | | | | | | | | | - Abigail L. Mackey
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, DK
- Institute of Sports Medicine Copenhagen, Department of Orthopaedic Surgery, Copenhagen University Hospital – Bispebjerg and Frederiksberg, Copenhagen, Denmark
| | | |
Collapse
|
7
|
Ungvari Z, Fazekas-Pongor V, Csiszar A, Kunutsor SK. The multifaceted benefits of walking for healthy aging: from Blue Zones to molecular mechanisms. GeroScience 2023; 45:3211-3239. [PMID: 37495893 PMCID: PMC10643563 DOI: 10.1007/s11357-023-00873-8] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Accepted: 07/11/2023] [Indexed: 07/28/2023] Open
Abstract
Physical activity, including walking, has numerous health benefits in older adults, supported by a plethora of observational and interventional studies. Walking decreases the risk or severity of various health outcomes such as cardiovascular and cerebrovascular diseases, type 2 diabetes mellitus, cognitive impairment and dementia, while also improving mental well-being, sleep, and longevity. Dose-response relationships for walking duration and intensity are established for adverse cardiovascular outcomes. Walking's favorable effects on cardiovascular risk factors are attributed to its impact on circulatory, cardiopulmonary, and immune function. Meeting current physical activity guidelines by walking briskly for 30 min per day for 5 days can reduce the risk of several age-associated diseases. Additionally, low-intensity physical exercise, including walking, exerts anti-aging effects and helps prevent age-related diseases, making it a powerful tool for promoting healthy aging. This is exemplified by the lifestyles of individuals in Blue Zones, regions of the world with the highest concentration of centenarians. Walking and other low-intensity physical activities contribute significantly to the longevity of individuals in these regions, with walking being an integral part of their daily lives. Thus, incorporating walking into daily routines and encouraging walking-based physical activity interventions can be an effective strategy for promoting healthy aging and improving health outcomes in all populations. The goal of this review is to provide an overview of the vast and consistent evidence supporting the health benefits of physical activity, with a specific focus on walking, and to discuss the impact of walking on various health outcomes, including the prevention of age-related diseases. Furthermore, this review will delve into the evidence on the impact of walking and low-intensity physical activity on specific molecular and cellular mechanisms of aging, providing insights into the underlying biological mechanisms through which walking exerts its beneficial anti-aging effects.
Collapse
Affiliation(s)
- Zoltan Ungvari
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Department of Public Health, Semmelweis University, Budapest, Hungary.
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
| | | | - Anna Csiszar
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Department of Translational Medicine, Semmelweis University, Budapest, Hungary
| | - Setor K Kunutsor
- Diabetes Research Centre, University of Leicester, Leicester General Hospital, Gwendolen Road, Leicester, LE5 4WP, UK.
| |
Collapse
|
8
|
Pasini E, Corsetti G, Dioguardi FS. Nutritional Supplementation and Exercise as Essential Allies in the Treatment of Chronic Heart Failure: The Metabolic and Molecular Bases. Nutrients 2023; 15:nu15102337. [PMID: 37242219 DOI: 10.3390/nu15102337] [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: 04/18/2023] [Revised: 05/07/2023] [Accepted: 05/15/2023] [Indexed: 05/28/2023] Open
Abstract
Chronic heart failure (CHF) is one of principal health problems in industrialized countries. Despite therapeutical improvement, based on drugs and exercise training, it is still characterized by elevated mortality and morbidity. Data show that protein energy malnutrition, clinically evident primarily with sarcopenia, is present in more than 50% of CHF patients and is an independent factor of CHF prognosis. Several pathophysiological mechanisms, primarily due to the increase in blood hypercatabolic molecules, have been proposed to explain this phenomenon. Nutritional supplementation with proteins, amino acids, vitamins and antioxidants have all been used to treat malnutrition. However, the success and efficacy of these procedures are often contradictory and not conclusive. Interestingly, data on exercise training show that exercise reduces mortality and increases functional capacity, although it also increases the catabolic state with energy expenditure and nitrogen-providing substrate needs. Therefore, this paper discusses the molecular mechanisms of specific nutritional supplementation and exercise training that may improve anabolic pathways. In our opinion, the relationship between exercise and the mTOR complex subunit as Deptor and/or related signaling proteins, such as AMPK or sestrin, is pivotal. Consequently, concomitantly with traditional medical therapies, we have proposed a combination of personalized and integrated nutritional supplementation, as well as exercise to treat malnutrition, and anthropometric and functional CHF-related disorders.
Collapse
Affiliation(s)
- Evasio Pasini
- Department of Clinical and Experimental Sciences, University of Brescia, 25100 Brescia, Italy
- Italian Association of Functional Medicine, 20855 Lesmo, Italy
| | - Giovanni Corsetti
- Department of Clinical and Experimental Sciences, University of Brescia, 25100 Brescia, Italy
| | | |
Collapse
|
9
|
Chen XK, Zheng C, Wong SHS, Ma ACH. Moderate-vigorous physical activity attenuates premature senescence of immune cells in sedentary adults with obesity: a pilot randomized controlled trial. Aging (Albany NY) 2022; 14:10137-10152. [PMID: 36585923 PMCID: PMC9831733 DOI: 10.18632/aging.204458] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Accepted: 12/20/2022] [Indexed: 12/31/2022]
Abstract
Despite the well-known senolytic effects of physical exercise on immune cells in older adults, the effect of physical activity (PA) on premature immune senescence in sedentary adults with obesity remains largely unknown. This pilot study aimed to investigate the role of objectively measured physical behaviors and Fitbit watch-based free-living PA intervention in premature senescence of immune cells in sedentary adults with obesity. Forty-five participants were recruited in the cross-sectional analysis, and forty of them further participated in the randomized controlled trial. We found that objectively measured moderate-vigorous PA was independently and inversely correlated with the expression of p16INK4a and p21Cip1 in the peripheral blood mononuclear cell (PBMCs) of adults with obesity; however, chronological age, body mass index, body fat, maximal oxygen consumption, light PA, sedentary behaviors, and sleep duration were not. More importantly, the 12-week PA intervention mitigated the elevated p16INK4a levels in PBMCs, though it showed no effect on p21Cip1 and senescence-associated secretory phenotypes. Taken together, physical inactivity is an independent determinant of premature senescence in immune cells, while the 12-week PA intervention is a promising strategy to alleviate premature immune senescence in adults with obesity.
Collapse
Affiliation(s)
- Xiang-Ke Chen
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hung Hom, Hong Kong SAR, China
| | - Chen Zheng
- Department of Sports Science and Physical Education, The Chinese University of Hong Kong, Sha Tin, Hong Kong SAR, China
| | - Stephen Heung-Sang Wong
- Department of Sports Science and Physical Education, The Chinese University of Hong Kong, Sha Tin, Hong Kong SAR, China
| | - Alvin Chun-Hang Ma
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hung Hom, Hong Kong SAR, China
| |
Collapse
|
10
|
Zhang X, Englund DA, Aversa Z, Jachim SK, White TA, LeBrasseur NK. Exercise Counters the Age-Related Accumulation of Senescent Cells. Exerc Sport Sci Rev 2022; 50:213-221. [PMID: 35776782 PMCID: PMC9680689 DOI: 10.1249/jes.0000000000000302] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
We propose the beneficial effects of exercise are in part mediated through the prevention and elimination of senescent cells. Exercise counters multiple forms of age-related molecular damage that initiate the senescence program and activates immune cells responsible for senescent cell clearance. Preclinical and clinical evidence for exercise as a senescence-targeting therapy and areas needing further investigation are discussed.
Collapse
Affiliation(s)
- Xu Zhang
- Robert and Arlene Kogod Center on Aging, Rochester, MN
- Department of Physical Medicine and Rehabilitation, Mayo Clinic, Rochester, MN
| | - Davis A. Englund
- Robert and Arlene Kogod Center on Aging, Rochester, MN
- Department of Physical Medicine and Rehabilitation, Mayo Clinic, Rochester, MN
| | - Zaira Aversa
- Robert and Arlene Kogod Center on Aging, Rochester, MN
- Department of Physical Medicine and Rehabilitation, Mayo Clinic, Rochester, MN
| | - Sarah K. Jachim
- Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN
| | | | - Nathan K. LeBrasseur
- Robert and Arlene Kogod Center on Aging, Rochester, MN
- Department of Physical Medicine and Rehabilitation, Mayo Clinic, Rochester, MN
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN
| |
Collapse
|
11
|
Zhu H, Zeng W, Zhao T, Shi W, Dong X, Zhang A, Li X, Xu L. Synthesis and evaluation of 5-aminimidazole-4-carboxamide riboside derivatives as anti-fatigue agents. ARAB J CHEM 2022. [DOI: 10.1016/j.arabjc.2022.104313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
|
12
|
Lee SM, Lee MC, Bae WR, Yoon KJ, Moon HY. Muscle fiber type-dependence effect of exercise on genomic networks in aged mice models. Aging (Albany NY) 2022; 14:3337-3364. [PMID: 35440516 PMCID: PMC9085230 DOI: 10.18632/aging.204024] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Accepted: 04/12/2022] [Indexed: 11/29/2022]
Abstract
Skeletal muscles are made up of various muscle fiber type including slow and fast-twitch fibers. Because each muscle fiber has its own physiological characteristics, the effects of aging and exercise vary depending on the type of muscle fiber. We used bioinformatics screening techniques such as differentially expressed gene analysis, gene ontology analysis and gene set enrichment analysis, to try to understand the genetic differences between muscle fiber types. The experiment and gene expression profiling in this study used the soleus (SOL, slow-twitch muscle) and gastrocnemius (GAS, fast-twitch muscle). According to our findings, fatty acid metabolism is significantly up-regulated in SOL compared to GAS, whereas the glucose metabolism pathway is significantly down-regulated in SOL compared to GAS. Furthermore, apoptosis and myogenesis patterns differ between SOL and GAS. SOL did not show differences in apoptosis due to the aging effect, but apoptosis in GAS was significantly up-regulated with age. Apoptosis in GAS of old groups is significantly reduced after 4 weeks of aerobic exercise, but no such finding was found in SOL. In terms of myogenesis, exercise intervention up-regulated this process in GAS of old groups but not in SOL. Taken together, muscle fiber type significantly interacts with aging and exercise. Despite the importance of the interaction between these factors, large-scale gene expression data has rarely been studied. We hope to contribute to a better understanding of the relationship between muscle fiber type, aging and exercise at the molecular level.
Collapse
Affiliation(s)
- Sun Min Lee
- Department of Physical Education, Seoul National University, Seoul, South Korea
| | - Min Chul Lee
- Department of Sports Medicine, College of Health Science, CHA University, Pocheon, South Korea
| | - Woo Ri Bae
- Department of Physical Education, Seoul National University, Seoul, South Korea
| | - Kyung Jin Yoon
- Department of Physical Education, Seoul National University, Seoul, South Korea
| | - Hyo Youl Moon
- Department of Physical Education, Seoul National University, Seoul, South Korea.,Institute of Sport Science, Seoul National University, Seoul, South Korea.,Institute on Aging, Seoul National University, Seoul, South Korea
| |
Collapse
|
13
|
Andreou C, Matsakas A. Current insights into cellular senescence and myotoxicity induced by doxorubicin. Int J Sports Med 2022; 43:1084-1096. [PMID: 35288882 DOI: 10.1055/a-1797-7622] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Doxorubicin is an anti-neoplasmic drug that prevents DNA replication but induces senescence and cellular toxicity. Intensive research has focused on strategies to alleviate the doxorubicin-induced skeletal myotoxicity. The aim of the present review is to critically discuss the relevant scientific evidence about the role of exercise and growth factor administration and offer novel insights about newly developed-tools to combat the adverse drug reactions of doxorubicin treatment on skeletal muscle. In the first part, we discuss current data and mechanistic details on the impact of doxorubicin on skeletal myotoxicity. We next, review key aspects about the role of regular exercise and the impact of growth factors either administered pharmacologically or via genetic interventions. Future strategies such as combination of exercise and growth factor administration remain to be established to combat the pharmacologically-induced myotoxicity.
Collapse
Affiliation(s)
- Charalampos Andreou
- Hull York Medical School, University of Hull, Hull, United Kingdom of Great Britain and Northern Ireland
| | - Antonios Matsakas
- Hull York Medical School, University of Hull, Hull, United Kingdom of Great Britain and Northern Ireland
| |
Collapse
|
14
|
Babaei P, Hoseini R. Exercise training modulates adipokine dysregulations in metabolic syndrome. SPORTS MEDICINE AND HEALTH SCIENCE 2022; 4:18-28. [PMID: 35782776 PMCID: PMC9219261 DOI: 10.1016/j.smhs.2022.01.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 01/01/2022] [Accepted: 01/07/2022] [Indexed: 12/16/2022] Open
Abstract
Metabolic syndrome (MetS) is a cluster of risk factors for various metabolic diseases, and it is characterized by central obesity, dyslipidemia, hypertension, and insulin resistance. The core component for MetS is adipose tissue, which releases adipokines and influences physical health. Adipokines consist of pro and anti-inflammatory cytokines and contribute to various physiological functions. Generally, a sedentary lifestyle promotes fat accumulation and secretion of pro-inflammatory adipokines. However, regular exercise has been known to exert various beneficial effects on metabolic and cognitive disorders. Although the mechanisms underlying exercise beneficial effects in MetS are not fully understood, changes in energy expenditure, fat accumulation, circulatory level of myokines, and adipokines might be involved. This review article focuses on some of the selected adipokines in MetS, and their responses to exercise training considering possible mechanisms.
Collapse
Affiliation(s)
- Parvin Babaei
- Cellular & Molecular Research Center, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
- Neuroscience Research Center, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
- Department of Physiology, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Rastegar Hoseini
- Department of Sports Physiology, Faculty of Sport Sciences, Razi University, Kermanshah, Iran
| |
Collapse
|
15
|
Babaei P, Azari HB. Exercise Training Improves Memory Performance in Older Adults: A Narrative Review of Evidence and Possible Mechanisms. Front Hum Neurosci 2022; 15:771553. [PMID: 35153701 PMCID: PMC8829997 DOI: 10.3389/fnhum.2021.771553] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Accepted: 12/16/2021] [Indexed: 12/15/2022] Open
Abstract
As human life expectancy increases, cognitive decline and memory impairment threaten independence and quality of life. Therefore, finding prevention and treatment strategies for memory impairment is an important health concern. Moreover, a better understanding of the mechanisms involved underlying memory preservation will enable the development of appropriate pharmaceuticals drugs for those who are activity limited. Exercise training as a non-pharmacological tool, has been known to increase the mean lifespan by maintaining general body health and improving the cardiovascular and nervous systems function. Among different exercise training protocols, aerobic exercise has been reported to prevent the progression of memory decline, provided adequate exertion level, duration, and frequency. Mechanisms underlying exercise training effects on memory performance have not been understood yet. Convergent evidence suggest several direct and indirect mechanisms at molecular and supramolecular levels. The supramolecular level includes improvement in blood circulation, synaptic plasticity and neurogenesis which are under controls of complex molecular signaling of neurotransmitters, neurotrophic factors, exerkines, and epigenetics factors. Among these various factors, irisin/BDNF signaling seems to be one of the important mediators of crosstalk between contracted skeletal muscles and the brain during exercise training. This review provides an affordable and effective method to improve cognitive function in old ages, particularly those who are most vulnerable to neurodegenerative disorders.
Collapse
Affiliation(s)
- Parvin Babaei
- Neuroscience Research Center, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
- Cellular and Molecular Research Center, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
- Department of Physiology, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Helya Bolouki Azari
- Neuroscience Research Center, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
- Department of Physiology, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
16
|
Hu MC, Moe OW. Phosphate and Cellular Senescence. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1362:55-72. [PMID: 35288873 PMCID: PMC10513121 DOI: 10.1007/978-3-030-91623-7_7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Cellular senescence is one type of permeant arrest of cell growth and one of increasingly recognized contributor to aging and age-associated disease. High phosphate and low Klotho individually and synergistically lead to age-related degeneration in multiple organs. Substantial evidence supports the causality of high phosphate in cellular senescence, and potential contribution to human aging, cancer, cardiovascular, kidney, neurodegenerative, and musculoskeletal diseases. Phosphate can induce cellular senescence both by direct phosphotoxicity, and indirectly through downregulation of Klotho and upregulation of plasminogen activator inhibitor-1. Restriction of dietary phosphate intake and blockage of intestinal absorption of phosphate help suppress cellular senescence. Supplementation of Klotho protein, cellular senescence inhibitor, and removal of senescent cells with senolytic agents are potential novel strategies to attenuate phosphate-induced cellular senescence, retard aging, and ameliorate age-associated, and phosphate-induced disorders.
Collapse
Affiliation(s)
- Ming Chang Hu
- Charles and Jane Pak Center for Mineral Metabolism and Clinical Research, University of Texas Southwestern Medical Center, Dallas, TX, USA.
- Departments of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| | - Orson W Moe
- Charles and Jane Pak Center for Mineral Metabolism and Clinical Research, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Departments of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Departments of Physiology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
17
|
Yan X, Shen Z, Yu D, Zhao C, Zou H, Ma B, Dong W, Chen W, Huang D, Yu Z. Nrf2 contributes to the benefits of exercise interventions on age-related skeletal muscle disorder via regulating Drp1 stability and mitochondrial fission. Free Radic Biol Med 2022; 178:59-75. [PMID: 34823019 DOI: 10.1016/j.freeradbiomed.2021.11.030] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 11/03/2021] [Accepted: 11/20/2021] [Indexed: 02/09/2023]
Abstract
The progressive and generalized loss of skeletal muscle mass and function, also known as sarcopenia, underlies disability, increasing adverse outcomes and poor quality of life in older people. Exercise interventions are commonly recommended as the primary treatment for sarcopenia. Nuclear factor erythroid 2-related factor 2 (Nrf2) plays a vital role in regulating metabolism, mitochondrial function, and the ROS-dependent adaptations of skeletal muscle, as the response to exercise. To investigate the contribution of Nrf2 to the benefits of exercise interventions in older age, aged (∼22 month old) Nrf2 knockout (Nrf2-KO) mice and age-matched wild-type (WT) C57BL6/J mice were randomly divided into 2 groups (sedentary or exercise group). We found that exercise interventions improved skeletal muscle function and restored the sarcopenia-like phenotype in WT mice, accompanied with the increasing mRNA level of Nrf2. While these alternations were minimal in Nrf2-KO mice after exercise. Further studies indicated that Nrf2 could increase the stability of Drp1 through deubiquitinating and promote Drp1-dependent mitochondrial fission to attenuate mitochondrial disorder. We also observed the effects of sulforaphane (SFN), a Nrf2 activator, in restoring mitochondrial function in senescent C2C12 cells and improving sarcopenia in older WT mice, which were abolished by Nrf2 deficiency. These results indicated that some benefits of exercise intervention to skeletal muscle were Nrf2 mediated, and a future work should focus on Nrf2 signaling to identify a pharmacological treatment for sarcopenia.
Collapse
Affiliation(s)
- Xialin Yan
- Department of Gastrointestinal Surgery, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Zile Shen
- Department of Gastrointestinal Surgery, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Dingye Yu
- Department of General Surgery, Shanghai Minimally Invasive Surgery Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chongke Zhao
- Department of Medical Ultrasound, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Hongbo Zou
- Department of Gastrointestinal Surgery, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China; Department of Gastrointestinal Surgery, People's Hospital of Deyang City, Deyang, Sichuan, China
| | - Bingwei Ma
- Department of Gastrointestinal Surgery, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Wenxi Dong
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Wenhao Chen
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Dongdong Huang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.
| | - Zhen Yu
- Department of Gastrointestinal Surgery, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China; Department of Gastrointestinal Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.
| |
Collapse
|
18
|
Ou HC, Chu PM, Huang YT, Cheng HC, Chou WC, Yang HL, Chen HI, Tsai KL. Low-level laser prevents doxorubicin-induced skeletal muscle atrophy by modulating AMPK/SIRT1/PCG-1α-mediated mitochondrial function, apoptosis and up-regulation of pro-inflammatory responses. Cell Biosci 2021; 11:200. [PMID: 34876217 PMCID: PMC8650328 DOI: 10.1186/s13578-021-00719-w] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Accepted: 11/22/2021] [Indexed: 02/06/2023] Open
Abstract
Background Doxorubicin (Dox) is a widely used anthracycline drug to treat cancer, yet numerous adverse effects influencing different organs may offset the treatment outcome, which in turn affects the patient’s quality of life. Low-level lasers (LLLs) have resulted in several novel indications in addition to traditional orthopedic conditions, such as increased fatigue resistance and muscle strength. However, the mechanisms by which LLL irradiation exerts beneficial effects on muscle atrophy are still largely unknown. Results The present study aimed to test our hypothesis that LLL irradiation protects skeletal muscles against Dox-induced muscle wasting by using both animal and C2C12 myoblast cell models. We established SD rats treated with 4 consecutive Dox injections (12 mg/kg cumulative dose) and C2C12 myoblast cells incubated with 2 μM Dox to explore the protective effects of LLL irradiation. We found that LLL irradiation markedly alleviated Dox-induced muscle wasting in rats. Additionally, LLL irradiation inhibited Dox-induced mitochondrial dysfunction, apoptosis, and oxidative stress via the activation of AMPK and upregulation of SIRT1 with its downstream signaling PGC-1α. These aforementioned beneficial effects of LLL irradiation were reversed by knockdown AMPK, SIRT1, and PGC-1α in C2C12 cells transfected with siRNA and were negated by cotreatment with mitochondrial antioxidant and P38MAPK inhibitor. Therefore, AMPK/SIRT1/PGC-1α pathway activation may represent a new mechanism by which LLL irradiation exerts protection against Dox myotoxicity through preservation of mitochondrial homeostasis and alleviation of oxidative stress and apoptosis. Conclusion Our findings may provide a novel adjuvant intervention that can potentially benefit cancer patients from Dox-induced muscle wasting. Supplementary Information The online version contains supplementary material available at 10.1186/s13578-021-00719-w.
Collapse
Affiliation(s)
- Hsiu-Chung Ou
- Department of Physical Therapy, College of Medical and Health Science, Asia University, Taichung, Taiwan, ROC
| | - Pei-Ming Chu
- Department of Anatomy, School of Medicine, China Medical University, Taichung, Taiwan, ROC
| | - Yu-Ting Huang
- Department of Physical Therapy, College of Medicine, National Cheng Kung University, Tainan, Taiwan, ROC
| | - Hui-Ching Cheng
- Department of Physical Therapy, College of Medicine, National Cheng Kung University, Tainan, Taiwan, ROC
| | - Wan-Ching Chou
- Department of Physical Therapy, College of Medicine, National Cheng Kung University, Tainan, Taiwan, ROC
| | - Hsin-Lun Yang
- Department of Physical Therapy, College of Medicine, National Cheng Kung University, Tainan, Taiwan, ROC.,Institute of Allied Health Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan, ROC
| | - Hsiu-I Chen
- Department of Physical Therapy, College of Medical and Health Science, Asia University, Taichung, Taiwan, ROC.,Department of Physical Therapy, Hungkuang University, Taichung, Taiwan, ROC
| | - Kun-Ling Tsai
- Department of Physical Therapy, College of Medicine, National Cheng Kung University, Tainan, Taiwan, ROC. .,Institute of Allied Health Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan, ROC.
| |
Collapse
|
19
|
Saito Y, Chikenji TS. Diverse Roles of Cellular Senescence in Skeletal Muscle Inflammation, Regeneration, and Therapeutics. Front Pharmacol 2021; 12:739510. [PMID: 34552495 PMCID: PMC8450382 DOI: 10.3389/fphar.2021.739510] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2021] [Accepted: 08/23/2021] [Indexed: 12/19/2022] Open
Abstract
Skeletal muscle undergoes vigorous tissue remodeling after injury. However, aging, chronic inflammatory diseases, sarcopenia, and neuromuscular disorders cause muscle loss and degeneration, resulting in muscular dysfunction. Cellular senescence, a state of irreversible cell cycle arrest, acts during normal embryonic development and remodeling after tissue damage; when these processes are complete, the senescent cells are eliminated. However, the accumulation of senescent cells is a hallmark of aging tissues or pathological contexts and may lead to progressive tissue degeneration. The mechanisms responsible for the effects of senescent cells have not been fully elucidated. Here, we review current knowledge about the beneficial and detrimental effects of senescent cells in tissue repair, regeneration, aging, and age-related disease, especially in skeletal muscle. We also discuss how senescence of muscle stem cells and muscle-resident fibro-adipogenic progenitors affects muscle pathologies or regeneration, and consider the possibility that immunosenescence leads to muscle pathogenesis. Finally, we explore senotherapy, the therapeutic targeting of senescence to treat age-related disease, from the standpoint of improving muscle regeneration.
Collapse
Affiliation(s)
- Yuki Saito
- Department of Anatomy, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Takako S Chikenji
- Department of Anatomy, Sapporo Medical University School of Medicine, Sapporo, Japan.,Department of Health Sciences, School of Medicine, Hokkaido University, Sapporo, Japan
| |
Collapse
|
20
|
Yoon KJ, Park S, Kwak SH, Moon HY. Effects of Voluntary Running Wheel Exercise-Induced Extracellular Vesicles on Anxiety. Front Mol Neurosci 2021; 14:665800. [PMID: 34276303 PMCID: PMC8280765 DOI: 10.3389/fnmol.2021.665800] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Accepted: 05/20/2021] [Indexed: 11/13/2022] Open
Abstract
Anxiety disorders are the most frequently diagnosed psychological condition, associated with serious comorbidities including excessive fear and interference with daily life. Drugs for anxiety disorders are typically prescribed but the side effects include weight gain, nausea, and sleepiness. Exercise is an effective treatment for anxiety. Exercise induces the release of extracellular vesicles (EVs) into the circulation, which transmit signals between organs. However, the effects of exercise-induced EVs on anxiety remain poorly understood. Here, we isolated EVs from the sera of mice that were sedentary or that voluntarily exercised. We characterized the changes in the miRNA profile of serum EVs after 4 weeks of voluntary exercise. miRNA sequencing showed that 82 miRNAs (46 of which were positive and 36 negative regulators) changed after exercise. We selected genes affected by at least two miRNAs. Of these, 27.27% were associated with neurotrophin signaling (9.09% with each of central nervous system neuronal development, cerebral cortical cell migration, and peripheral neuronal development). We also analyzed behavioral changes in mice with 3 weeks of restraint stress-induced anxiety after injection of 20 μg amounts of EVs from exercised or sedentary mice into the left cerebral ventricle. We found that exercise-derived EVs reduced anxiety (compared to a control group) in a nest-building test but found no between-group differences in the rotarod or open field tests. Exercise-derived EVs enhanced the expression of neuroactive ligand-receptor interaction genes. Thus, exercise-derived EVs may exhibit anti-anxiety effects and may be of therapeutic utility.
Collapse
Affiliation(s)
- Kyeong Jin Yoon
- Department of Physical Education, Seoul National University, Seoul, South Korea
| | - Suhong Park
- Department of Physical Education, Seoul National University, Seoul, South Korea
| | - Seung Hee Kwak
- Department of Physical Education, Seoul National University, Seoul, South Korea
| | - Hyo Youl Moon
- Department of Physical Education, Seoul National University, Seoul, South Korea.,Institute of Sport Science, Seoul National University, Seoul, South Korea.,Institute on Aging, Seoul National University, Seoul, South Korea
| |
Collapse
|
21
|
Yan C, Xu Z, Huang W. Cellular Senescence Affects Cardiac Regeneration and Repair in Ischemic Heart Disease. Aging Dis 2021; 12:552-569. [PMID: 33815882 PMCID: PMC7990367 DOI: 10.14336/ad.2020.0811] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Accepted: 08/11/2020] [Indexed: 01/10/2023] Open
Abstract
Ischemic heart disease (IHD) is defined as a syndrome of ischemic cardiomyopathy. Myogenesis and angiogenesis in the ischemic myocardium are important for cardiomyocyte (CM) survival, improving cardiac function and decreasing the progression of heart failure after IHD. Cellular senescence is a state of permanent irreversible cell cycle arrest caused by stress that results in a decline in cellular functions, such as proliferation, migration, homing, and differentiation. In addition, senescent cells produce the senescence-associated secretory phenotype (SASP), which affects the tissue microenvironment and surrounding cells by secreting proinflammatory cytokines, chemokines, growth factors, and extracellular matrix degradation proteins. The accumulation of cardiovascular-related senescent cells, including vascular endothelial cells (VECs), vascular smooth muscle cells (VSMCs), CMs and progenitor cells, is an important risk factor of cardiovascular diseases, such as vascular aging, atherosclerotic plaque formation, myocardial infarction (MI) and ventricular remodeling. This review summarizes the processes of angiogenesis, myogenesis and cellular senescence after IHD. In addition, this review focuses on the relationship between cellular senescence and cardiovascular disease and the mechanism of cellular senescence. Finally, we discuss a potential therapeutic strategy for MI targeting senescent cells.
Collapse
Affiliation(s)
- Chi Yan
- Department of Geriatric Cardiology, The First Affiliated Hospital of Guangxi Medical University, Guangxi, China.
- Guangxi Key Laboratory of Precision Medicine in Cardio-cerebrovascular Diseases Control and Prevention, Guangxi, China.
- Department of Cardiology, Guangxi Clinical Research Center for Cardio-cerebrovascular Diseases, Guangxi, China.
| | - Zhimeng Xu
- Department of Cardiology, The People's Hospital of Guangxi Zhuang Autonomous Region, Guangxi, China.
| | - Weiqiang Huang
- Department of Geriatric Cardiology, The First Affiliated Hospital of Guangxi Medical University, Guangxi, China.
- Guangxi Key Laboratory of Precision Medicine in Cardio-cerebrovascular Diseases Control and Prevention, Guangxi, China.
- Department of Cardiology, Guangxi Clinical Research Center for Cardio-cerebrovascular Diseases, Guangxi, China.
| |
Collapse
|
22
|
Yu M, Zhang H, Wang B, Zhang Y, Zheng X, Shao B, Zhuge Q, Jin K. Key Signaling Pathways in Aging and Potential Interventions for Healthy Aging. Cells 2021; 10:cells10030660. [PMID: 33809718 PMCID: PMC8002281 DOI: 10.3390/cells10030660] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2021] [Revised: 03/11/2021] [Accepted: 03/12/2021] [Indexed: 12/12/2022] Open
Abstract
Aging is a fundamental biological process accompanied by a general decline in tissue function. Indeed, as the lifespan increases, age-related dysfunction, such as cognitive impairment or dementia, will become a growing public health issue. Aging is also a great risk factor for many age-related diseases. Nowadays, people want not only to live longer but also healthier. Therefore, there is a critical need in understanding the underlying cellular and molecular mechanisms regulating aging that will allow us to modify the aging process for healthy aging and alleviate age-related disease. Here, we reviewed the recent breakthroughs in the mechanistic understanding of biological aging, focusing on the adenosine monophosphate-activated kinase (AMPK), Sirtuin 1 (SIRT1) and mammalian target of rapamycin (mTOR) pathways, which are currently considered critical for aging. We also discussed how these proteins and pathways may potentially interact with each other to regulate aging. We further described how the knowledge of these pathways may lead to new interventions for antiaging and against age-related disease.
Collapse
Affiliation(s)
- Mengdi Yu
- Zhejiang Provincial Key Laboratory of Aging and Neurological Disorder Research, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China; (M.Y.); (Y.Z.); (X.Z.)
| | - Hongxia Zhang
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA;
| | - Brian Wang
- Pathnova Laboratories Pte. Ltd. 1 Research Link, Singapore 117604, Singapore;
| | - Yinuo Zhang
- Zhejiang Provincial Key Laboratory of Aging and Neurological Disorder Research, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China; (M.Y.); (Y.Z.); (X.Z.)
| | - Xiaoying Zheng
- Zhejiang Provincial Key Laboratory of Aging and Neurological Disorder Research, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China; (M.Y.); (Y.Z.); (X.Z.)
| | - Bei Shao
- Department of Neurology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China;
| | - Qichuan Zhuge
- Zhejiang Provincial Key Laboratory of Aging and Neurological Disorder Research, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China; (M.Y.); (Y.Z.); (X.Z.)
- Correspondence: (Q.Z.); (K.J.); Tel.: +86-577-55579339 (Q.Z.); +1-81-7735-2579 (K.J.)
| | - Kunlin Jin
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
- Correspondence: (Q.Z.); (K.J.); Tel.: +86-577-55579339 (Q.Z.); +1-81-7735-2579 (K.J.)
| |
Collapse
|
23
|
Yoon KJ, Ahn A, Park SH, Kwak SH, Kwak SE, Lee W, Yang YR, Kim M, Shin HM, Kim HR, Moon HY. Exercise reduces metabolic burden while altering the immune system in aged mice. Aging (Albany NY) 2021; 13:1294-1313. [PMID: 33406502 PMCID: PMC7834985 DOI: 10.18632/aging.202312] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Accepted: 11/13/2020] [Indexed: 02/06/2023]
Abstract
Although several evidence has suggested the impact of exercise on the prevention of aging phenotypes, few studies have been conducted on the mechanism by which exercise alters the immune-cell profile, thereby improving metabolism in senile obesity. In this study, we confirmed that 4-week treadmill exercise sufficiently improved metabolic function, including increased lean mass and decreased fat mass, in 88-week-old mice. The expression level of the senescence marker p16 in the white adipose tissue (WAT) was decreased after 4-weeks of exercise. Exercise induced changes in the profiles of immune-cell subsets, including natural killer (NK) cells, central memory CD8+ T cells, eosinophils, and neutrophils, in the stromal vascular fraction of WAT. In addition, it has been shown through transcriptome analysis of WAT that exercise can activate pathways involved in the interaction between WAT and immune cells, in particular NK cells, in aged mice. These results suggest that exercise has a profound effect on changes in immune-cell distribution and senescent-cell scavenging in WAT of aged mice, eventually affecting overall energy metabolism toward a more youthful state.
Collapse
Affiliation(s)
- Kyeong Jin Yoon
- Department of Physical Education, Seoul National University, Gwanak-ro, Gwanak-gu, Seoul, Republic of Korea.,Institute of Sport Science, Seoul National University, Gwanak-ro, Gwanak-gu, Seoul, Republic of Korea
| | - Aram Ahn
- Department of Kinesiology, University of Connecticut, Storrs, CT 06269,USA
| | - Soo Hong Park
- Department of Physical Education, Seoul National University, Gwanak-ro, Gwanak-gu, Seoul, Republic of Korea.,Institute of Sport Science, Seoul National University, Gwanak-ro, Gwanak-gu, Seoul, Republic of Korea
| | - Seung Hee Kwak
- Department of Physical Education, Seoul National University, Gwanak-ro, Gwanak-gu, Seoul, Republic of Korea.,Institute of Sport Science, Seoul National University, Gwanak-ro, Gwanak-gu, Seoul, Republic of Korea
| | - Seong Eun Kwak
- Department of Physical Education, Seoul National University, Gwanak-ro, Gwanak-gu, Seoul, Republic of Korea.,School of Kinesiology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Wonsang Lee
- Department of Physical Education, Seoul National University, Gwanak-ro, Gwanak-gu, Seoul, Republic of Korea.,Institute of Sport Science, Seoul National University, Gwanak-ro, Gwanak-gu, Seoul, Republic of Korea
| | - Yong Ryoul Yang
- Aging Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Republic of Korea
| | - Minji Kim
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Republic of Korea.,Wide River Institute of Immunology, Seoul National University, Hongcheon, Republic of Korea
| | - Hyun Mu Shin
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Republic of Korea.,Wide River Institute of Immunology, Seoul National University, Hongcheon, Republic of Korea
| | - Hang-Rae Kim
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Republic of Korea.,BK21Plus Biomedical Science Project, Seoul National University College of Medicine, Seoul, Republic of Korea.,Medical Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea.,Wide River Institute of Immunology, Seoul National University, Hongcheon, Republic of Korea
| | - Hyo Youl Moon
- Department of Physical Education, Seoul National University, Gwanak-ro, Gwanak-gu, Seoul, Republic of Korea.,Institute of Sport Science, Seoul National University, Gwanak-ro, Gwanak-gu, Seoul, Republic of Korea.,Institute on Aging, Seoul National University, Gwanak-ro, Gwanak-gu, Seoul, Republic of Korea
| |
Collapse
|
24
|
Chen X, Yi Z, Wong GT, Hasan KMM, Kwan JS, Ma AC, Chang RC. Is exercise a senolytic medicine? A systematic review. Aging Cell 2021; 20:e13294. [PMID: 33378138 PMCID: PMC7811843 DOI: 10.1111/acel.13294] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 11/15/2020] [Accepted: 12/01/2020] [Indexed: 12/14/2022] Open
Abstract
Cellular senescence, a state of irreversible growth arrest triggered by various stressors, engages in a category of pathological processes, whereby senescent cells accumulate in mitotic tissues. Senolytics as novel medicine against aging and various diseases through the elimination of senescent cells has emerged rapidly in recent years. Exercise is a potent anti‐aging and anti‐chronic disease medicine, which has shown the capacity to lower the markers of cellular senescence over the past decade. However, whether exercise is a senolytic medicine for aging and various diseases remains unclear. Here, we have conducted a systematic review of the published literature studying the senolytic effects of exercise or physical activity on senescent cells under various states in both human and animal models. Exercise can reduce the markers of senescent cells in healthy humans, while it lowered the markers of senescent cells in obese but not healthy animals. The discrepancy between human and animal studies may be due to the relatively small volume of research and the variations in markers of senescent cells, types of cells/tissues, and health conditions. These findings suggest that exercise has senolytic properties under certain conditions, which warrant further investigations.
Collapse
Affiliation(s)
- Xiang‐Ke Chen
- Laboratory of Neurodegenerative Diseases School of Biomedical Sciences LKS Faculty of Medicine The University of Hong Kong Hong Kong China
| | - Zhen‐Ni Yi
- Department of Health Technology and Informatics Hong Kong Polytechnic University Hong Kong China
| | - Gordon Tin‐Chun Wong
- Department of Anaesthesiology LKS Faculty of Medicine The University of Hong Kong Hong Kong China
| | - Kazi Md. Mahmudul Hasan
- Department of Health Technology and Informatics Hong Kong Polytechnic University Hong Kong China
| | | | - Alvin Chun‐Hang Ma
- Department of Health Technology and Informatics Hong Kong Polytechnic University Hong Kong China
| | - Raymond Chuen‐Chung Chang
- Laboratory of Neurodegenerative Diseases School of Biomedical Sciences LKS Faculty of Medicine The University of Hong Kong Hong Kong China
- State Key Laboratory of Brain and Cognitive Sciences The University of Hong Kong Hong Kong China
| |
Collapse
|
25
|
Chen X, Yi Z, Wong GT, Hasan KMM, Kwan JS, Ma AC, Chang RC. Is exercise a senolytic medicine? A systematic review. Aging Cell 2020. [DOI: https://doi.org/10.1111/acel.13294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Affiliation(s)
- Xiang‐Ke Chen
- Laboratory of Neurodegenerative Diseases School of Biomedical Sciences LKS Faculty of Medicine The University of Hong Kong Hong Kong China
| | - Zhen‐Ni Yi
- Department of Health Technology and Informatics Hong Kong Polytechnic University Hong Kong China
| | - Gordon Tin‐Chun Wong
- Department of Anaesthesiology LKS Faculty of Medicine The University of Hong Kong Hong Kong China
| | - Kazi Md. Mahmudul Hasan
- Department of Health Technology and Informatics Hong Kong Polytechnic University Hong Kong China
| | | | - Alvin Chun‐Hang Ma
- Department of Health Technology and Informatics Hong Kong Polytechnic University Hong Kong China
| | - Raymond Chuen‐Chung Chang
- Laboratory of Neurodegenerative Diseases School of Biomedical Sciences LKS Faculty of Medicine The University of Hong Kong Hong Kong China
- State Key Laboratory of Brain and Cognitive Sciences The University of Hong Kong Hong Kong China
| |
Collapse
|
26
|
Yang Y, Liao Z, Xiao Q. Metformin ameliorates skeletal muscle atrophy in Grx1 KO mice by regulating intramuscular lipid accumulation and glucose utilization. Biochem Biophys Res Commun 2020; 533:1226-1232. [PMID: 33069361 DOI: 10.1016/j.bbrc.2020.09.119] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Accepted: 09/26/2020] [Indexed: 12/27/2022]
Abstract
Skeletal muscle is the largest tissue in the body, and plays a remarkable role in energy and metabolic homeostasis. Disorder in lipid metabolism and glucose utilization could impair the quality and function of skeletal muscle. Glutaredoxin-1 (Grx1) acts as a vital metabolic regulator of redox homeostasis. Recent studies have shown that Grx1 regulates hepatic lipid metabolism. The skeletal muscle also contains abundant Grx1, but the role of Grx1 in skeletal muscle remains unknown. Therefore, we investigated the effect of Grx1 on skeletal muscle. In this study, we found that Grx1-deficient mice (Grx1-/-) spontaneously developed muscle atrophy by 3 months of age. And the p-AMPK activity and Sirt1 activity were inhibited in Grx1-/- mice, which led to intramuscular lipid deposition and glucose utilization disorder in skeletal muscle. However, intraperitoneal injection of metformin for 15 consecutive days ameliorated skeletal muscle atrophy caused by Grx1 deficiency to a certain extent. Taken together, these findings indicate that Grx1 deficiency might induce skeletal muscle atrophy by regulating the intramuscular lipid deposition and glucose utilization, which could be attenuated by metformin. Therefore, the expression or activity of Grx1 may be a pharmacological approach to ameliorate muscle atrophy diseases, such as sarcopenia.
Collapse
Affiliation(s)
- Yunfei Yang
- Department of Geriatrics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Zhiyin Liao
- Department of Geriatrics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Qian Xiao
- Department of Geriatrics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China.
| |
Collapse
|
27
|
Effects of treadmill exercise on the regulation of tight junction proteins in aged mice. Exp Gerontol 2020; 141:111077. [PMID: 32898618 DOI: 10.1016/j.exger.2020.111077] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 08/11/2020] [Accepted: 08/26/2020] [Indexed: 02/06/2023]
Abstract
Tight junction protein is representative regulator of gut permeability. Also, it has been noted for controlling inflammatory responses through tight junction. Therefore, in this study, we examined that whether tight junction protein is changed in aged mice, and to further, confirmed the effect of treadmill exercise on the tight junction protein. In in vitro study, doxorubicin that induces cell senescence was treated to Caco2 cells (colon cell) to mimic aging effect. After that, 5-aminoimidazole-4-carboxamide-1-β-D-ribofuranoside (AICAR), exercise mimic chemical that stimulates AMPK level, was also administered to Caco2 cells. In animal study, 2 months and 21 months C57BL/6 J mouse were treated with treadmill exercise for 4 weeks (YE = 5, OE = 5). Then, the tight junction protein expression level was examined by western blot. Also, serum lipopolysaccharide (LPS) and zonulin level were analyzed to identify gut permeability. In vitro studies showed that doxorubicin downregulates tight junction protein expression levels in Caco2 cell, and also AICAR treatment upregulates tight junction protein expression levels. In animal study, 4 weeks treadmill exercise upregulated claudin-1 (p < 0.05) and occludin (p < 0.01) protein expression level in 21 months old mice. Also, zonula occluden-1 (ZO-1) protein expression level was not significant difference among all mice group. In addition, old mice group had higher level of serum LPS compared to young mice group, but the level was downregulated in both 2 months and 21 months mice group after four weeks of treadmill exercise. Zonulin, which is known as degrading tight junction protein, is not significantly changed by both age and exercise. This study compared that tight junction protein expression level in old mice compared to its level in young mice, and also clarified that the effect of treadmill exercise on tight junction protein in both young and old mice.
Collapse
|
28
|
Balan E, De Groote E, Bouillon M, Viceconte N, Mahieu M, Naslain D, Nielens H, Decottignies A, Deldicque L. No effect of the endurance training status on senescence despite reduced inflammation in skeletal muscle of older individuals. Am J Physiol Endocrinol Metab 2020; 319:E447-E454. [PMID: 32691630 DOI: 10.1152/ajpendo.00149.2020] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The aim of the present study was to determine if the training status decreases inflammation, slows down senescence, and preserves telomere health in skeletal muscle in older compared with younger subjects, with a specific focus on satellite cells. Analyses were conducted on skeletal muscle and cultured satellite cells from vastus lateralis biopsies (n = 34) of male volunteers divided into four groups: young sedentary (YS), young trained cyclists (YT), old sedentary (OS), and old trained cyclists (OT). The senescence state and inflammatory profile were evaluated by telomere dysfunction-induced foci (TIF) quantification, senescence-associated β-galactosidase (SA-β-Gal) staining, and quantitative (q)RT-PCR. Independently of the endurance training status, TIF levels (+35%, P < 0.001) and the percentage of SA-β-Gal-positive cells (+30%, P < 0.05) were higher in cultured satellite cells of older compared with younger subjects. p16 (4- to 5-fold) and p21 (2-fold) mRNA levels in skeletal muscle were higher with age but unchanged by the training status. Aging induced higher CD68 mRNA levels in human skeletal muscle (+102%, P = 0.009). Independently of age, both trained groups had lower IL-8 mRNA levels (-70%, P = 0.011) and tended to have lower TNF-α mRNA levels (-40%, P = 0.10) compared with the sedentary subjects. All together, we found that the endurance training status did not slow down senescence in skeletal muscle and satellite cells in older compared with younger subjects despite reduced inflammation in skeletal muscle. These findings highlight that the link between senescence and inflammation can be disrupted in skeletal muscle.
Collapse
Affiliation(s)
- Estelle Balan
- Institute of Neuroscience, Université Catholique de Louvain, Louvain-La-Neuve, Belgium
| | - Estelle De Groote
- Institute of Neuroscience, Université Catholique de Louvain, Louvain-La-Neuve, Belgium
| | - Margot Bouillon
- Institute of Neuroscience, Université Catholique de Louvain, Louvain-La-Neuve, Belgium
| | - Nikenza Viceconte
- De Duve Institute, Université Catholique de Louvain, Brussels, Belgium
| | - Manon Mahieu
- De Duve Institute, Université Catholique de Louvain, Brussels, Belgium
| | - Damien Naslain
- Institute of Neuroscience, Université Catholique de Louvain, Louvain-La-Neuve, Belgium
| | - Henri Nielens
- Institute of Neuroscience, Université Catholique de Louvain, Louvain-La-Neuve, Belgium
| | | | - Louise Deldicque
- Institute of Neuroscience, Université Catholique de Louvain, Louvain-La-Neuve, Belgium
| |
Collapse
|
29
|
The role of adipose tissue senescence in obesity- and ageing-related metabolic disorders. Clin Sci (Lond) 2020; 134:315-330. [PMID: 31998947 DOI: 10.1042/cs20190966] [Citation(s) in RCA: 84] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Revised: 01/07/2020] [Accepted: 01/14/2020] [Indexed: 12/19/2022]
Abstract
Adipose tissue as the largest energy reservoir and endocrine organ is essential for maintenance of systemic glucose, lipid and energy homeostasis, but these metabolic functions decline with ageing and obesity. Adipose tissue senescence is one of the common features in obesity and ageing. Although cellular senescence is a defensive mechanism preventing tumorigenesis, its occurrence in adipose tissue causatively induces defective adipogenesis, inflammation, aberrant adipocytokines production and insulin resistance, leading to adipose tissue dysfunction. In addition to these paracrine effects, adipose tissue senescence also triggers systemic inflammation and senescence as well as insulin resistance in the distal metabolic organs, resulting in Type 2 diabetes and other premature physiological declines. Multiple cell types including mature adipocytes, immune cells, endothelial cells and progenitor cells gradually senesce at different levels in different fat depots with ageing and obesity, highlighting the heterogeneity and complexity of adipose tissue senescence. In this review, we discuss the causes and consequences of adipose tissue senescence, and the major cell types responsible for adipose tissue senescence in ageing and obesity. In addition, we summarize the pharmacological approaches and lifestyle intervention targeting adipose tissue senescence for the treatment of obesity- and ageing-related metabolic diseases.
Collapse
|
30
|
Role of Non-coding RNA in Diabetic Cardiomyopathy. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1229:181-195. [PMID: 32285412 DOI: 10.1007/978-981-15-1671-9_10] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Diabetic cardiomyopathy (DCM) is the leading cause of morbidity and mortality in diabetic population worldwide, characteristic by cardiomyocyte hypertrophy, apoptosis and myocardial interstitial fibrosis and eventually developing into heart failure. Non-coding RNAs, such as microRNAs (miRNAs), circular RNAs (circRNAs), long non-coding RNAs (lncRNAs) and other RNAs without the protein encoding function were emerging as a popular regulator in various types of processes during human diseases. The evidences have shown that miRNAs are regulators in diabetic cardiomyopathy, such as insulin resistance, cardiomyocytes apoptosis, and inflammatory, especially their protective effect on heart function. Besides that, the functions of lncRNAs and circRNAs have been gradually confirmed in recent years, and their functions in DCM have become increasingly prominent. We highlighted the nonnegligible roles of non-coding RNAs in the pathological process of DCM and showed the future possibilities of these non-coding RNAs in DCM treatment. In this chapter, we summarized the present advance of the researches in this filed and raised the concern and the prospect in the future.
Collapse
|
31
|
Bolotta A, Filardo G, Abruzzo PM, Astolfi A, De Sanctis P, Di Martino A, Hofer C, Indio V, Kern H, Löfler S, Marcacci M, Zampieri S, Marini M, Zucchini C. Skeletal Muscle Gene Expression in Long-Term Endurance and Resistance Trained Elderly. Int J Mol Sci 2020; 21:ijms21113988. [PMID: 32498275 PMCID: PMC7312229 DOI: 10.3390/ijms21113988] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 05/23/2020] [Accepted: 05/28/2020] [Indexed: 02/06/2023] Open
Abstract
Physical exercise is deemed the most efficient way of counteracting the age-related decline of skeletal muscle. Here we report a transcriptional study by next-generation sequencing of vastus lateralis biopsies from elderly with a life-long high-level training practice (n = 9) and from age-matched sedentary subjects (n = 5). Unsupervised mixture distribution analysis was able to correctly categorize trained and untrained subjects, whereas it failed to discriminate between individuals who underwent a prevalent endurance (n = 5) or a prevalent resistance (n = 4) training, thus showing that the training mode was not relevant for sarcopenia prevention. KEGG analysis of transcripts showed that physical exercise affected a high number of metabolic and signaling pathways, in particular those related to energy handling and mitochondrial biogenesis, where AMPK and AKT-mTOR signaling pathways are both active and balance each other, concurring to the establishment of an insulin-sensitive phenotype and to the maintenance of a functional muscle mass. Other pathways affected by exercise training increased the efficiency of the proteostatic mechanisms, consolidated the cytoskeletal organization, lowered the inflammation level, and contrasted cellular senescence. This study on extraordinary individuals who trained at high level for at least thirty years suggests that aging processes and exercise training travel the same paths in the opposite direction.
Collapse
Affiliation(s)
- Alessandra Bolotta
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna School of Medicine, 40138 Bologna, Italy; (A.B.); (P.D.S.); (M.M.); (C.Z.)
- IRCCS Fondazione Don Carlo Gnocchi, 20148 Milan, Italy
| | - Giuseppe Filardo
- Applied and Translational Research Center, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy;
| | - Provvidenza Maria Abruzzo
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna School of Medicine, 40138 Bologna, Italy; (A.B.); (P.D.S.); (M.M.); (C.Z.)
- IRCCS Fondazione Don Carlo Gnocchi, 20148 Milan, Italy
- Correspondence: ; Tel.: +39-051-2094122
| | - Annalisa Astolfi
- Giorgio Prodi Interdepartimental Center for Cancer Research, S.Orsola-Malpighi Hospital, 40138 Bologna, Italy; (A.A.); (V.I.)
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, 44121 Ferrara, Italy
| | - Paola De Sanctis
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna School of Medicine, 40138 Bologna, Italy; (A.B.); (P.D.S.); (M.M.); (C.Z.)
| | - Alessandro Di Martino
- Second Orthopaedic and Traumatologic Clinic, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy;
| | - Christian Hofer
- Ludwig Boltzmann Institute for Rehabilitation Research, 1160 Wien, Austria; (C.H.); (H.K.); (S.L.)
| | - Valentina Indio
- Giorgio Prodi Interdepartimental Center for Cancer Research, S.Orsola-Malpighi Hospital, 40138 Bologna, Italy; (A.A.); (V.I.)
| | - Helmut Kern
- Ludwig Boltzmann Institute for Rehabilitation Research, 1160 Wien, Austria; (C.H.); (H.K.); (S.L.)
| | - Stefan Löfler
- Ludwig Boltzmann Institute for Rehabilitation Research, 1160 Wien, Austria; (C.H.); (H.K.); (S.L.)
| | - Maurilio Marcacci
- Department of Biomedical Sciences, Knee Joint Reconstruction Center, 3rd Orthopaedic Division, Humanitas Clinical Institute, Humanitas University, 20089 Milan, Italy;
| | - Sandra Zampieri
- Department of Surgery, Oncology and Gastroenterology, University of Padua, 35122 Padua, Italy;
- Department of Biomedical Sciences, University of Padua, 35131 Padua, Italy
| | - Marina Marini
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna School of Medicine, 40138 Bologna, Italy; (A.B.); (P.D.S.); (M.M.); (C.Z.)
- IRCCS Fondazione Don Carlo Gnocchi, 20148 Milan, Italy
| | - Cinzia Zucchini
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna School of Medicine, 40138 Bologna, Italy; (A.B.); (P.D.S.); (M.M.); (C.Z.)
| |
Collapse
|
32
|
Moon HY, Yoon KJ, Lee WS, Cho HS, Kim DY, Kim JS. Neural maturation enhanced by exercise-induced extracellular derivatives. Sci Rep 2020; 10:3893. [PMID: 32127592 PMCID: PMC7054262 DOI: 10.1038/s41598-020-60930-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Accepted: 02/19/2020] [Indexed: 12/31/2022] Open
Abstract
Physical activity has profound effects on neuronal progenitor cell growth, differentiation, and integration, but the mechanism for these effects is still ambiguous. Using a mouse model, we investigated the effects of two weeks of treadmill running on the dynamics of the size distribution and miRNA profiles of serum extracellular derivatives (EDs) using particle-sizing analysis and small RNA sequencing. We found that an increased average diameter of EDs in the running group compared with the sedentary group (p < 0.05), and 16 miRNAs were significantly altered (p < 0.05) in the running group. Furthermore, functional annotation analysis of differentially expressed miRNA-predicted target genes showed that many of these target genes are involved in the PI3K-Akt pathway. Exercise-induced serum EDs increased Neuro2A cell viability and Akt phosphorylation. We also found that expression levels of neuronal maturation markers such as Microtubule-Associated Protein 2 (MAP2ab) and Neuronal nuclei (NeuN) were increased (p < 0.05, respectively), and that inhibition of the PI3K-Akt pathway by LY294002 pre-treatment ameliorated their expression in Neuro2A cells. Finally, the administration of exercise-induced EDs for 3 days increased the Histone 3 phosphorylation and β-III tubulin expression in Ink/Arf null neural stem cells and progenitors (NSPCs) under each proliferation and differentiation condition. These results suggest that exercise-induced circulating EDs may mediate neuronal maturation during exercise.
Collapse
Affiliation(s)
- Hyo Youl Moon
- Department of Physical Education, Seoul National University, Seoul, Korea.,Institute of Sport Science, Seoul National University, Gwanak-ro, Gwanak-gu, Seoul, 08826, Republic of Korea.,School of Biological Sciences, Seoul National University, Seoul, 08826, Korea
| | - Kyeong Jin Yoon
- Department of Physical Education, Seoul National University, Seoul, Korea
| | - Won Sang Lee
- Department of Physical Education, Seoul National University, Seoul, Korea
| | - Hae-Sung Cho
- Department of Physical Education, Seoul National University, Seoul, Korea
| | - Do-Yeon Kim
- Department of Pharmacology, School of Dentistry, Brain Science and Engineering Institute, Kyungpook National University, Daegu, 41940, Republic of Korea
| | - Ji-Seok Kim
- Department of Physical Education, Gyeongsang National University, Jinju-daero, Jinju, 52828, Republic of Korea.
| |
Collapse
|
33
|
Baati N, Feillet-Coudray C, Fouret G, Vernus B, Goustard B, Jollet M, Bertrand-Gaday C, Coudray C, Lecomte J, Bonnieu A, Koechlin-Ramonatxo C. New evidence of exercise training benefits in myostatin-deficient mice: Effect on lipidomic abnormalities. Biochem Biophys Res Commun 2019; 516:89-95. [PMID: 31200956 DOI: 10.1016/j.bbrc.2019.06.014] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Accepted: 06/03/2019] [Indexed: 12/11/2022]
Abstract
Myostatin (Mstn) inactivation or inhibition is considered as a promising treatment for various muscle-wasting disorders because it promotes muscle growth. However, myostatin-deficient hypertrophic muscles show strong fatigability associated with abnormal mitochondria and lipid metabolism. Here, we investigated whether endurance training could improve lipid metabolism and mitochondrial membrane lipid composition in mice where the Mstn gene was genetically ablated (Mstn-/- mice). In Mstn-/- mice, 4 weeks of daily running exercise sessions (65-70% of the maximal aerobic speed for 1 h) improved significantly aerobic performance, particularly the endurance capacity (up to +280% compared with untrained Mstn-/- mice), to levels comparable to those of trained wild type (WT) littermates. The expression of oxidative and lipid metabolism markers also was increased, as indicated by the upregulation of the Cpt1, Ppar-δ and Fasn genes. Moreover, endurance training also increased, but far less than WT, citrate synthase level and mitochondrial protein content. Interestingly endurance training normalized the cardiolipin fraction in the mitochondrial membrane of Mstn-/- muscle compared with WT. These results suggest that the combination of myostatin inhibition and endurance training could increase the muscle mass while preserving the physical performance with specific effects on cardiolipin and lipid-related pathways.
Collapse
Affiliation(s)
- Narjes Baati
- INRA, UMR866 Dynamique Musculaire Et Métabolisme, Université Montpellier, 34000, Montpellier, France
| | - Christine Feillet-Coudray
- INRA, UMR866 Dynamique Musculaire Et Métabolisme, Université Montpellier, 34000, Montpellier, France
| | - Gilles Fouret
- INRA, UMR866 Dynamique Musculaire Et Métabolisme, Université Montpellier, 34000, Montpellier, France
| | - Barbara Vernus
- INRA, UMR866 Dynamique Musculaire Et Métabolisme, Université Montpellier, 34000, Montpellier, France
| | - Bénédicte Goustard
- INRA, UMR866 Dynamique Musculaire Et Métabolisme, Université Montpellier, 34000, Montpellier, France
| | - Maxence Jollet
- INRA, UMR866 Dynamique Musculaire Et Métabolisme, Université Montpellier, 34000, Montpellier, France
| | - Christelle Bertrand-Gaday
- INRA, UMR866 Dynamique Musculaire Et Métabolisme, Université Montpellier, 34000, Montpellier, France
| | - Charles Coudray
- INRA, UMR866 Dynamique Musculaire Et Métabolisme, Université Montpellier, 34000, Montpellier, France
| | - Jérôme Lecomte
- Centre de Recherche Agronomique pour le Développement)/SupAgro, UMR IATE, F-34398, Montpellier, France
| | - Anne Bonnieu
- INRA, UMR866 Dynamique Musculaire Et Métabolisme, Université Montpellier, 34000, Montpellier, France
| | | |
Collapse
|