1
|
Moradi N, Kuznyetsova A, Sanfrancesco VC, Champsi S, Hood DA. Focus on the Forgotten Organelle: Regulation of Lysosomes in Skeletal Muscle. Exerc Sport Sci Rev 2025; 53:87-95. [PMID: 39820215 DOI: 10.1249/jes.0000000000000358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2025]
Abstract
Research on the role of the lysosome as the terminal organelle in autophagy and in communicating with other organelles in skeletal muscle is in its infancy. We hypothesize that the lysosome can adapt positively to exercise to improve the clearance of cargo, like dysfunctional mitochondria, within muscle, representing an important therapy for protein homeostasis in aging and muscle disuse.
Collapse
Affiliation(s)
- Neushaw Moradi
- Muscle Health Research Centre, Kinesiology and Health Science, York University, Toronto, ON, Canada
| | | | | | | | | |
Collapse
|
2
|
Rahman FA, Graham MQ, Adam AM, Juracic ES, Tupling AR, Quadrilatero J. Mitophagy is required to protect against excessive skeletal muscle atrophy following hindlimb immobilization. J Biomed Sci 2025; 32:29. [PMID: 39979946 PMCID: PMC11844018 DOI: 10.1186/s12929-025-01118-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Accepted: 01/10/2025] [Indexed: 02/22/2025] Open
Abstract
BACKGROUND Skeletal muscle atrophy involves significant remodeling of fibers and is characterized by deficits in mitochondrial content and function. These changes are intimately connected to shifts in mitochondrial turnover, encompassing processes such as mitophagy and mitochondrial biogenesis. However, the role of these mitochondrial turnover processes in muscle atrophy remains poorly understood. METHODS We used a novel mitophagy reporter model, mt-Keima mice, to perform hindlimb immobilization and accurately measure mitophagy. A comprehensive set of analyses were conducted to investigate biochemical and molecular changes at the muscle and mitochondrial levels. We also performed image analyses to determine mitophagic flux. To further explore the role of mitophagy in immobilization-induced atrophy, we treated animals with N-acetylcysteine (NAC; 150 mg/kg/day) to modify reactive oxygen species (ROS) signaling and colchicine (0.4 mg/kg/day) to inhibit autophagy. RESULTS Our study revealed that hindlimb immobilization leads to muscle weakness and atrophy of fast-twitch muscle fibers (types IIA, IIX, and IIB), with recovery observed in IIA fibers following remobilization. This atrophy was accompanied by a significant increase in mitophagic flux. Additionally, immobilization induced notable mitochondrial dysfunction, as shown by diminished respiration, increased mitochondrial ROS, and greater whole muscle lipid peroxidation. Treatment of immobilized mice with NAC enhanced mitochondrial respiration and reduced ROS generation but suppressed mitophagic flux and intensified atrophy of type IIX and IIB fibers. Additionally, administration of colchicine to immobilized mice suppressed mitophagic flux, which also exacerbated atrophy of IIX and IIB fibers. Colchicine treatment led to significant reductions in mitochondrial function, accompanied by CASP9 and CASP3 activation. CONCLUSION These findings emphasize the role of mitophagy in limiting excessive muscle atrophy during immobilization. Targeting mitophagy may offer new strategies to preserve muscle function during prolonged periods of immobilization.
Collapse
Affiliation(s)
- Fasih A Rahman
- Faculty of Health, Department of Kinesiology and Health Sciences, University of Waterloo, 200 University Ave. West, Waterloo, ON, N2L 3G1, Canada
| | - Mackenzie Q Graham
- Faculty of Health, Department of Kinesiology and Health Sciences, University of Waterloo, 200 University Ave. West, Waterloo, ON, N2L 3G1, Canada
| | - Amanda M Adam
- Faculty of Health, Department of Kinesiology and Health Sciences, University of Waterloo, 200 University Ave. West, Waterloo, ON, N2L 3G1, Canada
| | - Emma S Juracic
- Faculty of Health, Department of Kinesiology and Health Sciences, University of Waterloo, 200 University Ave. West, Waterloo, ON, N2L 3G1, Canada
| | - A Russell Tupling
- Faculty of Health, Department of Kinesiology and Health Sciences, University of Waterloo, 200 University Ave. West, Waterloo, ON, N2L 3G1, Canada
| | - Joe Quadrilatero
- Faculty of Health, Department of Kinesiology and Health Sciences, University of Waterloo, 200 University Ave. West, Waterloo, ON, N2L 3G1, Canada.
| |
Collapse
|
3
|
Zhou XH, Luo YX, Yao XQ. Exercise-driven cellular autophagy: A bridge to systematic wellness. J Adv Res 2025:S2090-1232(24)00613-1. [PMID: 39756575 DOI: 10.1016/j.jare.2024.12.036] [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: 08/18/2024] [Revised: 11/28/2024] [Accepted: 12/21/2024] [Indexed: 01/07/2025] Open
Abstract
BACKGROUND Exercise enhances health by supporting homeostasis, bolstering defenses, and aiding disease recovery. It activates autophagy, a conserved cellular process essential for maintaining balance, while dysregulated autophagy contributes to disease progression. Despite extensive research on exercise and autophagy independently, their interplay remains insufficiently understood. AIM OF REVIEW This review explores the molecular mechanisms of exercise-induced autophagy in various tissues, focusing on key transduction pathways. It examines how different types of exercise trigger specific autophagic responses, supporting cellular balance and addressing systemic dysfunctions. The review also highlights the signaling pathways involved, their roles in protecting organ function, reducing disease risk, and promoting longevity, offering a clear understanding of the link between exercise and autophagy. KEY SCIENTIFIC CONCEPTS OF REVIEW Exercise-induced autophagy is governed by highly coordinated and dynamic pathways integrating direct and indirect mechanical forces and biochemical signals, linking physical activity to cellular and systemic health across multiple organ systems. Its activation is influenced by exercise modality, intensity, duration, and individual biological characteristics, including age, sex, and muscle fiber composition. Aerobic exercises primarily engage AMPK and mTOR pathways, supporting mitochondrial quality and cellular homeostasis. Anaerobic training activates PI3K/Akt signaling, modulating molecules like FOXO3a and Beclin1 to drive muscle autophagy and repair. In pathological contexts, exercise-induced autophagy enhances mitochondrial function, proteostasis, and tissue regeneration, benefiting conditions like sarcopenia, neurodegeneration, myocardial ischemia, metabolic disorders, and cancer. However, excessive exercise may lead to autophagic overactivation, leading to muscle atrophy or pathological cardiac remodeling. This underscores the critical need for balanced exercise regimens to maximize therapeutic efficacy while minimizing risks. Future research should prioritize identifying reliable biomarkers, optimizing exercise protocols, and integrating exercise with pharmacological strategies to enhance therapeutic outcomes.
Collapse
Affiliation(s)
- Xiao-Han Zhou
- Department of Rehabilitation, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, PR China
| | - Ya-Xi Luo
- Department of Rehabilitation, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, PR China
| | - Xiu-Qing Yao
- Department of Rehabilitation, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, PR China; Chongqing Municipality Clinical Research Center for Geriatric Medicine, Chongqing, PR China; Department of Rehabilitation Therapy, Chongqing Medical University, Chongqing, PR China.
| |
Collapse
|
4
|
Zhang X, Liao S, Huang L, Wang J. Prospective Intervention Strategies Between Skeletal Muscle Health and Mitochondrial Changes During Aging. Adv Biol (Weinh) 2025; 9:e2400235. [PMID: 39410835 DOI: 10.1002/adbi.202400235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Revised: 09/14/2024] [Indexed: 01/19/2025]
Abstract
Sarcopenia is a geriatric condition characterized by a decrease in skeletal muscle mass and function, significantly impacting both quality of life and overall health. Mitochondria are the main sites of energy production within the cell, and also produce reactive oxygen species (ROS), which maintain mitochondrial homeostasis-mitophagy (clearing damaged mitochondria); mitochondrial dynamics, which involve fusion and fission to regulate mitochondrial morphology; mitochondrial biogenesis, which ensures the functionality and homeostasis of mitochondria. Sarcopenia is linked to mitochondrial dysfunction, suggesting that muscle mitochondrial function therapy should be investigated. Extrinsic therapies are extensively examined to identify new treatments for muscular illnesses including sarcopenia. Changes in muscle physiology and lifestyle interventions, such as pharmacological treatments and exercise, can modulate mitochondrial activity in older adults. This PubMed review encompasses the most significant mitophagy and sarcopenia research from the past five years. Animal models, cellular models, and human samples are well covered. The review will inform the development of novel mitochondria-targeted therapies aimed at combating age-related muscle atrophy.
Collapse
Affiliation(s)
- Xin Zhang
- School of Basic Medical Sciences, Youjiang Medical University for Nationalities, Baise, 533000, China
| | - Suchan Liao
- School of Basic Medical Sciences, Youjiang Medical University for Nationalities, Baise, 533000, China
- Department of Physiology, School of Basic Medical Sciences, Youjiang Medical University for Nationalities, Baise, 533000, China
| | - Lingling Huang
- School of Basic Medical Sciences, Youjiang Medical University for Nationalities, Baise, 533000, China
| | - Jinhua Wang
- School of Basic Medical Sciences, Youjiang Medical University for Nationalities, Baise, 533000, China
- Modern Industrial College of Biomedicine and Great Health, Youjiang Medical University for Nationalities, Baise, Guangxi, 533000, China
| |
Collapse
|
5
|
Kim JT, Jeon DH, Lee HJ. Molecular mechanism of skeletal muscle loss and its prevention by natural resources. Food Sci Biotechnol 2024; 33:3387-3400. [PMID: 39493391 PMCID: PMC11525361 DOI: 10.1007/s10068-024-01678-x] [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: 05/15/2024] [Revised: 07/23/2024] [Accepted: 08/04/2024] [Indexed: 11/05/2024] Open
Abstract
A skeletal muscle disorder has drawn attention due to the global aging issues. The loss of skeletal muscle mass has been suggested to be from the reduced muscle regeneration by dysfunction of muscle satellite cell/fibro-adipogenic progenitor cells and the muscle atrophy by dysfunction of mitochondria, ubiquitin-proteasome system, and autophagy. In this review, we highlighted the underlying mechanisms of skeletal muscle mass loss including Notch signaling, Wnt/β-catenin signaling, Hedgehog signaling, AMP-activated protein kinase (AMPK) signaling, and mammalian target of rapamycin (mTOR) signaling. In addition, we summarized accumulated studies of natural resources investigating their roles in ameliorating the loss of skeletal muscle mass and demonstrating the underlying mechanisms in vitro and in vivo. In conclusion, following the studies of natural resources exerting the preventive activity in muscle mass loss, the signaling-based approaches may accelerate the development of functional foods for sarcopenia prevention.
Collapse
Affiliation(s)
- Jin Tae Kim
- Department of Food Science and Biotechnology, Chung-Ang University, Anseong, 17546 South Korea
- GreenTech-Based Food Safety Research Group, BK21 Four, Chung-Ang University, Anseong, 17546 South Korea
| | - Dong Hyeon Jeon
- Department of Food Science and Biotechnology, Chung-Ang University, Anseong, 17546 South Korea
- GreenTech-Based Food Safety Research Group, BK21 Four, Chung-Ang University, Anseong, 17546 South Korea
| | - Hong Jin Lee
- Department of Food Science and Biotechnology, Chung-Ang University, Anseong, 17546 South Korea
- GreenTech-Based Food Safety Research Group, BK21 Four, Chung-Ang University, Anseong, 17546 South Korea
| |
Collapse
|
6
|
Moradi N, Sanfrancesco VC, Champsi S, Hood DA. Regulation of lysosomes in skeletal muscle during exercise, disuse and aging. Free Radic Biol Med 2024; 225:323-332. [PMID: 39332541 DOI: 10.1016/j.freeradbiomed.2024.09.028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 08/29/2024] [Accepted: 09/16/2024] [Indexed: 09/29/2024]
Abstract
Lysosomes play a critical role as a terminal organelle in autophagy flux and in regulating protein degradation, but their function and adaptability in skeletal muscle is understudied. Lysosome functions include both housekeeping and signaling functions essential for cellular homeostasis. This review focuses on the regulation of lysosomes in skeletal muscle during exercise, disuse, and aging, with a consideration of sex differences as well as the role of lysosomes in mediating the degradation of mitochondria, termed mitophagy. Exercise enhances mitophagy during elevated mitochondrial stress and energy demand. A critical response to this deviation from homeostasis is the activation of transcription factors TFEB and TFE3, which drive the expression of lysosomal and autophagic genes. Conversely, during muscle disuse, the suppression of lysosomal activity contributes to the accumulation of defective mitochondria and other cellular debris, impairing muscle function. Aging further exacerbates these effects by diminishing lysosomal efficacy, leading to the accumulation of damaged cellular components. mTORC1, a key nutrient sensor, modulates lysosomal activity by inhibiting TFEB/TFE3 translocation to the nucleus under nutrient-rich conditions, thereby suppressing autophagy. During nutrient deprivation or exercise, AMPK activation inhibits mTORC1, facilitating TFEB/TFE3 nuclear translocation and promoting lysosomal biogenesis and autophagy. TRPML1 activation by mitochondrial ROS enhances lysosomal calcium release, which is essential for autophagy and maintaining mitochondrial quality. Overall, the intricate regulation of lysosomal functions and signaling pathways in skeletal muscle is crucial for adaptation to physiological demands, and disruptions in these processes during disuse and aging underscore the ubiquitous power of exercise-induced adaptations, and also highlight the potential for targeted therapeutic interventions to preserve muscle health.
Collapse
Affiliation(s)
- N Moradi
- Muscle Health Research Centre, Kinesiology and Health Science, York University, Toronto, ON, Canada
| | - V C Sanfrancesco
- Muscle Health Research Centre, Kinesiology and Health Science, York University, Toronto, ON, Canada
| | - S Champsi
- Muscle Health Research Centre, Kinesiology and Health Science, York University, Toronto, ON, Canada
| | - D A Hood
- Muscle Health Research Centre, Kinesiology and Health Science, York University, Toronto, ON, Canada.
| |
Collapse
|
7
|
He Y, Fan Y, Ahmadpoor X, Wang Y, Li ZA, Zhu W, Lin H. Targeting lysosomal quality control as a therapeutic strategy against aging and diseases. Med Res Rev 2024; 44:2472-2509. [PMID: 38711187 DOI: 10.1002/med.22047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2023] [Revised: 04/04/2024] [Accepted: 04/21/2024] [Indexed: 05/08/2024]
Abstract
Previously, lysosomes were primarily referred to as the digestive organelles and recycling centers within cells. Recent discoveries have expanded the lysosomal functional scope and revealed their critical roles in nutrient sensing, epigenetic regulation, plasma membrane repair, lipid transport, ion homeostasis, and cellular stress response. Lysosomal dysfunction is also found to be associated with aging and several diseases. Therefore, function of macroautophagy, a lysosome-dependent intracellular degradation system, has been identified as one of the updated twelve hallmarks of aging. In this review, we begin by introducing the concept of lysosomal quality control (LQC), which is a cellular machinery that maintains the number, morphology, and function of lysosomes through different processes such as lysosomal biogenesis, reformation, fission, fusion, turnover, lysophagy, exocytosis, and membrane permeabilization and repair. Next, we summarize the results from studies reporting the association between LQC dysregulation and aging/various disorders. Subsequently, we explore the emerging therapeutic strategies that target distinct aspects of LQC for treating diseases and combatting aging. Lastly, we underscore the existing knowledge gap and propose potential avenues for future research.
Collapse
Affiliation(s)
- Yuchen He
- Department of Orthopaedics, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
- Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Yishu Fan
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Xenab Ahmadpoor
- Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Yumin Wang
- Department of Otolaryngology Head and Neck Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Zhong Alan Li
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Shatin, NT, Hong Kong SAR, China
- Center for Neuromusculoskeletal Restorative Medicine, Hong Kong Science Park, Shatin, NT, Hong Kong SAR, China
| | - Weihong Zhu
- Department of Orthopaedics, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
- Department of Orthopaedic Surgery, Mayo Clinic, Rochester, Minnesota, USA
| | - Hang Lin
- Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- Department of Bioengineering, University of Pittsburgh Swanson School of Engineering, Pittsburgh, Pennsylvania, USA
- McGowan Institute for Regenerative Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
8
|
Roths M, Rudolph TE, Krishna S, Michael A, Selsby JT. One day of environment-induced heat stress damages the murine myocardium. Am J Physiol Heart Circ Physiol 2024; 327:H978-H988. [PMID: 39212770 PMCID: PMC11482254 DOI: 10.1152/ajpheart.00180.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 08/19/2024] [Accepted: 08/19/2024] [Indexed: 09/04/2024]
Abstract
The physiological consequences of environment-induced heat stress (EIHS), caused by prolonged exposure to excess heat and humidity, are largely unknown. The purpose of this investigation was to determine the extent to which EIHS alters cardiac health. We hypothesized that 24 h of EIHS would cause cardiac injury and cellular dysfunction in a murine EIHS model. To test this hypothesis, 7-wk-old female mice were housed under thermoneutral (TN) conditions (n = 12; 31.2 ± 1.01°C, 35 ± 0.7% humidity) or EIHS conditions (n = 14; 37.6 ± 0.01°C, 42.0 ± 0.06% humidity) for 24 h. Environment-induced heat stress increased rectal temperature by 2.1°C (P < 0.01) and increased subcutaneous temperature by 1.8°C (P < 0.01). Body weight was decreased by 10% (P = 0.03), heart weight/body weight was increased by 26% (P < 0.01), and tissue water content was increased by 11% (P < 0.05) in EIHS compared with TN. In comparison with TN, EIHS increased protein abundance of heat shock protein (HSP) 27 by 84% (P = 0.01); however, HSPs 90, 60, 70, and phosphorylated HSP 27 were similar between groups. Histological inspection of the heart revealed that EIHS animals had increased myocyte vacuolation in the left ventricle (P = 0.01), right ventricle (P < 0.01), and septum (P = 0.01) compared with TN animals. Biochemical indices are suggestive of mitochondrial remodeling, increased autophagic flux, and robust activation of endoplasmic reticulum stress in hearts from EIHS mice compared with TN mice. These data demonstrate that 1 day of EIHS is sufficient to induce myocardial injury and biochemical dysregulation.NEW & NOTEWORTHY The consequences of prolonged environment-induced heat stress (EIHS) on heart health are largely unknown. We discovered that a 24-h exposure to environmental conditions sufficient to cause EIHS resulted in cardiac edema and histopathologic changes in the right and left ventricles. Furthermore, among other biochemical changes, EIHS increased autophagic flux and caused endoplasmic reticulum stress. These data raise the possibility that thermic injury, even when insufficient to cause heat stroke, can damage the myocardium.
Collapse
Affiliation(s)
- Melissa Roths
- Department of Animal Science, Iowa State University, Ames, Iowa, United States
| | - Tori E Rudolph
- Department of Animal Science, Iowa State University, Ames, Iowa, United States
| | - Swathy Krishna
- Department of Animal Science, Iowa State University, Ames, Iowa, United States
| | - Alyona Michael
- Veterinary Diagnostic Laboratory, Iowa State University College of Veterinary Medicine, Ames, Iowa, United States
| | - Joshua T Selsby
- Department of Animal Science, Iowa State University, Ames, Iowa, United States
| |
Collapse
|
9
|
Jeong I, Cho EJ, Yook JS, Choi Y, Park DH, Kang JH, Lee SH, Seo DY, Jung SJ, Kwak HB. Mitochondrial Adaptations in Aging Skeletal Muscle: Implications for Resistance Exercise Training to Treat Sarcopenia. Life (Basel) 2024; 14:962. [PMID: 39202704 PMCID: PMC11355854 DOI: 10.3390/life14080962] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Revised: 07/13/2024] [Accepted: 07/29/2024] [Indexed: 09/03/2024] Open
Abstract
Sarcopenia, the age-related decline in muscle mass and function, poses a significant health challenge as the global population ages. Mitochondrial dysfunction is a key factor in sarcopenia, as evidenced by the role of mitochondrial reactive oxygen species (mtROS) in mitochondrial biogenesis and dynamics, as well as mitophagy. Resistance exercise training (RET) is a well-established intervention for sarcopenia; however, its effects on the mitochondria in aging skeletal muscles remain unclear. This review aims to elucidate the relationship between mitochondrial dynamics and sarcopenia, with a specific focus on the implications of RET. Although aerobic exercise training (AET) has traditionally been viewed as more effective for mitochondrial enhancement, emerging evidence suggests that RET may also confer beneficial effects. Here, we highlight the potential of RET to modulate mtROS, drive mitochondrial biogenesis, optimize mitochondrial dynamics, and promote mitophagy in aging skeletal muscles. Understanding this interplay offers insights for combating sarcopenia and preserving skeletal muscle health in aging individuals.
Collapse
Affiliation(s)
- Ilyoung Jeong
- Program in Biomedical Science & Engineering, Department of Biomedical Science, Inha University, Incheon 22212, Republic of Korea; (I.J.); (E.-J.C.); (D.-H.P.); (J.-H.K.)
| | - Eun-Jeong Cho
- Program in Biomedical Science & Engineering, Department of Biomedical Science, Inha University, Incheon 22212, Republic of Korea; (I.J.); (E.-J.C.); (D.-H.P.); (J.-H.K.)
| | - Jang-Soo Yook
- Institute of Sports and Arts Convergence, Inha University, Incheon 22212, Republic of Korea; (J.-S.Y.); (Y.C.)
| | - Youngju Choi
- Institute of Sports and Arts Convergence, Inha University, Incheon 22212, Republic of Korea; (J.-S.Y.); (Y.C.)
- Institute of Specialized Teaching and Research, Inha University, Incheon 22212, Republic of Korea
| | - Dong-Ho Park
- Program in Biomedical Science & Engineering, Department of Biomedical Science, Inha University, Incheon 22212, Republic of Korea; (I.J.); (E.-J.C.); (D.-H.P.); (J.-H.K.)
- Institute of Sports and Arts Convergence, Inha University, Incheon 22212, Republic of Korea; (J.-S.Y.); (Y.C.)
- Department of Kinesiology, Inha University, Incheon 22212, Republic of Korea
| | - Ju-Hee Kang
- Program in Biomedical Science & Engineering, Department of Biomedical Science, Inha University, Incheon 22212, Republic of Korea; (I.J.); (E.-J.C.); (D.-H.P.); (J.-H.K.)
- Institute of Sports and Arts Convergence, Inha University, Incheon 22212, Republic of Korea; (J.-S.Y.); (Y.C.)
- Department of Pharmacology, College of Medicine, Inha University, Incheon 22212, Republic of Korea
| | - Seok-Hun Lee
- Combat Institute of Australia, Leederville, WA 6007, Australia;
| | - Dae-Yun Seo
- Basic Research Laboratory, Department of Physiology, College of Medicine, Smart Marine Therapeutic Center, Cardiovascular and Metabolic Disease Core Research Support Center, Inje University, Busan 47392, Republic of Korea
| | - Su-Jeen Jung
- Department of Leisure Sports, Seoil University, Seoul 02192, Republic of Korea
| | - Hyo-Bum Kwak
- Program in Biomedical Science & Engineering, Department of Biomedical Science, Inha University, Incheon 22212, Republic of Korea; (I.J.); (E.-J.C.); (D.-H.P.); (J.-H.K.)
- Institute of Sports and Arts Convergence, Inha University, Incheon 22212, Republic of Korea; (J.-S.Y.); (Y.C.)
- Department of Kinesiology, Inha University, Incheon 22212, Republic of Korea
| |
Collapse
|
10
|
Stouth DW, vanLieshout TL, Mikhail AI, Ng SY, Raziee R, Edgett BA, Vasam G, Webb EK, Gilotra KS, Markou M, Pineda HC, Bettencourt-Mora BG, Noor H, Moll Z, Bittner ME, Gurd BJ, Menzies KJ, Ljubicic V. CARM1 drives mitophagy and autophagy flux during fasting-induced skeletal muscle atrophy. Autophagy 2024; 20:1247-1269. [PMID: 38018843 PMCID: PMC11210918 DOI: 10.1080/15548627.2023.2288528] [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: 01/17/2023] [Revised: 11/17/2023] [Accepted: 11/20/2023] [Indexed: 11/30/2023] Open
Abstract
CARM1 (coactivator associated arginine methyltransferase 1) has recently emerged as a powerful regulator of skeletal muscle biology. However, the molecular mechanisms by which the methyltransferase remodels muscle remain to be fully understood. In this study, carm1 skeletal muscle-specific knockout (mKO) mice exhibited lower muscle mass with dysregulated macroautophagic/autophagic and atrophic signaling, including depressed AMP-activated protein kinase (AMPK) site-specific phosphorylation of ULK1 (unc-51 like autophagy activating kinase 1; Ser555) and FOXO3 (forkhead box O3; Ser588), as well as MTOR (mechanistic target of rapamycin kinase)-induced inhibition of ULK1 (Ser757), along with AKT/protein kinase B site-specific suppression of FOXO1 (Ser256) and FOXO3 (Ser253). In addition to lower mitophagy and autophagy flux in skeletal muscle, carm1 mKO led to increased mitochondrial PRKN/parkin accumulation, which suggests that CARM1 is required for basal mitochondrial turnover and autophagic clearance. carm1 deletion also elicited PPARGC1A (PPARG coactivator 1 alpha) activity and a slower, more oxidative muscle phenotype. As such, these carm1 mKO-evoked adaptations disrupted mitophagy and autophagy induction during food deprivation and collectively served to mitigate fasting-induced muscle atrophy. Furthermore, at the threshold of muscle atrophy during food deprivation experiments in humans, skeletal muscle CARM1 activity decreased similarly to our observations in mice, and was accompanied by site-specific activation of ULK1 (Ser757), highlighting the translational impact of the methyltransferase in human skeletal muscle. Taken together, our results indicate that CARM1 governs mitophagic, autophagic, and atrophic processes fundamental to the maintenance and remodeling of muscle mass. Targeting the enzyme may provide new therapeutic approaches for mitigating skeletal muscle atrophy.Abbreviation: ADMA: asymmetric dimethylarginine; AKT/protein kinase B: AKT serine/threonine kinase; AMPK: AMP-activated protein kinase; ATG: autophagy related; BECN1: beclin 1; BNIP3: BCL2 interacting protein 3; CARM1: coactivator associated arginine methyltransferase 1; Col: colchicine; CSA: cross-sectional area; CTNS: cystinosin, lysosomal cystine transporter; EDL: extensor digitorum longus; FBXO32/MAFbx: F-box protein 32; FOXO: forkhead box O; GAST: gastrocnemius; H2O2: hydrogen peroxide; IMF: intermyofibrillar; LAMP1: lysosomal associated membrane protein 1; MAP1LC3B: microtubule associated protein 1 light chain 3 beta; mKO: skeletal muscle-specific knockout; MMA: monomethylarginine; MTOR: mechanistic target of rapamycin kinase; MYH: myosin heavy chain; NFE2L2/NRF2: NFE2 like bZIP transcription factor 2; OXPHOS: oxidative phosphorylation; PABPC1/PABP1: poly(A) binding protein cytoplasmic 1; PPARGC1A/PGC-1α: PPARG coactivator 1 alpha; PRKN/parkin: parkin RBR E3 ubiquitin protein ligase; PRMT: protein arginine methyltransferase; Sal: saline; SDMA: symmetric dimethylarginine; SIRT1: sirtuin 1; SKP2: S-phase kinase associated protein 2; SMARCC1/BAF155: SWI/SNF related, matrix associated, actin dependent regulator of chromatin subfamily c member 1; SOL: soleus; SQSTM1/p62: sequestosome 1; SS: subsarcolemmal; TA: tibialis anterior; TFAM: transcription factor A, mitochondrial; TFEB: transcription factor EB; TOMM20: translocase of outer mitochondrial membrane 20; TRIM63/MuRF1: tripartite motif containing 63; ULK1: unc-51 like autophagy activating kinase 1; VPS11: VPS11 core subunit of CORVET and HOPS complexes; WT: wild-type.
Collapse
Affiliation(s)
- Derek W. Stouth
- Department of Kinesiology, McMaster University, Hamilton, Ontario, Canada
| | | | - Andrew I. Mikhail
- Department of Kinesiology, McMaster University, Hamilton, Ontario, Canada
| | - Sean Y. Ng
- Department of Kinesiology, McMaster University, Hamilton, Ontario, Canada
| | - Rozhin Raziee
- Department of Kinesiology, McMaster University, Hamilton, Ontario, Canada
| | - Brittany A. Edgett
- School of Kinesiology and Health Studies, Queen’s University, Kingston, Ontario, Canada
| | - Goutham Vasam
- Interdisciplinary School of Health Sciences, Faculty of Health Sciences, University of Ottawa, Ottawa, Ontario, Canada
| | - Erin K. Webb
- Department of Kinesiology, McMaster University, Hamilton, Ontario, Canada
| | - Kevin S. Gilotra
- Department of Kinesiology, McMaster University, Hamilton, Ontario, Canada
| | - Matthew Markou
- Department of Kinesiology, McMaster University, Hamilton, Ontario, Canada
| | - Hannah C. Pineda
- Department of Kinesiology, McMaster University, Hamilton, Ontario, Canada
| | | | - Haleema Noor
- Department of Kinesiology, McMaster University, Hamilton, Ontario, Canada
| | - Zachary Moll
- Department of Kinesiology, McMaster University, Hamilton, Ontario, Canada
| | - Megan E. Bittner
- Department of Kinesiology, McMaster University, Hamilton, Ontario, Canada
| | - Brendon J. Gurd
- School of Kinesiology and Health Studies, Queen’s University, Kingston, Ontario, Canada
| | - Keir J. Menzies
- Interdisciplinary School of Health Sciences, Faculty of Health Sciences, University of Ottawa, Ottawa, Ontario, Canada
- Ottawa Institute of Systems Biology and the Centre for Neuromuscular Disease, Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Vladimir Ljubicic
- Department of Kinesiology, McMaster University, Hamilton, Ontario, Canada
| |
Collapse
|
11
|
Bhatt V, Shukla H, Tiwari AK. Parkinson's Disease and Mitotherapy-Based Approaches towards α-Synucleinopathies. J Integr Neurosci 2024; 23:109. [PMID: 38940084 DOI: 10.31083/j.jin2306109] [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: 11/28/2023] [Revised: 01/25/2024] [Accepted: 02/05/2024] [Indexed: 06/29/2024] Open
Abstract
Parkinson's disease (PD) is a neurodegenerative disorder characterized by the progressive loss of dopaminergic neurons in the substantia nigra pars compacta region of the midbrain and the formation of intracellular protein aggregates known as Lewy bodies, of which a major component is the protein α-synuclein. Several studies have suggested that mitochondria play a central role in the pathogenesis of PD, encompassing both familial and sporadic forms of the disease. Mitochondrial dysfunction is attributed to bioenergetic impairment, increased oxidative stress, damage to mitochondrial DNA, and alteration in mitochondrial morphology. These alterations may contribute to improper functioning of the central nervous system and ultimately lead to neurodegeneration. The perturbation of mitochondrial function makes it a potential target, worthy of exploration for neuroprotective therapies and to improve mitochondrial health in PD. Thus, in the current review, we provide an update on mitochondria-based therapeutic approaches toward α-synucleinopathies in PD.
Collapse
Affiliation(s)
- Vidhi Bhatt
- Genetics & Developmental Biology Laboratory, Department of Biotechnology & Bioengineering, Institute of Advanced Research (IAR), 382426 Gandhinagar, Gujarat, India
| | - Halak Shukla
- Genetics & Developmental Biology Laboratory, Department of Biotechnology & Bioengineering, Institute of Advanced Research (IAR), 382426 Gandhinagar, Gujarat, India
| | - Anand Krishna Tiwari
- Genetics & Developmental Biology Laboratory, Department of Biotechnology & Bioengineering, Institute of Advanced Research (IAR), 382426 Gandhinagar, Gujarat, India
| |
Collapse
|
12
|
Du ZY, Zhu HL, Chang W, Zhang YF, Ling Q, Wang KW, Zhang J, Zhang QB, Kan XL, Wang QN, Wang H, Zhou Y. Maternal prednisone exposure during pregnancy elevates susceptibility to osteoporosis in female offspring: The role of mitophagy/FNDC5 alteration in skeletal muscle. JOURNAL OF HAZARDOUS MATERIALS 2024; 469:133997. [PMID: 38508115 DOI: 10.1016/j.jhazmat.2024.133997] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Revised: 03/01/2024] [Accepted: 03/08/2024] [Indexed: 03/22/2024]
Abstract
Maternal exposure to glucocorticoids has been associated with adverse outcomes in offspring. However, the consequences and mechanisms of gestational exposure to prednisone on susceptibility to osteoporosis in the offspring remain unclear. Here, we found that gestational prednisone exposure enhanced susceptibility to osteoporosis in adult mouse offspring. In a further exploration of myogenic mechanisms, results showed that gestational prednisone exposure down-regulated FNDC5/irisin protein expression and activation of OPTN-dependent mitophagy in skeletal muscle of adult offspring. Additional experiments elucidated that activated mitophagy significantly inhibited the expression of FNDC5/irisin in skeletal muscle cells. Likewise, we observed delayed fetal bone development, downregulated FNDC5/irisin expression, and activated mitophagy in fetal skeletal muscle upon gestational prednisone exposure. In addition, an elevated total m6A level was observed in fetal skeletal muscle after gestational prednisone exposure. Finally, gestational supplementation with S-adenosylhomocysteine (SAH), an inhibitor of m6A activity, attenuated mitophagy and restored FNDC5/irisin expression in fetal skeletal muscle, which in turn reversed fetal bone development. Overall, these data indicate that gestational prednisone exposure increases m6A modification, activates mitophagy, and decreases FNDC5/irisin expression in skeletal muscle, thus elevating osteoporosis susceptibility in adult offspring. Our results provide a new perspective on the earlier prevention and treatment of fetal-derived osteoporosis.
Collapse
Affiliation(s)
- Zun-Yu Du
- Department of Rehabilitation Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Hefei, China
| | - Hua-Long Zhu
- Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Hefei, China; Department of Toxicology, School of Public Health, Anhui Medical University, Hefei, China
| | - Wei Chang
- Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Hefei, China; Department of Toxicology, School of Public Health, Anhui Medical University, Hefei, China
| | - Yu-Feng Zhang
- Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Hefei, China; Department of Toxicology, School of Public Health, Anhui Medical University, Hefei, China; Teaching and Research Section of Nuclear Medicine, School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China
| | - Qing Ling
- Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Hefei, China; Department of Toxicology, School of Public Health, Anhui Medical University, Hefei, China
| | - Kai-Wen Wang
- Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Hefei, China; Department of Toxicology, School of Public Health, Anhui Medical University, Hefei, China
| | - Jin Zhang
- Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Hefei, China; Department of Toxicology, School of Public Health, Anhui Medical University, Hefei, China
| | - Quan-Bing Zhang
- Department of Rehabilitation Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Xiu-Li Kan
- Department of Rehabilitation Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Qu-Nan Wang
- Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Hefei, China; Department of Toxicology, School of Public Health, Anhui Medical University, Hefei, China
| | - Hua Wang
- Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Hefei, China; Department of Toxicology, School of Public Health, Anhui Medical University, Hefei, China.
| | - Yun Zhou
- Department of Rehabilitation Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, China.
| |
Collapse
|
13
|
Noh SG, Ahn A, Davi SM, Lepley LK, Kwon OS. Quadriceps muscle atrophy after non-invasive anterior cruciate ligament injury: evidence linking to autophagy and mitophagy. Front Physiol 2024; 15:1341723. [PMID: 38496299 PMCID: PMC10940348 DOI: 10.3389/fphys.2024.1341723] [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: 11/20/2023] [Accepted: 02/20/2024] [Indexed: 03/19/2024] Open
Abstract
Introduction: Anterior cruciate ligament (ACL) injury is frequently accompanied by quadriceps muscle atrophy, a process closely linked to mitochondrial health and mitochondria-specific autophagy. However, the temporal progression of key quadricep atrophy-mediating events following ACL injury remains poorly understood. To advance our understanding, we conducted a longitudinal study to elucidate key parameters in quadriceps autophagy and mitophagy. Methods: Long-Evans rats were euthanized at 7, 14, 28, and 56 days after non-invasive ACL injury that was induced via tibial compression overload; controls were not injured. Vastus lateralis muscle was extracted, and subsequent immunoblotting analysis was conducted using primary antibodies targeting key proteins involved in autophagy and mitophagy cellular processes. Results: Our findings demonstrated dynamic changes in autophagy and mitophagy markers in the quadriceps muscle during the recovery period after ACL injury. The early response to the injury was characterized by the induction of autophagy at 14 days (Beclin1), indicating an initial cellular response to the injury. Subsequently, at 14 days we observed increase in the elongation of autophagosomes (Atg4B), suggesting a potential remodeling process. The autophagosome flux was also augmented between 14- and 28 days (LC3-II/LC3-I ratio and p62). Notably, at 56 days, markers associated with the elimination of damaged mitochondria were elevated (PINK1, Parkin, and VDAC1), indicating a possible ongoing cellular repair and restoration process. Conclusion: These data highlight the complexity of muscle recovery after ACL injury and underscore the overlooked but crucial role of autophagy and mitophagy in promoting the recovery process.
Collapse
Affiliation(s)
- Sung Gi Noh
- Department of Kinesiology, University of Connecticut, Storrs, CT, United States
| | - Ahram Ahn
- Department of Kinesiology, University of Connecticut, Storrs, CT, United States
| | - Steven M. Davi
- Department of Kinesiology, University of Connecticut, Storrs, CT, United States
- Cooperative Studies Program Coordinating Center (CSPCC), VA Connecticut Healthcare System, West Haven, CT, United States
| | - Lindsey K. Lepley
- School of Kinesiology, University of Michigan, Ann Arbor, MI, United States
| | - Oh Sung Kwon
- Department of Kinesiology, University of Connecticut, Storrs, CT, United States
- Department of Orthopaedic Surgery and Center on Aging, University of Connecticut School of Medicine, Farmington, CT, United States
| |
Collapse
|
14
|
Guglielmi V, Cheli M, Tonin P, Vattemi G. Sporadic Inclusion Body Myositis at the Crossroads between Muscle Degeneration, Inflammation, and Aging. Int J Mol Sci 2024; 25:2742. [PMID: 38473988 PMCID: PMC10932328 DOI: 10.3390/ijms25052742] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Revised: 02/19/2024] [Accepted: 02/22/2024] [Indexed: 03/14/2024] Open
Abstract
Sporadic inclusion body myositis (sIBM) is the most common muscle disease of older people and is clinically characterized by slowly progressive asymmetrical muscle weakness, predominantly affecting the quadriceps, deep finger flexors, and foot extensors. At present, there are no enduring treatments for this relentless disease that eventually leads to severe disability and wheelchair dependency. Although sIBM is considered a rare muscle disorder, its prevalence is certainly higher as the disease is often undiagnosed or misdiagnosed. The histopathological phenotype of sIBM muscle biopsy includes muscle fiber degeneration and endomysial lymphocytic infiltrates that mainly consist of cytotoxic CD8+ T cells surrounding nonnecrotic muscle fibers expressing MHCI. Muscle fiber degeneration is characterized by vacuolization and the accumulation of congophilic misfolded multi-protein aggregates, mainly in their non-vacuolated cytoplasm. Many players have been identified in sIBM pathogenesis, including environmental factors, autoimmunity, abnormalities of protein transcription and processing, the accumulation of several toxic proteins, the impairment of autophagy and the ubiquitin-proteasome system, oxidative and nitrative stress, endoplasmic reticulum stress, myonuclear degeneration, and mitochondrial dysfunction. Aging has also been proposed as a contributor to the disease. However, the interplay between these processes and the primary event that leads to the coexistence of autoimmune and degenerative changes is still under debate. Here, we outline our current understanding of disease pathogenesis, focusing on degenerative mechanisms, and discuss the possible involvement of aging.
Collapse
Affiliation(s)
- Valeria Guglielmi
- Cellular and Molecular Biology of Cancer Program, NCI-Designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA;
- Immunity and Pathogenesis Program, Infectious and Inflammatory Disease Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | - Marta Cheli
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, 37134 Verona, Italy; (M.C.); (P.T.)
| | - Paola Tonin
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, 37134 Verona, Italy; (M.C.); (P.T.)
| | - Gaetano Vattemi
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, 37134 Verona, Italy; (M.C.); (P.T.)
| |
Collapse
|
15
|
Wong JC, Oliveira AN, Khemraj P, Hood DA. The role of TFE3 in mediating skeletal muscle mitochondrial adaptations to exercise training. J Appl Physiol (1985) 2024; 136:262-273. [PMID: 38095014 DOI: 10.1152/japplphysiol.00484.2023] [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: 07/17/2023] [Revised: 12/07/2023] [Accepted: 12/11/2023] [Indexed: 01/26/2024] Open
Abstract
Transcription factor E3 (TFE3) is a transcription factor that activates the expression of lysosomal genes involved in the clearance of dysfunctional mitochondria, termed mitophagy. With exercise, TFE3 is presumed to optimize the mitochondrial pool through the removal of organelles via lysosomes. However, the molecular mechanisms of the involved pathways remain unknown. Wild-type (WT) and TFE3 knockout (KO) mice were subjected to 6 wk of voluntary wheel running as an endurance training regimen. This was followed by a 45-min bout of in situ stimulation of the sciatic nerve innervating hindlimb muscles to evaluate muscle fatigue and contractile properties. A subset of animals was treated with colchicine to measure autophagy and mitophagy flux. Fatigability during stimulation was reduced with training in WT animals, as seen by a 13% increase in the percentage of maximum force at 5 min of stimulation, and a 30% increase at 30 minutes. Permeabilized fiber oxygen consumption was also improved with training. Concurrent with improved muscle and mitochondrial function, cytochrome c oxidase (COX) activity and COX I protein expression were increased in trained WT animals compared to untrained animals, signifying an increase in mitochondrial content. These training adaptations were abolished with the loss of TFE3. Surprisingly, the absence of TFE3 did not affect lysosomal content nor did it blunt the induction of mitophagy flux with contractile activity compared to WT mice. Our results suggest that the loss of TFE3 compromises beneficial training adaptations that lead to improved muscle endurance and mitochondrial function.NEW & NOTEWORTHY Our understanding of the role of transcription factor E3 (TFE3) in skeletal muscle is very limited. This research shows that TFE3 plays a direct role in skeletal muscle mitochondrial enhancement with exercise training, thereby introducing a paradigm shift in our perception of the function of TFE3 in mitochondrial maintenance, beyond mitophagy. This research serves to introduce TFE3 as a protein that holds promise as a future therapeutic target for metabolic diseases and skeletal muscle dysfunction.
Collapse
Affiliation(s)
- Jenna C Wong
- Muscle Health Research Centre, School of Kinesiology and Health Science, York University, Toronto, Ontario, Canada
| | - Ashley N Oliveira
- Muscle Health Research Centre, School of Kinesiology and Health Science, York University, Toronto, Ontario, Canada
| | - Priyanka Khemraj
- Muscle Health Research Centre, School of Kinesiology and Health Science, York University, Toronto, Ontario, Canada
| | - David A Hood
- Muscle Health Research Centre, School of Kinesiology and Health Science, York University, Toronto, Ontario, Canada
| |
Collapse
|
16
|
Nevi L, Pöllänen N, Penna F, Caretti G. Targeting Epigenetic Regulators with HDAC and BET Inhibitors to Modulate Muscle Wasting. Int J Mol Sci 2023; 24:16404. [PMID: 38003594 PMCID: PMC10671811 DOI: 10.3390/ijms242216404] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Revised: 11/07/2023] [Accepted: 11/13/2023] [Indexed: 11/26/2023] Open
Abstract
Epigenetic changes contribute to the profound alteration in the transcriptional program associated with the onset and progression of muscle wasting in several pathological conditions. Although HDACs and their inhibitors have been extensively studied in the field of muscular dystrophies, the potential of epigenetic inhibitors has only been marginally explored in other disorders associated with muscle atrophy, such as in cancer cachexia and sarcopenia. BET inhibitors represent a novel class of recently developed epigenetic drugs that display beneficial effects in a variety of diseases beyond malignancies. Based on the preliminary in vitro and preclinical data, HDACs and BET proteins contribute to the pathogenesis of cancer cachexia and sarcopenia, modulating processes related to skeletal muscle mass maintenance and/or metabolism. Thus, epigenetic drugs targeting HDACs and BET proteins may emerge as promising strategies to reverse the catabolic phenotype associated with cachexia and sarcopenia. Further preclinical studies are warranted to delve deeper into the molecular mechanisms associated with the functions of HDACs and BET proteins in muscle atrophy and to establish whether their epigenetic inhibitors represent a prospective therapeutic avenue to alleviate muscle wasting.
Collapse
Affiliation(s)
- Lorenzo Nevi
- Department of Biosciences, University of Milan, 20133 Milan, Italy;
| | - Noora Pöllänen
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, 00014 Helsinki, Finland
| | - Fabio Penna
- Department of Clinical and Biological Sciences, University of Torino, 10125 Torino, Italy
| | | |
Collapse
|
17
|
Picca A, Guerra F, Calvani R, Romano R, Coelho-Junior HJ, Bucci C, Leeuwenburgh C, Marzetti E. Mitochondrial-derived vesicles in skeletal muscle remodeling and adaptation. Semin Cell Dev Biol 2023; 143:37-45. [PMID: 35367122 DOI: 10.1016/j.semcdb.2022.03.023] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 02/25/2022] [Accepted: 03/19/2022] [Indexed: 12/24/2022]
Abstract
Mitochondrial remodeling is crucial to meet the bioenergetic demand to support muscle contractile activity during daily tasks and muscle regeneration following injury. A set of mitochondrial quality control (MQC) processes, including mitochondrial biogenesis, dynamics, and mitophagy, are in place to maintain a well-functioning mitochondrial network and support muscle regeneration. Alterations in any of these pathways compromises mitochondrial quality and may potentially lead to impaired myogenesis, defective muscle regeneration, and ultimately loss of muscle function. Among MQC processes, mitophagy has gained special attention for its implication in the clearance of dysfunctional mitochondria via crosstalk with the endo-lysosomal system, a major cell degradative route. Along this pathway, additional opportunities for mitochondrial disposal have been identified that may also signal at the systemic level. This communication occurs via inclusion of mitochondrial components within membranous shuttles named mitochondrial-derived vesicles (MDVs). Here, we discuss MDV generation and release as a mitophagy-complementing route for the maintenance of mitochondrial homeostasis in skeletal myocytes. We also illustrate the possible role of muscle-derived MDVs in immune signaling during muscle remodeling and adaptation.
Collapse
Affiliation(s)
- Anna Picca
- Fondazione Policlinico Universitario "Agostino Gemelli" IRCCS, Rome, Italy
| | - Flora Guerra
- Department of Biological and Environmental Sciences and Technologies, Università del Salento, Lecce, Italy
| | - Riccardo Calvani
- Fondazione Policlinico Universitario "Agostino Gemelli" IRCCS, Rome, Italy
| | - Roberta Romano
- Department of Biological and Environmental Sciences and Technologies, Università del Salento, Lecce, Italy
| | | | - Cecilia Bucci
- Department of Biological and Environmental Sciences and Technologies, Università del Salento, Lecce, Italy
| | - Christiaan Leeuwenburgh
- Department of Aging and Geriatric Research, Institute on Aging, Division of Biology of Aging, University of Florida, Gainesville, USA
| | - Emanuele Marzetti
- Fondazione Policlinico Universitario "Agostino Gemelli" IRCCS, Rome, Italy; Università Cattolica del Sacro Cuore, Department of Geriatrics and Orthopedics, Rome, Italy.
| |
Collapse
|
18
|
F AR, Quadrilatero J. Emerging role of mitophagy in myoblast differentiation and skeletal muscle remodeling. Semin Cell Dev Biol 2023; 143:54-65. [PMID: 34924331 DOI: 10.1016/j.semcdb.2021.11.026] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2021] [Revised: 11/26/2021] [Accepted: 11/30/2021] [Indexed: 12/17/2022]
Abstract
Mitochondrial turnover in the form of mitophagy is emerging as a central process in maintaining cellular function. The degradation of damaged mitochondria through mitophagy is particularly important in cells/tissues that exhibit high energy demands. Skeletal muscle is one such tissue that requires precise turnover of mitochondria in several conditions in order to optimize energy production and prevent bioenergetic crisis. For instance, the formation of skeletal muscle (i.e., myogenesis) is accompanied by robust turnover of low-functioning mitochondria to eventually allow the formation of high-functioning mitochondria. In mature skeletal muscle, alterations in mitophagy-related signaling occur during exercise, aging, and various disease states. Nonetheless, several questions regarding the direct role of mitophagy in various skeletal muscle conditions remain unknown. Furthermore, given the heterogenous nature of skeletal muscle with respect to various cellular and molecular properties, and the plasticity in these properties in various conditions, the involvement and characterization of mitophagy requires more careful consideration in this tissue. Therefore, this review will highlight the known mechanisms of mitophagy in skeletal muscle, and discuss their involvement during myogenesis and various skeletal muscle conditions. This review also provides important considerations for the accurate measurement of mitophagy and interpretation of data in skeletal muscle.
Collapse
Affiliation(s)
- Ahmad Rahman F
- Department of Kinesiology & Health Sciences, University of Waterloo, Waterloo, ON, Canada
| | - Joe Quadrilatero
- Department of Kinesiology & Health Sciences, University of Waterloo, Waterloo, ON, Canada.
| |
Collapse
|
19
|
Picca A, Lozanoska-Ochser B, Calvani R, Coelho-Júnior HJ, Leewenburgh C, Marzetti E. Inflammatory, mitochondrial, and senescence-related markers: Underlying biological pathways of muscle aging and new therapeutic targets. Exp Gerontol 2023; 178:112204. [PMID: 37169101 DOI: 10.1016/j.exger.2023.112204] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 05/04/2023] [Accepted: 05/09/2023] [Indexed: 05/13/2023]
Abstract
The maintenance of functional health is pivotal for achieving independent life in older age. The aged muscle is characterized by ultrastructural changes, including loss of type I and type II myofibers and a greater proportion of cytochrome c oxidase deficient and succinate dehydrogenase positive fibers. Both intrinsic (e.g., altered proteostasis, DNA damage, and mitochondrial dysfunction) and extrinsic factors (e.g., denervation, altered metabolic regulation, declines in satellite cells, and inflammation) contribute to muscle aging. Being a hub for several cellular activities, mitochondria are key to myocyte viability and mitochondrial dysfunction has been implicated in age-associated physical decline. The maintenance of functional organelles via mitochondrial quality control (MQC) processes is, therefore, crucial to skeletal myofiber viability and organismal health. The autophagy-lysosome pathway has emerged as a critical step of MQC in muscle by disposing organelles and proteins via their tagging for autophagosome incorporation and delivery to the lysosome for clearance. This pathway was found to be altered in muscle of physically inactive older adults. A relationship between this pathway and muscle tissue composition of the lower extremities as well as physical performance was also identified. Therefore, integrating muscle structure and myocyte quality control measures in the evaluation of muscle health may be a promising strategy for devising interventions fostering muscle health.
Collapse
Affiliation(s)
- Anna Picca
- Department of Medicine and Surgery, LUM University, Casamassima, 70100 Bari, Italy; Fondazione Policlinico Universitario "Agostino Gemelli" IRCSS, 00168 Rome, Italy
| | - Biliana Lozanoska-Ochser
- Department of Medicine and Surgery, LUM University, Casamassima, 70100 Bari, Italy; DAHFMO Unit of Histology and Medical Embryology, Sapienza University of Rome, 00161 Rome, Italy
| | - Riccardo Calvani
- Fondazione Policlinico Universitario "Agostino Gemelli" IRCSS, 00168 Rome, Italy; Department of Geriatrics and Orthopedics, Università Cattolica del Sacro Cuore, 00168 Rome, Italy.
| | - Hélio José Coelho-Júnior
- Department of Geriatrics and Orthopedics, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | | | - Emanuele Marzetti
- Fondazione Policlinico Universitario "Agostino Gemelli" IRCSS, 00168 Rome, Italy; Department of Geriatrics and Orthopedics, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| |
Collapse
|
20
|
Galasso L, Cappella A, Mulè A, Castelli L, Ciorciari A, Stacchiotti A, Montaruli A. Polyamines and Physical Activity in Musculoskeletal Diseases: A Potential Therapeutic Challenge. Int J Mol Sci 2023; 24:9798. [PMID: 37372945 DOI: 10.3390/ijms24129798] [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: 05/10/2023] [Revised: 06/02/2023] [Accepted: 06/04/2023] [Indexed: 06/29/2023] Open
Abstract
Autophagy dysregulation is commonplace in the pathogenesis of several invalidating diseases, such as musculoskeletal diseases. Polyamines, as spermidine and spermine, are small aliphatic cations essential for cell growth and differentiation, with multiple antioxidant, anti-inflammatory, and anti-apoptotic effects. Remarkably, they are emerging as natural autophagy regulators with strong anti-aging effects. Polyamine levels were significantly altered in the skeletal muscles of aged animals. Therefore, supplementation of spermine and spermidine may be important to prevent or treat muscle atrophy. Recent in vitro and in vivo experimental studies indicate that spermidine reverses dysfunctional autophagy and stimulates mitophagy in muscles and heart, preventing senescence. Physical exercise, as polyamines, regulates skeletal muscle mass inducing proper autophagy and mitophagy. This narrative review focuses on the latest evidence regarding the efficacy of polyamines and exercise as autophagy inducers, alone or coupled, in alleviating sarcopenia and aging-dependent musculoskeletal diseases. A comprehensive description of overall autophagic steps in muscle, polyamine metabolic pathways, and effects of the role of autophagy inducers played by both polyamines and exercise has been presented. Although literature shows few data in regard to this controversial topic, interesting effects on muscle atrophy in murine models have emerged when the two "autophagy-inducers" were combined. We hope these findings, with caution, can encourage researchers to continue investigating in this direction. In particular, if these novel insights could be confirmed in further in vivo and clinical studies, and the two synergic treatments could be optimized in terms of dose and duration, then polyamine supplementation and physical exercise might have a clinical potential in sarcopenia, and more importantly, implications for a healthy lifestyle in the elderly population.
Collapse
Affiliation(s)
- Letizia Galasso
- Department of Biomedical Sciences for Health, University of Milan, 20133 Milan, Italy
| | - Annalisa Cappella
- Department of Biomedical Sciences for Health, University of Milan, 20133 Milan, Italy
- U.O. Laboratorio di Morfologia Umana Applicata, I.R.C.C.S. Policlinico San Donato, San Donato Milanese, 20097 Milan, Italy
| | - Antonino Mulè
- Department of Biomedical Sciences for Health, University of Milan, 20133 Milan, Italy
| | - Lucia Castelli
- Department of Biomedical Sciences for Health, University of Milan, 20133 Milan, Italy
| | - Andrea Ciorciari
- Department of Biomedical Sciences for Health, University of Milan, 20133 Milan, Italy
| | - Alessandra Stacchiotti
- Department of Biomedical Sciences for Health, University of Milan, 20133 Milan, Italy
- U.O. Laboratorio di Morfologia Umana Applicata, I.R.C.C.S. Policlinico San Donato, San Donato Milanese, 20097 Milan, Italy
| | - Angela Montaruli
- Department of Biomedical Sciences for Health, University of Milan, 20133 Milan, Italy
- I.R.C.C.S. Ospedale Galeazzi-Sant'Ambrogio, 20157 Milan, Italy
| |
Collapse
|
21
|
Saud Gany SL, Chin KY, Tan JK, Aminuddin A, Makpol S. Curcumin as a Therapeutic Agent for Sarcopenia. Nutrients 2023; 15:nu15112526. [PMID: 37299489 DOI: 10.3390/nu15112526] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 05/24/2023] [Accepted: 05/25/2023] [Indexed: 06/12/2023] Open
Abstract
Sarcopenia is the progressive loss of muscle mass, strength, and functions as we age. The pathogenesis of sarcopenia is underlined by oxidative stress and inflammation. As such, it is reasonable to suggest that a natural compound with both antioxidant and anti-inflammatory activities could prevent sarcopenia. Curcumin, a natural compound derived from turmeric with both properties, could benefit muscle health. This review aims to summarise the therapeutic effects of curcumin on cellular, animal, and human studies. The available evidence found in the literature showed that curcumin prevents muscle degeneration by upregulating the expression of genes related to protein synthesis and suppressing genes related to muscle degradation. It also protects muscle health by maintaining satellite cell number and function, protecting the mitochondrial function of muscle cells, and suppressing inflammation and oxidative stress. However, it is noted that most studies are preclinical. Evidence from randomised control trials in humans is lacking. In conclusion, curcumin has the potential to be utilised to manage muscle wasting and injury, pending more evidence from carefully planned human clinical trials.
Collapse
Affiliation(s)
- Siti Liyana Saud Gany
- Department of Biochemistry, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia
| | - Kok-Yong Chin
- Department of Pharmacology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia
| | - Jen Kit Tan
- Department of Biochemistry, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia
| | - Amilia Aminuddin
- Department of Physiology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia
| | - Suzana Makpol
- Department of Biochemistry, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia
| |
Collapse
|
22
|
A cross-talk between sestrins, chronic inflammation and cellular senescence governs the development of age-associated sarcopenia and obesity. Ageing Res Rev 2023; 86:101852. [PMID: 36642190 DOI: 10.1016/j.arr.2023.101852] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 12/20/2022] [Accepted: 01/10/2023] [Indexed: 01/15/2023]
Abstract
The rapid increase in both the lifespan and proportion of older adults is accompanied by the unprecedented rise in age-associated chronic diseases, including sarcopenia and obesity. Aging is also manifested by increased susceptibility to multiple endogenous and exogenous stresses enabling such chronic conditions to develop. Among the main physiological regulators of cellular adaption to various stress stimuli, such as DNA damage, hypoxia, and oxidative stress, are sestrins (Sesns), a family of three evolutionarily conserved proteins, Sesn1, 2, and 3. Age-associated sarcopenia and obesity are characterized by two key processes: (i) accumulation of senescent cells in the skeletal muscle and adipose tissue and (ii) creation of a systemic, chronic, low-grade inflammation (SCLGI). Presumably, failed SCLGI resolution governs the development of these chronic conditions. Noteworthy, Sesns activate senolytics, which are agents that selectively eliminate senescent cells, as well as specialized pro-resolving mediators, which are factors that physiologically provide inflammation resolution. Sesns reveal clear beneficial effects in pre-clinical models of sarcopenia and obesity. Based on these observations, we propose a novel treatment strategy for age-associated sarcopenia and obesity, complementary to the conventional therapeutic modalities: Sesn activation, SCLGI resolution, and senescent cell elimination.
Collapse
|
23
|
Wu J, Ding P, Wu H, Yang P, Guo H, Tian Y, Meng L, Zhao Q. Sarcopenia: Molecular regulatory network for loss of muscle mass and function. Front Nutr 2023; 10:1037200. [PMID: 36819699 PMCID: PMC9932270 DOI: 10.3389/fnut.2023.1037200] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Accepted: 01/16/2023] [Indexed: 02/05/2023] Open
Abstract
Skeletal muscle is the foundation of human function and plays a key role in producing exercise, bone protection, and energy metabolism. Sarcopenia is a systemic disease, which is characterized by degenerative changes in skeletal muscle mass, strength, and function. Therefore, sarcopenia often causes weakness, prolonged hospitalization, falls and other adverse consequences that reduce the quality of life, and even lead to death. In recent years, sarcopenia has become the focus of in-depth research. Researchers have suggested some molecular mechanisms for sarcopenia according to different muscle physiology. These mechanisms cover neuromuscular junction lesion, imbalance of protein synthesis and breakdown, satellite cells dysfunction, etc. We summarize the latest research progress on the molecular mechanism of sarcopenia in this review in order to provide new ideas for future researchers to find valuable therapeutic targets and develop relevant prevention strategies.
Collapse
Affiliation(s)
- Jiaxiang Wu
- The Third Department of Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China,Hebei Key Laboratory of Precision Diagnosis and Comprehensive Treatment of Gastric Cancer, Shijiazhuang, China
| | - Ping’an Ding
- The Third Department of Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China,Hebei Key Laboratory of Precision Diagnosis and Comprehensive Treatment of Gastric Cancer, Shijiazhuang, China
| | - Haotian Wu
- The Third Department of Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China,Hebei Key Laboratory of Precision Diagnosis and Comprehensive Treatment of Gastric Cancer, Shijiazhuang, China
| | - Peigang Yang
- The Third Department of Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China,Hebei Key Laboratory of Precision Diagnosis and Comprehensive Treatment of Gastric Cancer, Shijiazhuang, China
| | - Honghai Guo
- The Third Department of Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China,Hebei Key Laboratory of Precision Diagnosis and Comprehensive Treatment of Gastric Cancer, Shijiazhuang, China
| | - Yuan Tian
- The Third Department of Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China,Hebei Key Laboratory of Precision Diagnosis and Comprehensive Treatment of Gastric Cancer, Shijiazhuang, China
| | - Lingjiao Meng
- Hebei Key Laboratory of Precision Diagnosis and Comprehensive Treatment of Gastric Cancer, Shijiazhuang, China,Research Center of the Fourth Hospital of Hebei Medical University, Shijiazhuang, China,Lingjiao Meng,
| | - Qun Zhao
- The Third Department of Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China,Hebei Key Laboratory of Precision Diagnosis and Comprehensive Treatment of Gastric Cancer, Shijiazhuang, China,*Correspondence: Qun Zhao,
| |
Collapse
|
24
|
Liang J, Zhang H, Zeng Z, Lv J, Huang J, Wu X, Wang M, Xu J, Fan J, Chen N. MicroRNA profiling of different exercise interventions for alleviating skeletal muscle atrophy in naturally aging rats. J Cachexia Sarcopenia Muscle 2023; 14:356-368. [PMID: 36457259 PMCID: PMC9891923 DOI: 10.1002/jcsm.13137] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 10/06/2022] [Accepted: 11/03/2022] [Indexed: 12/04/2022] Open
Abstract
BACKGROUND Exercise is an affordable and practical strategy to alleviate several detrimental outcomes from the aging process, including sarcopenia. The elucidation of molecular mechanisms to alleviate sarcopenia is one of the most important steps towards understanding human aging. Although microRNAs (miRNAs) regulate muscle growth, regeneration and aging, the potential role of exercise-mediated miRNAs during the prevention and rehabilitation of skeletal muscle atrophy upon exercise interventions remains unclear. METHODS A miRNA profile by miRNA sequencing for gastrocnemius muscle of a 24-month-old aged male rat model mimicking the naturally aging process was established through screening the differentially expressed miRNAs (DEMs) for alleviating aging-induced skeletal muscle atrophy upon optimal exercise intervention. The screened miRNAs and hub genes, as well as biomarkers with the most significantly enriched pathways, were validated by quantitative real-time polymerase chain reaction and western blotting. RESULTS The sarcopenia index (SI) value and cross-sectional area (CSA) of rats from the old control (OC) group significantly decreased when compared with the youth control (YC) group (P < 0.001, P < 0.01), whereas an increased SI value and an enlarged CSA of rats from the old-aerobic exercise (OE), old-resistance exercise (OR) and old-mixed exercise (OM) groups were determined (P < 0.01, P < 0.001, P < 0.05; P < 0.01, P < 0.01, P < 0.05). Our results demonstrate that 764 known miRNAs, 201 novel miRNAs and 505 miRNA-mRNA interaction networks were identified to be related to aging-induced muscular atrophy. Among them, 13 miRNAs were differentially expressed (P < 0.05 and log2 |fold change| > 1) between the YC group and the OC group. Compared with the OC group, 7, 2 and 11 miRNAs were differentially expressed in the OE, OR and OM groups after exercise interventions, respectively. Meanwhile, Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses revealed that the identified DEMs were primarily related to apoptosis, autophagy and the NF-κB/MuRF1 signalling pathways (P < 0.05). Meanwhile, four DEMs (miR-7a-1-3p, miR-135a-5p, miR-151-5p and miR-196b-5p), six hub genes (Ar, Igf1, Hif1a, Bdnf, Fak and Nras) and several biomarkers (LC3, Beclin1, p62, Bax, Bcl-2 and NF-κB/MuRF1) with the most significantly enriched pathways were confirmed, which may play a key role in muscular atrophy during the aging process. CONCLUSIONS These findings are closely correlated with the progression of sarcopenia and could act as potential biomarkers for the diagnosis and interventional monitoring of aging-induced skeletal muscle atrophy.
Collapse
Affiliation(s)
- Jiling Liang
- Tianjiu Research and Development Center for Exercise Nutrition and Foods, Hubei Key Laboratory of Exercise Training and Monitoring, College of Health Science, Wuhan Sports University, Wuhan, Hubei, China
| | - Hu Zhang
- Tianjiu Research and Development Center for Exercise Nutrition and Foods, Hubei Key Laboratory of Exercise Training and Monitoring, College of Health Science, Wuhan Sports University, Wuhan, Hubei, China
| | - Zhengzhong Zeng
- Tianjiu Research and Development Center for Exercise Nutrition and Foods, Hubei Key Laboratory of Exercise Training and Monitoring, College of Health Science, Wuhan Sports University, Wuhan, Hubei, China
| | - Jun Lv
- Tianjiu Research and Development Center for Exercise Nutrition and Foods, Hubei Key Laboratory of Exercise Training and Monitoring, College of Health Science, Wuhan Sports University, Wuhan, Hubei, China
| | - Jielun Huang
- Tianjiu Research and Development Center for Exercise Nutrition and Foods, Hubei Key Laboratory of Exercise Training and Monitoring, College of Health Science, Wuhan Sports University, Wuhan, Hubei, China
| | - Xiaowen Wu
- Tianjiu Research and Development Center for Exercise Nutrition and Foods, Hubei Key Laboratory of Exercise Training and Monitoring, College of Health Science, Wuhan Sports University, Wuhan, Hubei, China
| | - Minghui Wang
- Tianjiu Research and Development Center for Exercise Nutrition and Foods, Hubei Key Laboratory of Exercise Training and Monitoring, College of Health Science, Wuhan Sports University, Wuhan, Hubei, China
| | - Jiahao Xu
- Tianjiu Research and Development Center for Exercise Nutrition and Foods, Hubei Key Laboratory of Exercise Training and Monitoring, College of Health Science, Wuhan Sports University, Wuhan, Hubei, China
| | - Jingjing Fan
- Tianjiu Research and Development Center for Exercise Nutrition and Foods, Hubei Key Laboratory of Exercise Training and Monitoring, College of Health Science, Wuhan Sports University, Wuhan, Hubei, China
| | - Ning Chen
- Tianjiu Research and Development Center for Exercise Nutrition and Foods, Hubei Key Laboratory of Exercise Training and Monitoring, College of Health Science, Wuhan Sports University, Wuhan, Hubei, China
| |
Collapse
|
25
|
Christensen NM, Ringholm S, Buch BT, Gudiksen A, Halling JF, Pilegaard H. Muscle PGC-1α modulates hepatic mitophagy regulation during aging. Exp Gerontol 2023; 172:112046. [PMID: 36521568 DOI: 10.1016/j.exger.2022.112046] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 11/10/2022] [Accepted: 12/05/2022] [Indexed: 12/15/2022]
Abstract
Aging has been suggested to be associated with changes in oxidative capacity, autophagy, and mitophagy in the liver, but a simultaneous evaluation of these key cellular processes is lacking. Moreover, skeletal muscle transcriptional coactivator peroxisome proliferator-activated receptor gamma coactivator (PGC)-1α has been reported to mediate inter-organ signaling through myokines with regulatory effects in the liver, but the potential role of muscle PGC-1α on hepatic changes with age remains to be resolved. The aim of the present study was therefore to investigate 1) the effect of aging on mitochondrial autophagy and mitophagy capacity in mouse liver and 2) whether muscle PGC-1α is required for maintaining autophagy and mitophagy capacity in the liver during aging. The liver was obtained from young (Young) and aged (Aged) inducible muscle-specific PGC-1α knockout (iMKO) and floxed littermate control mice (Lox). Aging increased liver p62, Parkin and BCL2/adenovirus E1B 19 kDa protein-interacting protein (BNIP)3 protein with no effect of muscle specific PGC-1α knockout, while liver Microtubule-associated protein 1A/1B-light chain 3(LC3) II/I was unchanged with age, but tended to be lower in iMKO mice than in controls. Markers of liver mitochondrial oxidative capacity and oxidative stress were unchanged with age and iMKO. However, Parkin protein levels in isolated liver mitochondria were 2-fold higher in Aged iMKO mice than in Aged controls. In conclusion, aging had no effect on oxidative capacity and lipid peroxidation in the liver. However, aging was associated with increased levels of autophagy and mitophagy markers. Moreover, muscle PGC-1α appears to regulate hepatic mitochondrial translocation of Parkin in aged mice, suggesting that the metabolic capacity of skeletal muscle can modulate mitophagy regulation in the liver during aging.
Collapse
Affiliation(s)
- Natascha Masselkhi Christensen
- Section for Cell Biology and Physiology, Department of Biology, University of Copenhagen, Universitetsparken 13, 2100 Copenhagen, Denmark
| | - Stine Ringholm
- Section for Cell Biology and Physiology, Department of Biology, University of Copenhagen, Universitetsparken 13, 2100 Copenhagen, Denmark
| | - Bjørg Thiellesen Buch
- Section for Cell Biology and Physiology, Department of Biology, University of Copenhagen, Universitetsparken 13, 2100 Copenhagen, Denmark
| | - Anders Gudiksen
- Section for Cell Biology and Physiology, Department of Biology, University of Copenhagen, Universitetsparken 13, 2100 Copenhagen, Denmark
| | - Jens Frey Halling
- Section for Cell Biology and Physiology, Department of Biology, University of Copenhagen, Universitetsparken 13, 2100 Copenhagen, Denmark
| | - Henriette Pilegaard
- Section for Cell Biology and Physiology, Department of Biology, University of Copenhagen, Universitetsparken 13, 2100 Copenhagen, Denmark.
| |
Collapse
|
26
|
Jin J, Yang Z, Liu H, Guo M, Chen B, Zhu H, Wang Y, Lin J, Wang S, Chen S. Effects of acupuncture on the miR-146a-mediated IRAK1/TRAF6/NF-κB signaling pathway in rats with sarcopenia induced by D-galactose. ANNALS OF TRANSLATIONAL MEDICINE 2023; 11:47. [PMID: 36819511 PMCID: PMC9929824 DOI: 10.21037/atm-22-6082] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Accepted: 01/03/2023] [Indexed: 01/15/2023]
Abstract
Background Sarcopenia during aging is closely linked to sterile, low-grade, chronic inflammation. However, considering the increasingly aging global population, the effectiveness of existing treatments for sarcopenia is not exact, and acupuncture, as an effective anti-inflammatory therapy, has the potential to treat it. Methods Fifty Sprague-Dawley rats were randomly allocated into five groups, including Control group, D-galactose (D-gal) group, D-gal + acupuncture (DA) group, D-gal + non-acupoint (DN) group and D-gal amino acid mixture (DAA) group. An aging rat was model constructed using D-gal for 12 weeks. Rats in the control group received 0.9% physiological saline daily. Treatment groups were acupunctured or given amino acid mixture interventions daily, and lasted for last 4 consecutive weeks. The effects of acupuncture were evaluated by the hematoxylin and eosin staining (H&E), transmission electron microscopic (TEM) examination and terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) assays. The anti-inflammatory mechanism of acupuncture was studied by using the expressions of microRNA-146a (miR-146a) mediated nuclear factor-kappa B (NF-κB) signaling pathway-related proteins were detected by immunofluorescence, western blotting, quantitative real-time polymerase chain reaction (PCR) and enzyme-linked immunosorbent assay (ELISA). Results Rats injected by D-galactose (D-gal) revealed apparent skeletal muscle atrophy with significantly reduced cross-sectional area and fiber diameter. In contrast, acupuncture treatment alleviated these hallmarks of skeletal muscle atrophy and mitigated the mitochondrial aberrations and skeletal muscle apoptosis in D-gal rats. In addition, acupuncture also downgraded the overexpression of inflammatory factors in skeletal muscle, influenced miR-146a and the target genes level, and inhibited NF-κB nuclear translation in D-gal rats. Conclusions Acupuncture may ameliorate skeletal muscle atrophy, and its effects may be associated with the control of mitochondrial function regulation and the suppression of inflammation.
Collapse
Affiliation(s)
- Jing Jin
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Zhengyu Yang
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Haichao Liu
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Mingling Guo
- The School of Health, Fujian Medical University, Fuzhou, China
| | - Borui Chen
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Haoming Zhu
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Yu Wang
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Jianping Lin
- The School of Health, Fujian Medical University, Fuzhou, China
| | - Shizhong Wang
- The School of Health, Fujian Medical University, Fuzhou, China
| | - Shaoqing Chen
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| |
Collapse
|
27
|
Paez HG, Pitzer CR, Alway SE. Age-Related Dysfunction in Proteostasis and Cellular Quality Control in the Development of Sarcopenia. Cells 2023; 12:cells12020249. [PMID: 36672183 PMCID: PMC9856405 DOI: 10.3390/cells12020249] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2022] [Revised: 01/04/2023] [Accepted: 01/04/2023] [Indexed: 01/11/2023] Open
Abstract
Sarcopenia is a debilitating skeletal muscle disease that accelerates in the last decades of life and is characterized by marked deficits in muscle strength, mass, quality, and metabolic health. The multifactorial causes of sarcopenia have proven difficult to treat and involve a complex interplay between environmental factors and intrinsic age-associated changes. It is generally accepted that sarcopenia results in a progressive loss of skeletal muscle function that exceeds the loss of mass, indicating that while loss of muscle mass is important, loss of muscle quality is the primary defect with advanced age. Furthermore, preclinical models have suggested that aged skeletal muscle exhibits defects in cellular quality control such as the degradation of damaged mitochondria. Recent evidence suggests that a dysregulation of proteostasis, an important regulator of cellular quality control, is a significant contributor to the aging-associated declines in muscle quality, function, and mass. Although skeletal muscle mammalian target of rapamycin complex 1 (mTORC1) plays a critical role in cellular control, including skeletal muscle hypertrophy, paradoxically, sustained activation of mTORC1 recapitulates several characteristics of sarcopenia. Pharmaceutical inhibition of mTORC1 as well as caloric restriction significantly improves muscle quality in aged animals, however, the mechanisms controlling cellular proteostasis are not fully known. This information is important for developing effective therapeutic strategies that mitigate or prevent sarcopenia and associated disability. This review identifies recent and historical understanding of the molecular mechanisms of proteostasis driving age-associated muscle loss and suggests potential therapeutic interventions to slow or prevent sarcopenia.
Collapse
Affiliation(s)
- Hector G. Paez
- Department of Physiology, College of Medicine, University of Tennessee Health Science Center, Memphis, TN 38163, USA
- Integrated Biomedical Sciences Graduate Program, College of Graduate Health Sciences, University of Tennessee Health Science Center, Memphis, TN 38163, USA
- Laboratory of Muscle Biology and Sarcopenia, Department of Physical Therapy, College of Health Professions, University of Tennessee Health Science Center, Memphis, TN 38163, USA
- Center for Muscle, Metabolism and Neuropathology, Division of Regenerative and Rehabilitation Sciences, College of Health Professions, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Christopher R. Pitzer
- Department of Physiology, College of Medicine, University of Tennessee Health Science Center, Memphis, TN 38163, USA
- Integrated Biomedical Sciences Graduate Program, College of Graduate Health Sciences, University of Tennessee Health Science Center, Memphis, TN 38163, USA
- Laboratory of Muscle Biology and Sarcopenia, Department of Physical Therapy, College of Health Professions, University of Tennessee Health Science Center, Memphis, TN 38163, USA
- Center for Muscle, Metabolism and Neuropathology, Division of Regenerative and Rehabilitation Sciences, College of Health Professions, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Stephen E. Alway
- Department of Physiology, College of Medicine, University of Tennessee Health Science Center, Memphis, TN 38163, USA
- Integrated Biomedical Sciences Graduate Program, College of Graduate Health Sciences, University of Tennessee Health Science Center, Memphis, TN 38163, USA
- Laboratory of Muscle Biology and Sarcopenia, Department of Physical Therapy, College of Health Professions, University of Tennessee Health Science Center, Memphis, TN 38163, USA
- Center for Muscle, Metabolism and Neuropathology, Division of Regenerative and Rehabilitation Sciences, College of Health Professions, University of Tennessee Health Science Center, Memphis, TN 38163, USA
- The Tennessee Institute of Regenerative Medicine, Memphis, TN 38163, USA
- Correspondence:
| |
Collapse
|
28
|
Picca A, Triolo M, Wohlgemuth SE, Martenson MS, Mankowski RT, Anton SD, Marzetti E, Leeuwenburgh C, Hood DA. Relationship between Mitochondrial Quality Control Markers, Lower Extremity Tissue Composition, and Physical Performance in Physically Inactive Older Adults. Cells 2023; 12:183. [PMID: 36611976 PMCID: PMC9818256 DOI: 10.3390/cells12010183] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 12/29/2022] [Accepted: 12/30/2022] [Indexed: 01/03/2023] Open
Abstract
Altered mitochondrial quality and function in muscle may be involved in age-related physical function decline. The role played by the autophagy-lysosome system, a major component of mitochondrial quality control (MQC), is incompletely understood. This study was undertaken to obtain initial indications on the relationship between autophagy, mitophagy, and lysosomal markers in muscle and measures of physical performance and lower extremity tissue composition in young and older adults. Twenty-three participants were enrolled, nine young (mean age: 24.3 ± 4.3 years) and 14 older adults (mean age: 77.9 ± 6.3 years). Lower extremity tissue composition was quantified volumetrically by magnetic resonance imaging and a tissue composition index was calculated as the ratio between muscle and intermuscular adipose tissue volume. Physical performance in older participants was assessed via the Short Physical Performance Battery (SPPB). Protein levels of the autophagy marker p62, the mitophagy mediator BCL2/adenovirus E1B 19 kDa protein-interacting protein 3 (BNIP3), the lysosomal markers transcription factor EB, vacuolar-type ATPase, and lysosomal-associated membrane protein 1 were measured by Western immunoblotting in vastus lateralis muscle biopsies. Older adults had smaller muscle volume and lower tissue composition index than young participants. The protein content of p62 and BNIP3 was higher in older adults. A negative correlation was detected between p62 and BNIP3 and the tissue composition index. p62 and BNIP3 were also related to the performance on the 5-time sit-to-stand test of the SPPB. Our results suggest that an altered expression of markers of the autophagy/mitophagy-lysosomal system is related to deterioration of lower extremity tissue composition and muscle dysfunction. Additional studies are needed to clarify the role of defective MQC in human muscle aging and identify novel biological targets for drug development.
Collapse
Affiliation(s)
- Anna Picca
- Fondazione Policlinico Universitario “A. Gemelli”, IRCCS, 00168 Rome, Italy
- Department of Medicine and Surgery, LUM University, 70100 Casamassima, Italy
| | - Matthew Triolo
- Muscle Health Research Centre, York University, Toronto, ON M3J 1P3, Canada
- School of Kinesiology and Health Science, York University, Toronto, ON M3J 1P3, Canada
| | | | - Matthew S. Martenson
- Department of Physiology and Aging, University of Florida, Gainesville, FL 32610, USA
| | - Robert T. Mankowski
- Department of Physiology and Aging, University of Florida, Gainesville, FL 32610, USA
| | - Stephen D. Anton
- Department of Physiology and Aging, University of Florida, Gainesville, FL 32610, USA
| | - Emanuele Marzetti
- Fondazione Policlinico Universitario “A. Gemelli”, IRCCS, 00168 Rome, Italy
- Department of Geriatrics and Orthopedics, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | | | - David A. Hood
- Muscle Health Research Centre, York University, Toronto, ON M3J 1P3, Canada
- School of Kinesiology and Health Science, York University, Toronto, ON M3J 1P3, Canada
| |
Collapse
|
29
|
Pires RA, Correia TML, Almeida AA, Coqueiro RDS, Machado M, Teles MF, Peixoto ÁS, Queiroz RF, Pereira R. Time-Course of Redox Status, Redox-Related, and Mitochondrial-Dynamics-Related Gene Expression after an Acute Bout of Different Physical Exercise Protocols. Life (Basel) 2022; 12:life12122113. [PMID: 36556478 PMCID: PMC9781780 DOI: 10.3390/life12122113] [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: 11/17/2022] [Revised: 12/10/2022] [Accepted: 12/13/2022] [Indexed: 12/23/2022] Open
Abstract
We investigated the magnitude of exercise-induced changes in muscular bioenergetics, redox balance, mitochondrial function, and gene expression within 24 h after the exercise bouts performed with different intensities, durations, and execution modes (continuous or with intervals). Sixty-five male Swiss mice were divided into four groups: one control (n = 5) and three experimental groups (20 animals/group), submitted to a forced swimming bout with an additional load (% of animal weight): low-intensity continuous (LIC), high-intensity continuous (HIC), and high-intensity interval (HII). Five animals from each group were euthanized at 0 h, 6 h, 12 h, and 24 h postexercise. Gastrocnemius muscle was removed to analyze the expression of genes involved in mitochondrial biogenesis (Ppargc1a), fusion (Mfn2), fission (Dnm1L), and mitophagy (Park2), as well as inflammation (Nos2) and antioxidant defense (Nfe2l2, GPx1). Lipid peroxidation (TBARS), total peroxidase, glutathione peroxidase (GPx), and citrate synthase (CS) activity were also measured. Lactacidemia was measured from a blood sample obtained immediately postexercise. Lactacidemia was higher the higher the exercise intensity (LIC < HIC < HII), while the inverse was observed for TBARS levels. The CS activity was higher in the HII group than the other groups. The antioxidant activity was higher 24 h postexercise in all groups compared to the control and greater in the HII group than the LIC and HIC groups. The gene expression profile exhibited a particular profile for each exercise protocol, but with some similarities between the LIC and HII groups. Taken together, these results suggest that the intervals applied to high-intensity exercise seem to minimize the signs of oxidative damage and drive the mitochondrial dynamics to maintain the mitochondrial network, similar to low-intensity continuous exercise.
Collapse
Affiliation(s)
- Ramon Alves Pires
- Integrative Physiology Research Center, Department of Biological Sciences, Universidade Estadual do Sudoeste da Bahia (UESB), Jequie 45210-506, Brazil
- Multicentric Postgraduate Program in Biochemistry and Molecular (Brazilian Society for Biochemistry and Molecular Biology), Universidade Estadual do Sudoeste da Bahia (UESB), Vitoria da Conquista, Jequie 45210-506, Brazil
| | - Thiago Macedo Lopes Correia
- Integrative Physiology Research Center, Department of Biological Sciences, Universidade Estadual do Sudoeste da Bahia (UESB), Jequie 45210-506, Brazil
- Multicentric Postgraduate Program in Physiological Sciences (Brazilian Society of Physiology), Universidade Federal da Bahia (UFBA), Vitoria da Conquista, Jequie 45210-506, Brazil
| | - Amanda Alves Almeida
- Integrative Physiology Research Center, Department of Biological Sciences, Universidade Estadual do Sudoeste da Bahia (UESB), Jequie 45210-506, Brazil
- Multicentric Postgraduate Program in Physiological Sciences (Brazilian Society of Physiology), Universidade Federal da Bahia (UFBA), Vitoria da Conquista, Jequie 45210-506, Brazil
| | - Raildo da Silva Coqueiro
- Integrative Physiology Research Center, Department of Biological Sciences, Universidade Estadual do Sudoeste da Bahia (UESB), Jequie 45210-506, Brazil
| | - Marco Machado
- Integrative Physiology Research Center, Department of Biological Sciences, Universidade Estadual do Sudoeste da Bahia (UESB), Jequie 45210-506, Brazil
- Fundação Universitária de Itaperuna (FUNITA), Itaperuna 28300-000, Brazil
- Laboratory of Physiology and Biokinetic, Faculty of Biological Sciences and Health, Universidade Iguaçu Campus V, Itaperuna 28300-000, Brazil
| | - Mauro Fernandes Teles
- Integrative Physiology Research Center, Department of Biological Sciences, Universidade Estadual do Sudoeste da Bahia (UESB), Jequie 45210-506, Brazil
| | - Álbert Souza Peixoto
- Instituto de Ciências Biomédicas (ICB), Universidade de São Paulo (USP), São Paulo 05508-000, Brazil
| | - Raphael Ferreira Queiroz
- Multicentric Postgraduate Program in Biochemistry and Molecular (Brazilian Society for Biochemistry and Molecular Biology), Universidade Estadual do Sudoeste da Bahia (UESB), Vitoria da Conquista, Jequie 45210-506, Brazil
- Postgraduate Program in Biosciences, Universidade Federal da Bahia, Campus Anísio Teixeira, Vitória da Conquista 40110-100, Brazil
| | - Rafael Pereira
- Integrative Physiology Research Center, Department of Biological Sciences, Universidade Estadual do Sudoeste da Bahia (UESB), Jequie 45210-506, Brazil
- Multicentric Postgraduate Program in Biochemistry and Molecular (Brazilian Society for Biochemistry and Molecular Biology), Universidade Estadual do Sudoeste da Bahia (UESB), Vitoria da Conquista, Jequie 45210-506, Brazil
- Multicentric Postgraduate Program in Physiological Sciences (Brazilian Society of Physiology), Universidade Federal da Bahia (UFBA), Vitoria da Conquista, Jequie 45210-506, Brazil
- Correspondence:
| |
Collapse
|
30
|
Kolodziej F, McDonagh B, Burns N, Goljanek-Whysall K. MicroRNAs as the Sentinels of Redox and Hypertrophic Signalling. Int J Mol Sci 2022; 23:ijms232314716. [PMID: 36499053 PMCID: PMC9737617 DOI: 10.3390/ijms232314716] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 11/22/2022] [Accepted: 11/23/2022] [Indexed: 11/26/2022] Open
Abstract
Oxidative stress and inflammation are associated with skeletal muscle function decline with ageing or disease or inadequate exercise and/or poor diet. Paradoxically, reactive oxygen species and inflammatory cytokines are key for mounting the muscular and systemic adaptive responses to endurance and resistance exercise. Both ageing and lifestyle-related metabolic dysfunction are strongly linked to exercise redox and hypertrophic insensitivity. The adaptive inability and consequent exercise intolerance may discourage people from physical training resulting in a vicious cycle of under-exercising, energy surplus, chronic mitochondrial stress, accelerated functional decline and increased susceptibility to serious diseases. Skeletal muscles are malleable and dynamic organs, rewiring their metabolism depending on the metabolic or mechanical stress resulting in a specific phenotype. Endogenous RNA silencing molecules, microRNAs, are regulators of these metabolic/phenotypic shifts in skeletal muscles. Skeletal muscle microRNA profiles at baseline and in response to exercise have been observed to differ between adult and older people, as well as trained vs. sedentary individuals. Likewise, the circulating microRNA blueprint varies based on age and training status. Therefore, microRNAs emerge as key regulators of metabolic health/capacity and hormetic adaptability. In this narrative review, we summarise the literature exploring the links between microRNAs and skeletal muscle, as well as systemic adaptation to exercise. We expand a mathematical model of microRNA burst during adaptation to exercise through supporting data from the literature. We describe a potential link between the microRNA-dependent regulation of redox-signalling sensitivity and the ability to mount a hypertrophic response to exercise or nutritional cues. We propose a hypothetical model of endurance exercise-induced microRNA "memory cloud" responsible for establishing a landscape conducive to aerobic as well as anabolic adaptation. We suggest that regular aerobic exercise, complimented by a healthy diet, in addition to promoting mitochondrial health and hypertrophic/insulin sensitivity, may also suppress the glycolytic phenotype and mTOR signalling through miRNAs which in turn promote systemic metabolic health.
Collapse
Affiliation(s)
- Filip Kolodziej
- Department of Physiology, School of Medicine, CMNHS, University of Galway, H91TK33 Galway, Ireland
| | - Brian McDonagh
- Department of Physiology, School of Medicine, CMNHS, University of Galway, H91TK33 Galway, Ireland
| | - Nicole Burns
- Department of Physiology, School of Medicine, CMNHS, University of Galway, H91TK33 Galway, Ireland
| | - Katarzyna Goljanek-Whysall
- Department of Physiology, School of Medicine, CMNHS, University of Galway, H91TK33 Galway, Ireland
- Institute of Life Course and Medical Science, University of Liverpool, Liverpool L69 3BX, UK
| |
Collapse
|
31
|
Bellanti F, Lo Buglio A, Vendemiale G. Muscle Delivery of Mitochondria-Targeted Drugs for the Treatment of Sarcopenia: Rationale and Perspectives. Pharmaceutics 2022; 14:pharmaceutics14122588. [PMID: 36559079 PMCID: PMC9782427 DOI: 10.3390/pharmaceutics14122588] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 11/18/2022] [Accepted: 11/19/2022] [Indexed: 11/27/2022] Open
Abstract
An impairment in mitochondrial homeostasis plays a crucial role in the process of aging and contributes to the incidence of age-related diseases, including sarcopenia, which is defined as an age-dependent loss of muscle mass and strength. Mitochondrial dysfunction exerts a negative impact on several cellular activities, including bioenergetics, metabolism, and apoptosis. In sarcopenia, mitochondria homeostasis is disrupted because of reduced oxidative phosphorylation and ATP generation, the enhanced production of reactive species, and impaired antioxidant defense. This review re-establishes the most recent evidence on mitochondrial defects that are thought to be relevant in the pathogenesis of sarcopenia and that may represent promising therapeutic targets for its prevention/treatment. Furthermore, we describe mechanisms of action and translational potential of promising mitochondria-targeted drug delivery systems, including molecules able to boost the metabolism and bioenergetics, counteract apoptosis, antioxidants to scavenge reactive species and decrease oxidative stress, and target mitophagy. Even though these mitochondria-delivered strategies demonstrate to be promising in preclinical models, their use needs to be promoted for clinical studies. Therefore, there is a compelling demand to further understand the mechanisms modulating mitochondrial homeostasis, to characterize powerful compounds that target muscle mitochondria to prevent sarcopenia in aged people.
Collapse
|
32
|
Triolo M, Bhattacharya D, Hood DA. Denervation induces mitochondrial decline and exacerbates lysosome dysfunction in middle-aged mice. Aging (Albany NY) 2022; 14:8900-8913. [PMID: 36342767 PMCID: PMC9740366 DOI: 10.18632/aging.204365] [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: 08/25/2022] [Accepted: 10/24/2022] [Indexed: 11/06/2022]
Abstract
With age, skeletal muscle undergoes a progressive decline in size and quality. Imbalanced mitochondrial turnover and the resultant dysfunction contribute to these phenotypic alterations. Motor neuron denervation (Den) is a contributor to the etiology of muscle atrophy associated with age. Further, aged muscle exhibits reduced plasticity to both enhanced and suppressed contractile activity. It remains unclear when the onset of this blunted response occurs, and how middle-aged muscle adapts to denervation. The purpose of this study was to compare mitochondrial turnover pathways in young (Y, ~5months) and middle-aged (MA, ~15months) mice, and determine the influence of Den. Transgenic mt-Keima mice were subjected to 1,3 or 7 days of Den. Muscle mass, mitochondrial content, and PGC-1α protein were not different between Y and MA mice. However, indications of enhanced mitochondrial fission and mitophagy were evident in MA muscle which were supported by a greater abundance of lysosome proteins. Den resulted in muscle atrophy and reductions in mitochondrial protein content by 7-days. These changes occurred concomitant with modest decreases in PGC-1α protein, but without further elevations in mitophagy. Although both autophagosomal and lysosomal proteins were elevated, evidence of lysosome dysfunction was present following Den in MA mice. These data suggest that increases in fission drive an acceleration of mitophagy in muscle of MA mice to preserve mitochondrial quality. Den exacerbates the aging phenotype by reducing biogenesis in the absence of a change in mitophagy, perhaps limited by lysosomal capacity, leading to an accumulation of dysfunctional mitochondria with an age-related loss of neuromuscular innervation.
Collapse
Affiliation(s)
- Matthew Triolo
- Muscle Health Research Centre, York University, Toronto, Ontario M3J 1P3, Canada,School of Kinesiology and Health Science, York University, Toronto, Ontario M3J 1P3, Canada
| | - Debasmita Bhattacharya
- Muscle Health Research Centre, York University, Toronto, Ontario M3J 1P3, Canada,School of Kinesiology and Health Science, York University, Toronto, Ontario M3J 1P3, Canada
| | - David A. Hood
- Muscle Health Research Centre, York University, Toronto, Ontario M3J 1P3, Canada,School of Kinesiology and Health Science, York University, Toronto, Ontario M3J 1P3, Canada
| |
Collapse
|
33
|
Pinto AP, Muñoz VR, da Rocha AL, Rovina RL, Ferrari GD, Alberici LC, Simabuco FM, Teixeira GR, Pauli JR, de Moura LP, Cintra DE, Ropelle ER, Freitas EC, Rivas DA, da Silva ASR. IL-6 deletion decreased REV-ERBα protein and influenced autophagy and mitochondrial markers in the skeletal muscle after acute exercise. Front Immunol 2022; 13:953272. [PMID: 36311768 PMCID: PMC9608639 DOI: 10.3389/fimmu.2022.953272] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Accepted: 09/27/2022] [Indexed: 01/28/2024] Open
Abstract
Interleukin 6 (IL-6) acts as a pro and anti-inflammatory cytokine, has an intense correlation with exercise intensity, and activates various pathways such as autophagy and mitochondrial unfolded protein response. Also, IL-6 is interconnected to circadian clock-related inflammation and can be suppressed by the nuclear receptor subfamily 1, group D, member 1 (Nr1d1, protein product REV-ERBα). Since IL-6 is linked to physical exercise-modulated metabolic pathways such as autophagy and mitochondrial metabolism, we investigated the relationship of IL-6 with REV-ERBα in the adaptations of these molecular pathways in response to acute intense physical exercise in skeletal muscle. The present study was divided into three experiments. In the first one, wild-type (WT) and IL-6 knockout (IL-6 KO) mice were divided into three groups: Basal time (Basal; sacrificed before the acute exercise), 1 hour (1hr post-Ex; sacrificed 1 hour after the acute exercise), and 3 hours (3hr post-Ex; sacrificed 3 hours after the acute exercise). In the second experiment, C2C12 cells received IL-6 physiological concentrations or REV-ERBα agonist, SR9009. In the last experiment, WT mice received SR9009 injections. After the protocols, the gastrocnemius muscle or the cells were collected for reverse transcription-quantitative polymerase chain reaction (RTq-PCR) and immunoblotting techniques. In summary, the downregulation of REV-ERBα, autophagic flux, and most mitochondrial genes was verified in the IL-6 KO mice independent of exercise. The WT and IL-6 KO treated with SR9009 showed an upregulation of autophagic genes. C2C12 cells receiving IL-6 did not modulate the Nr1d1 mRNA levels but upregulated the expression of some mitochondrial genes. However, when treated with SR9009, IL-6 and mitochondrial gene expression were upregulated in C2C12 cells. The autophagic flux in C2C12 suggest the participation of REV-ERBα protein in the IL-6-induced autophagy. In conclusion, the present study verified that the adaptations required through physical exercise (increases in mitochondrial content and improvement of autophagy machinery) might be intermediated by an interaction between IL-6 and REVERBα.
Collapse
Affiliation(s)
- Ana P. Pinto
- School of Physical Education and Sport of Ribeirão Preto, University of São Paulo (USP), São Paulo, Brazil
| | - Vitor R. Muñoz
- School of Physical Education and Sport of Ribeirão Preto, University of São Paulo (USP), São Paulo, Brazil
| | - Alisson L. da Rocha
- Postgraduate Program in Rehabilitation and Functional Performance, Ribeirão Preto Medical School, University of São Paulo (USP), São Paulo, Brazil
| | - Rafael L. Rovina
- School of Physical Education and Sport of Ribeirão Preto, University of São Paulo (USP), São Paulo, Brazil
| | - Gustavo D. Ferrari
- Department of Biomolecular Sciences, School of Pharmaceutical Sciences of Ribeirao Preto, Faculty of Pharmaceutical Sciences of Ribeirao Preto, University of São Paulo (FCFRP USP), Sao Paulo, Brazil
| | - Luciane C. Alberici
- Department of Biomolecular Sciences, School of Pharmaceutical Sciences of Ribeirao Preto, Faculty of Pharmaceutical Sciences of Ribeirao Preto, University of São Paulo (FCFRP USP), Sao Paulo, Brazil
| | - Fernando M. Simabuco
- Laboratory of Molecular Biology of Exercise (LaBMEx), School of Applied Sciences, University of Campinas (UNICAMP), São Paulo, Brazil
- Department of Biochemistry, Federal University of São Paulo (UNIFESP), São Paulo, Brazil
| | - Giovana R. Teixeira
- Multicentric Program of Postgraduate in Physiological Sciences, School of Dentistry of Araçatuba, São Paulo State University (UNESP), São Paulo, Brazil
- Department of Physical Education, State University of São Paulo (UNESP), São Paulo, Brazil
| | - José R. Pauli
- Laboratory of Molecular Biology of Exercise (LaBMEx), School of Applied Sciences, University of Campinas (UNICAMP), São Paulo, Brazil
| | - Leandro P. de Moura
- Laboratory of Molecular Biology of Exercise (LaBMEx), School of Applied Sciences, University of Campinas (UNICAMP), São Paulo, Brazil
| | - Dennys E. Cintra
- Laboratory of Molecular Biology of Exercise (LaBMEx), School of Applied Sciences, University of Campinas (UNICAMP), São Paulo, Brazil
| | - Eduardo R. Ropelle
- Laboratory of Molecular Biology of Exercise (LaBMEx), School of Applied Sciences, University of Campinas (UNICAMP), São Paulo, Brazil
| | - Ellen C. Freitas
- School of Physical Education and Sport of Ribeirão Preto, University of São Paulo (USP), São Paulo, Brazil
| | - Donato A. Rivas
- Multicentric Program of Postgraduate in Physiological Sciences, School of Dentistry of Araçatuba, São Paulo State University (UNESP), São Paulo, Brazil
| | - Adelino S. R. da Silva
- School of Physical Education and Sport of Ribeirão Preto, University of São Paulo (USP), São Paulo, Brazil
- Postgraduate Program in Rehabilitation and Functional Performance, Ribeirão Preto Medical School, University of São Paulo (USP), São Paulo, Brazil
| |
Collapse
|
34
|
Gremminger VL, Omosule CL, Crawford TK, Cunningham R, Rector RS, Phillips CL. Skeletal muscle mitochondrial function and whole-body metabolic energetics in the +/G610C mouse model of osteogenesis imperfecta. Mol Genet Metab 2022; 136:315-323. [PMID: 35725939 PMCID: PMC11587666 DOI: 10.1016/j.ymgme.2022.06.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Revised: 06/08/2022] [Accepted: 06/09/2022] [Indexed: 11/17/2022]
Abstract
Osteogenesis imperfecta (OI) is rare heritable connective tissue disorder that most often arises from mutations in the type I collagen genes, COL1A1 and COL1A2, displaying a range of symptoms including skeletal fragility, short stature, blue-gray sclera, and muscle weakness. Recent investigations into the intrinsic muscle weakness have demonstrated reduced contractile generating force in some murine models consistent with patient population studies, as well as alterations in whole body bioenergetics. Muscle weakness is found in approximately 80% of patients and has been equivocal in OI mouse models. Understanding the mechanism responsible for OI muscle weakness is crucial in building our knowledge of muscle bone cross-talk via mechanotransduction and biochemical signaling, and for potential novel therapeutic approaches. In this study we evaluated skeletal muscle mitochondrial function and whole-body bioenergetics in the heterozygous +/G610C (Amish) mouse modeling mild/moderate human type I/VI OI and minimal skeletal muscle weakness. Our analyses revealed several changes in the +/G610C mouse relative to their wildtype littermates including reduced state 3 mitochondrial respiration, increased mitochondrial citrate synthase activity, increased Parkin and p62 protein content, and an increased respiratory quotient. These changes may represent the ability of the +/G610C mouse to compensate for mitochondrial and metabolic changes that may arise due to type I collagen mutations and may also account for the lack of muscle weakness observed in the +/G610C model relative to the more severe OI models.
Collapse
Affiliation(s)
- Victoria L Gremminger
- Department of Biochemistry, University of Missouri, Columbia, MO 65211, United States of America
| | - Catherine L Omosule
- Department of Biochemistry, University of Missouri, Columbia, MO 65211, United States of America
| | - Tara K Crawford
- Department of Biochemistry, University of Missouri, Columbia, MO 65211, United States of America
| | - Rory Cunningham
- Departments of Nutrition and Exercise Physiology and Medicine-GI, University of Missouri, Research Service-Harry S Truman Memorial VA Hospital, Columbia, MO 65201, United States of America
| | - R Scott Rector
- Departments of Nutrition and Exercise Physiology and Medicine-GI, University of Missouri, Research Service-Harry S Truman Memorial VA Hospital, Columbia, MO 65201, United States of America
| | - Charlotte L Phillips
- Department of Biochemistry, University of Missouri, Columbia, MO 65211, United States of America; Department of Child Health, University of Missouri, Columbia, MO 65212, United States of America.
| |
Collapse
|
35
|
Pinto AP, da Rocha AL, Teixeira GR, Rovina RL, Veras ASC, Frantz F, Pauli JR, de Moura LP, Cintra DE, Ropelle ER, Quadrilatero J, da Silva ASR. Rapamycin did not prevent the excessive exercise-induced hepatic fat accumulation. Life Sci 2022; 306:120800. [PMID: 35839860 DOI: 10.1016/j.lfs.2022.120800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 06/30/2022] [Accepted: 07/09/2022] [Indexed: 01/18/2023]
Affiliation(s)
- Ana P Pinto
- Postgraduate Program in Rehabilitation and Functional Performance, Ribeirão Preto Medical School, University of São Paulo (USP), Ribeirão Preto, São Paulo, Brazil
| | - Alisson L da Rocha
- Postgraduate Program in Rehabilitation and Functional Performance, Ribeirão Preto Medical School, University of São Paulo (USP), Ribeirão Preto, São Paulo, Brazil
| | - Giovana R Teixeira
- Multicentric Program of Postgraduate in Physiological Sciences, São Paulo State University (UNESP), School of Dentistry of Araçatuba, Araçatuba, São Paulo, Brazil; Department of Physical Education, State University of São Paulo (UNESP), Presidente Prudente, São Paulo, Brazil
| | - Rafael L Rovina
- School of Physical Education and Sport of Ribeirão Preto, University of São Paulo (USP), Ribeirão Preto, São Paulo, Brazil
| | - Allice S C Veras
- Multicentric Program of Postgraduate in Physiological Sciences, São Paulo State University (UNESP), School of Dentistry of Araçatuba, Araçatuba, São Paulo, Brazil
| | - Fabiani Frantz
- Faculty of Pharmaceutical Sciences of Ribeirão Preto, Department of Clinical, Toxicological, and Bromatological Analysis, University of São Paulo (USP), Ribeirão Preto, São Paulo, Brazil
| | - José R Pauli
- Laboratory of Molecular Biology of Exercise (LaBMEx), School of Applied Sciences, University of Campinas (UNICAMP), Limeira, São Paulo, Brazil
| | - Leandro P de Moura
- Laboratory of Molecular Biology of Exercise (LaBMEx), School of Applied Sciences, University of Campinas (UNICAMP), Limeira, São Paulo, Brazil
| | - Dennys E Cintra
- Laboratory of Molecular Biology of Exercise (LaBMEx), School of Applied Sciences, University of Campinas (UNICAMP), Limeira, São Paulo, Brazil
| | - Eduardo R Ropelle
- Laboratory of Molecular Biology of Exercise (LaBMEx), School of Applied Sciences, University of Campinas (UNICAMP), Limeira, São Paulo, Brazil
| | - Joe Quadrilatero
- Department of Kinesiology and Health Sciences, University of Waterloo, Waterloo, Ontario, Canada
| | - Adelino S R da Silva
- Postgraduate Program in Rehabilitation and Functional Performance, Ribeirão Preto Medical School, University of São Paulo (USP), Ribeirão Preto, São Paulo, Brazil; School of Physical Education and Sport of Ribeirão Preto, University of São Paulo (USP), Ribeirão Preto, São Paulo, Brazil.
| |
Collapse
|
36
|
Hughes DC, Baehr LM, Waddell DS, Sharples AP, Bodine SC. Ubiquitin Ligases in Longevity and Aging Skeletal Muscle. Int J Mol Sci 2022; 23:7602. [PMID: 35886949 PMCID: PMC9315556 DOI: 10.3390/ijms23147602] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 07/07/2022] [Accepted: 07/08/2022] [Indexed: 12/07/2022] Open
Abstract
The development and prevalence of diseases associated with aging presents a global health burden on society. One hallmark of aging is the loss of proteostasis which is caused in part by alterations to the ubiquitin-proteasome system (UPS) and lysosome-autophagy system leading to impaired function and maintenance of mass in tissues such as skeletal muscle. In the instance of skeletal muscle, the impairment of function occurs early in the aging process and is dependent on proteostatic mechanisms. The UPS plays a pivotal role in degradation of misfolded and aggregated proteins. For the purpose of this review, we will discuss the role of the UPS system in the context of age-related loss of muscle mass and function. We highlight the significant role that E3 ubiquitin ligases play in the turnover of key components (e.g., mitochondria and neuromuscular junction) essential to skeletal muscle function and the influence of aging. In addition, we will briefly discuss the contribution of the UPS system to lifespan. By understanding the UPS system as part of the proteostasis network in age-related diseases and disorders such as sarcopenia, new discoveries can be made and new interventions can be developed which will preserve muscle function and maintain quality of life with advancing age.
Collapse
Affiliation(s)
- David C. Hughes
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA; (L.M.B.); (S.C.B.)
| | - Leslie M. Baehr
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA; (L.M.B.); (S.C.B.)
| | - David S. Waddell
- Department of Biology, University of North Florida, Jacksonville, FL 32224, USA;
| | - Adam P. Sharples
- Institute for Physical Performance, Norwegian School of Sport Sciences (NiH), 0863 Oslo, Norway;
| | - Sue C. Bodine
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA; (L.M.B.); (S.C.B.)
| |
Collapse
|
37
|
Li J, Wang Z, Li C, Song Y, Wang Y, Bo H, Zhang Y. Impact of Exercise and Aging on Mitochondrial Homeostasis in Skeletal Muscle: Roles of ROS and Epigenetics. Cells 2022; 11:cells11132086. [PMID: 35805170 PMCID: PMC9266156 DOI: 10.3390/cells11132086] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 06/22/2022] [Accepted: 06/28/2022] [Indexed: 02/01/2023] Open
Abstract
Aging causes degenerative changes such as epigenetic changes and mitochondrial dysfunction in skeletal muscle. Exercise can upregulate muscle mitochondrial homeostasis and enhance antioxidant capacity and represents an effective treatment to prevent muscle aging. Epigenetic changes such as DNA methylation, histone posttranslational modifications, and microRNA expression are involved in the regulation of exercise-induced adaptive changes in muscle mitochondria. Reactive oxygen species (ROS) play an important role in signaling molecules in exercise-induced muscle mitochondrial health benefits, and strong evidence emphasizes that exercise-induced ROS can regulate gene expression via epigenetic mechanisms. The majority of mitochondrial proteins are imported into mitochondria from the cytosol, so mitochondrial homeostasis is regulated by nuclear epigenetic mechanisms. Exercise can reverse aging-induced changes in myokine expression by modulating epigenetic mechanisms. In this review, we provide an overview of the role of exercise-generated ROS in the regulation of mitochondrial homeostasis mediated by epigenetic mechanisms. In addition, the potential epigenetic mechanisms involved in exercise-induced myokine expression are reviewed.
Collapse
Affiliation(s)
- Jialin Li
- Tianjin Key Laboratory of Exercise Physiology and Sports Medicine, Institute of Exercise and Health, Tianjin University of Sport, Tianjin 301617, China; (J.L.); (Z.W.); (C.L.); (Y.S.); (Y.W.)
| | - Zhe Wang
- Tianjin Key Laboratory of Exercise Physiology and Sports Medicine, Institute of Exercise and Health, Tianjin University of Sport, Tianjin 301617, China; (J.L.); (Z.W.); (C.L.); (Y.S.); (Y.W.)
| | - Can Li
- Tianjin Key Laboratory of Exercise Physiology and Sports Medicine, Institute of Exercise and Health, Tianjin University of Sport, Tianjin 301617, China; (J.L.); (Z.W.); (C.L.); (Y.S.); (Y.W.)
| | - Yu Song
- Tianjin Key Laboratory of Exercise Physiology and Sports Medicine, Institute of Exercise and Health, Tianjin University of Sport, Tianjin 301617, China; (J.L.); (Z.W.); (C.L.); (Y.S.); (Y.W.)
| | - Yan Wang
- Tianjin Key Laboratory of Exercise Physiology and Sports Medicine, Institute of Exercise and Health, Tianjin University of Sport, Tianjin 301617, China; (J.L.); (Z.W.); (C.L.); (Y.S.); (Y.W.)
| | - Hai Bo
- Tianjin Key Laboratory of Exercise Physiology and Sports Medicine, Institute of Exercise and Health, Tianjin University of Sport, Tianjin 301617, China; (J.L.); (Z.W.); (C.L.); (Y.S.); (Y.W.)
- Department of Military Training Medicines, Logistics University of Chinese People’s Armed Police Force, Tianjin 300162, China
- Correspondence: (H.B.); (Y.Z.)
| | - Yong Zhang
- Tianjin Key Laboratory of Exercise Physiology and Sports Medicine, Institute of Exercise and Health, Tianjin University of Sport, Tianjin 301617, China; (J.L.); (Z.W.); (C.L.); (Y.S.); (Y.W.)
- Correspondence: (H.B.); (Y.Z.)
| |
Collapse
|
38
|
Triolo M, Oliveira AN, Kumari R, Hood DA. The influence of age, sex, and exercise on autophagy, mitophagy, and lysosome biogenesis in skeletal muscle. Skelet Muscle 2022; 12:13. [PMID: 35690879 PMCID: PMC9188089 DOI: 10.1186/s13395-022-00296-7] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Accepted: 05/15/2022] [Indexed: 12/25/2022] Open
Abstract
Background Aging decreases skeletal muscle mass and quality. Maintenance of healthy muscle is regulated by a balance between protein and organellar synthesis and their degradation. The autophagy-lysosome system is responsible for the selective degradation of protein aggregates and organelles, such as mitochondria (i.e., mitophagy). Little data exist on the independent and combined influence of age, biological sex, and exercise on the autophagy system and lysosome biogenesis. The purpose of this study was to characterize sex differences in autophagy and lysosome biogenesis in young and aged muscle and to determine if acute exercise influences these processes. Methods Young (4–6 months) and aged (22–24 months) male and female mice were assigned to a sedentary or an acute exercise group. Mitochondrial content, the autophagy-lysosome system, and mitophagy were measured via protein analysis. A TFEB-promoter-construct was utilized to examine Tfeb transcription, and nuclear-cytosolic fractions allowed us to examine TFEB localization in sedentary and exercised muscle with age and sex. Results Our results indicate that female mice, both young and old, had more mitochondrial protein than age-matched males. However, mitochondria in the muscle of females had a reduced respiratory capacity. Mitochondrial content was only reduced with age in the male cohort. Young female mice had a greater abundance of autophagy, mitophagy, and lysosome proteins than young males; however, increases were evident with age irrespective of sex. Young sedentary female mice had indices of greater autophagosomal turnover than male counterparts. Exhaustive exercise was able to stimulate autophagic clearance solely in young male mice. Similarly, nuclear TFEB protein was enhanced to a greater extent in young male, compared to young female mice following exercise, but no changes were observed in aged mice. Finally, TFEB-promoter activity was upregulated following exercise in both young and aged muscle. Conclusions The present study demonstrates that biological sex influences mitochondrial homeostasis, the autophagy-lysosome system, and mitophagy in skeletal muscle with age. Furthermore, our data suggest that young male mice have a more profound ability to activate these processes with exercise than in the other groups. Ultimately, this may contribute to a greater remodeling of muscle in response to exercise training in males.
Collapse
Affiliation(s)
- Matthew Triolo
- School of Kinesiology and Health Science, Muscle Health Research Centre, York University, Toronto, Ontario, M3J 1P3, Canada.,Muscle Health Research Centre, York University, Toronto, Ontario, M3J 1P3, Canada
| | - Ashley N Oliveira
- School of Kinesiology and Health Science, Muscle Health Research Centre, York University, Toronto, Ontario, M3J 1P3, Canada.,Muscle Health Research Centre, York University, Toronto, Ontario, M3J 1P3, Canada
| | - Rita Kumari
- School of Kinesiology and Health Science, Muscle Health Research Centre, York University, Toronto, Ontario, M3J 1P3, Canada.,Muscle Health Research Centre, York University, Toronto, Ontario, M3J 1P3, Canada
| | - David A Hood
- School of Kinesiology and Health Science, Muscle Health Research Centre, York University, Toronto, Ontario, M3J 1P3, Canada. .,Muscle Health Research Centre, York University, Toronto, Ontario, M3J 1P3, Canada.
| |
Collapse
|
39
|
Pinto AP, Ropelle ER, Quadrilatero J, da Silva ASR. Physical Exercise and Liver Autophagy: Potential Roles of IL-6 and Irisin. Exerc Sport Sci Rev 2022; 50:89-96. [PMID: 34961755 DOI: 10.1249/jes.0000000000000278] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Autophagic dysregulation contributes to liver diseases. Although some investigations have examined the effects of endurance and resistance exercise on autophagy activation, potential myokines responsible for skeletal muscle-liver crosstalk are still unknown. Based on experimental studies and bioinformatics, we hypothesized that interleukin 6 (IL-6) and irisin might be key players in the contraction-induced release of molecules that regulate liver autophagic responses.
Collapse
Affiliation(s)
- Ana P Pinto
- Postgraduate Program in Rehabilitation and Functional Performance, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto São Paulo, Brazil
| | - Eduardo R Ropelle
- Laboratory of Molecular Biology of Exercise, School of Applied Sciences, University of Campinas, Limeira, São Paulo, Brazil
| | - Joe Quadrilatero
- Department of Kinesiology, University of Waterloo, Waterloo, ON, Canada
| | | |
Collapse
|
40
|
Slavin MB, Memme JM, Oliveira AN, Moradi N, Hood DA. Regulatory networks controlling mitochondrial quality control in skeletal muscle. Am J Physiol Cell Physiol 2022; 322:C913-C926. [PMID: 35353634 DOI: 10.1152/ajpcell.00065.2022] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The adaptive plasticity of mitochondria within skeletal muscle is regulated by signals converging on a myriad of regulatory networks that operate during conditions of increased (i.e. exercise) and decreased (inactivity, disuse) energy requirements. Notably, some of the initial signals that induce adaptive responses are common to both conditions, differing in their magnitude and temporal pattern, to produce vastly opposing mitochondrial phenotypes. In response to exercise, signaling to PGC-1α and other regulators ultimately produces an abundance of high quality mitochondria, leading to reduced mitophagy and a higher mitochondrial content. This is accompanied by the presence of an enhanced protein quality control system that consists of the protein import machinery as well chaperones and proteases termed the UPRmt. The UPRmt monitors intra-organelle proteostasis, and strives to maintain a mito-nuclear balance between nuclear- and mtDNA-derived gene products via retrograde signaling from the organelle to the nucleus. In addition, antioxidant capacity is improved, affording greater protection against oxidative stress. In contrast, chronic disuse conditions produce similar signaling but result in decrements in mitochondrial quality and content. Thus, the interactive cross-talk of the regulatory networks that control organelle turnover during wide variations in muscle use and disuse remain incompletely understood, despite our improving knowledge of the traditional regulators of organelle content and function. This brief review acknowledges existing regulatory networks and summarizes recent discoveries of novel biological pathways involved in determining organelle biogenesis, dynamics, mitophagy, protein quality control and antioxidant capacity, identifying ample protein targets for therapeutic intervention that determine muscle and mitochondrial health.
Collapse
Affiliation(s)
- Mikhaela B Slavin
- Muscle Health Research Centre, School of Kinesiology and Health Science, York University, Toronto, Ontario, Canada.,School of Kinesiology and Health Science, York University, Toronto, ON, Canada
| | - Jonathan M Memme
- Muscle Health Research Centre, School of Kinesiology and Health Science, York University, Toronto, Ontario, Canada.,School of Kinesiology and Health Science, York University, Toronto, ON, Canada
| | - Ashley N Oliveira
- Muscle Health Research Centre, School of Kinesiology and Health Science, York University, Toronto, Ontario, Canada.,School of Kinesiology and Health Science, York University, Toronto, ON, Canada
| | - Neushaw Moradi
- Muscle Health Research Centre, School of Kinesiology and Health Science, York University, Toronto, Ontario, Canada.,School of Kinesiology and Health Science, York University, Toronto, ON, Canada
| | - David A Hood
- Muscle Health Research Centre, School of Kinesiology and Health Science, York University, Toronto, Ontario, Canada.,School of Kinesiology and Health Science, York University, Toronto, ON, Canada
| |
Collapse
|
41
|
McCoin CS, Franczak E, Deng F, Pei D, Ding WX, Thyfault JP. Acute exercise rapidly activates hepatic mitophagic flux. J Appl Physiol (1985) 2022; 132:862-873. [PMID: 35142562 PMCID: PMC8934677 DOI: 10.1152/japplphysiol.00704.2021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 01/25/2022] [Accepted: 02/02/2022] [Indexed: 01/18/2023] Open
Abstract
Exercise is critical for improving metabolic health and putatively maintains or enhances mitochondrial quality control in metabolic tissues. Although previous work has shown that exercise elicits hepatic mitochondrial biogenesis, it is unknown if acute exercise activates hepatic mitophagy, the selective degradation of damaged or low-functioning mitochondria. We tested if an acute bout of treadmill running increased hepatic mitophagic flux both right after and 2-h postexercise in 15- to 24-wk-old C57BL/6J female mice. Acute exercise did not significantly increase markers of autophagic flux, however, mitophagic flux was activated 2-h post-treadmill running as measured by accumulation of both LC3-II and p62 in isolated mitochondria in the presence of leupeptin, an inhibitor of autophagosome degradation. Furthermore, mitochondrial-associated ubiquitin, which recruits the autophagy receptor protein p62, was also significantly increased at 2 h. Further examination via Western blot and proteomics analysis revealed that acute exercise elicits a time-dependent, dynamic activation of mitophagy pathways. Moreover, the results suggest that exercise-induced hepatic mitophagy is likely mediated by both polyubiquitination and receptor-mediated signaling pathways. Overall, we provide evidence that acute exercise activates hepatic mitophagic flux while also revealing specific receptor-mediated proteins by which exercise maintains mitochondrial quality control in the liver.NEW & NOTEWORTHY This study provides evidence that acute exercise activates hepatic mitophagic flux and mitochondrial polyubiquitination while additionally revealing specific receptor-mediated proteins by which exercise maintains mitochondrial quality control in the liver.
Collapse
Affiliation(s)
- Colin S McCoin
- Department of Molecular & Integrative Physiology, University of Kansas Medical Center, Kansas City, Missouri
- Center for Children's Healthy Lifestyles and Nutrition, Children's Mercy Kansas City, Kansas City, Missouri
- Kansas City Veterans Affairs Medical Center, Kansas City, Missouri
| | - Edziu Franczak
- Department of Molecular & Integrative Physiology, University of Kansas Medical Center, Kansas City, Missouri
| | - Fengyan Deng
- Stowers Institute for Medical Research, Kansas City, Missouri
| | - Dong Pei
- Department of Biostatistics and Data Science, University of Kansas Medical Center, Kansas City, Kansas
| | - Wen-Xing Ding
- Department of Pharmacology, Toxicology & Therapeutics, University of Kansas Medical Center, Kansas City, Kansas
| | - John P Thyfault
- Department of Molecular & Integrative Physiology, University of Kansas Medical Center, Kansas City, Missouri
- Division of Endocrinology and Metabolism, Department of Internal Medicine, University of Kansas Medical Center, Kansas City, Kansas
- Center for Children's Healthy Lifestyles and Nutrition, Children's Mercy Kansas City, Kansas City, Missouri
- Kansas City Veterans Affairs Medical Center, Kansas City, Missouri
| |
Collapse
|
42
|
Sligar J, Debruin DA, Saner NJ, Philp AM, Philp A. The importance of mitochondrial quality control for maintaining skeletal muscle function across healthspan. Am J Physiol Cell Physiol 2022; 322:C461-C467. [PMID: 35108118 DOI: 10.1152/ajpcell.00388.2021] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
As the principal energy-producing organelles of the cell, mitochondria support numerous biological processes related to metabolism, growth and regeneration in skeletal muscle. Deterioration in skeletal muscle functional capacity with age is thought to be driven in part by a reduction in skeletal muscle oxidative capacity and reduced fatigue resistance. Underlying this maladaptive response is the development of mitochondrial dysfunction caused by alterations in mitochondrial quality control (MQC), a term encompassing processes of mitochondrial synthesis (biogenesis), remodelling (dynamics) and degradation (mitophagy). Knowledge regarding the role and regulation of MQC in skeletal muscle and the influence of ageing in this process have rapidly advanced in the last decade. Given the emerging link between ageing and MQC, therapeutic approaches to manipulate MQC to prevent mitochondrial dysfuntion during ageing hold tremendous therapeutic potential.
Collapse
Affiliation(s)
- James Sligar
- Mitochondrial Metabolism and Ageing Laboratory, Garvan Institute of Medical Research, Sydney, New South Wales, Australia.,St Vincent's Medical School, UNSW Medicine, UNSW Sydney, Sydney, New South Wales, Australia
| | - Danielle A Debruin
- Australian Institute for Musculoskeletal Science (AIMSS), Victoria University, Sunshine Hospital, St Albans, Australia.,Institute for Health and Sport, Victoria University, Melbourne, Australia
| | - Nicholas J Saner
- Human Integrative Physiology, Baker Heart and Diabetes Institute, Melbourne, Australia
| | - Ashleigh M Philp
- Mitochondrial Metabolism and Ageing Laboratory, Garvan Institute of Medical Research, Sydney, New South Wales, Australia.,St Vincent's Medical School, UNSW Medicine, UNSW Sydney, Sydney, New South Wales, Australia
| | - Andrew Philp
- Mitochondrial Metabolism and Ageing Laboratory, Garvan Institute of Medical Research, Sydney, New South Wales, Australia.,St Vincent's Medical School, UNSW Medicine, UNSW Sydney, Sydney, New South Wales, Australia
| |
Collapse
|
43
|
Memme JM, Oliveira AN, Hood DA. p53 regulates skeletal muscle mitophagy and mitochondrial quality control following denervation-induced muscle disuse. J Biol Chem 2022; 298:101540. [PMID: 34958797 PMCID: PMC8790503 DOI: 10.1016/j.jbc.2021.101540] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 12/10/2021] [Accepted: 12/13/2021] [Indexed: 02/06/2023] Open
Abstract
Persistent inactivity promotes skeletal muscle atrophy, marked by mitochondrial aberrations that affect strength, mobility, and metabolic health leading to the advancement of disease. Mitochondrial quality control (MQC) pathways include biogenesis (synthesis), mitophagy/lysosomal turnover, and the mitochondrial unfolded protein response, which serve to maintain an optimal organelle network. Tumor suppressor p53 has been implicated in regulating muscle mitochondria in response to cellular stress; however, its role in the context of muscle disuse has yet to be explored, and whether p53 is necessary for MQC remains unclear. To address this, we subjected p53 muscle-specific KO (mKO) and WT mice to unilateral denervation. Transcriptomic and pathway analyses revealed dysregulation of pathways pertaining to mitochondrial function, and especially turnover, in mKO muscle following denervation. Protein and mRNA data of the MQC pathways indicated activation of the mitochondrial unfolded protein response and mitophagy-lysosome systems along with reductions in mitochondrial biogenesis and content in WT and mKO tissue following chronic denervation. However, p53 ablation also attenuated the expression of autophagy-mitophagy machinery, reduced autophagic flux, and enhanced lysosomal dysfunction. While similar reductions in mitochondrial biogenesis and content were observed between genotypes, MQC dysregulation exacerbated mitochondrial dysfunction in mKO fibers, evidenced by elevated reactive oxygen species. Moreover, acute experiments indicate that p53 mediates the expression of transcriptional regulators of MQC pathways as early as 1 day following denervation. Together, our data illustrate exacerbated mitochondrial dysregulation with denervation stress in p53 mKO tissue, thus indicating that p53 contributes to organellar maintenance via regulation of MQC pathways during muscle atrophy.
Collapse
Affiliation(s)
- Jonathan M Memme
- Muscle Health Research Centre, School of Kinesiology and Health Science, York University, Toronto, Ontario, Canada
| | - Ashley N Oliveira
- Muscle Health Research Centre, School of Kinesiology and Health Science, York University, Toronto, Ontario, Canada
| | - David A Hood
- Muscle Health Research Centre, School of Kinesiology and Health Science, York University, Toronto, Ontario, Canada.
| |
Collapse
|
44
|
Triolo M, Slavin M, Moradi N, Hood DA. Time‐dependent changes in autophagy, mitophagy and lysosomes in skeletal muscle during denervation‐induced disuse. J Physiol 2022; 600:1683-1701. [DOI: 10.1113/jp282173] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Accepted: 01/13/2022] [Indexed: 12/09/2022] Open
Affiliation(s)
- Matthew Triolo
- Muscle Health Research Centre York University Toronto Ontario M3J 1P3 Canada
- School of Kinesiology and Health Science York University Toronto Ontario M3J 1P3 Canada
| | - Mikhaela Slavin
- Muscle Health Research Centre York University Toronto Ontario M3J 1P3 Canada
- School of Kinesiology and Health Science York University Toronto Ontario M3J 1P3 Canada
| | - Neushaw Moradi
- Muscle Health Research Centre York University Toronto Ontario M3J 1P3 Canada
- School of Kinesiology and Health Science York University Toronto Ontario M3J 1P3 Canada
| | - David A. Hood
- Muscle Health Research Centre York University Toronto Ontario M3J 1P3 Canada
- School of Kinesiology and Health Science York University Toronto Ontario M3J 1P3 Canada
| |
Collapse
|
45
|
Wang Z, Niu Y, Lei B, Yu L, Ke Z, Cao C, Wang R, Li J. Downhill Running Decreases the Acetylation of Tubulins and Impairs Autophagosome Degradation in Rat Skeletal Muscle. Med Sci Sports Exerc 2021; 53:2477-2484. [PMID: 34115728 DOI: 10.1249/mss.0000000000002728] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
PURPOSE This study was designed to probe the effect of downhill running on microtubule acetylation and autophagic flux in rat skeletal muscle. METHODS Sprague-Dawley rats were subjected to an exercise protocol of a 90-min downhill run with a slope of -16° and a speed of 16 m·min-1, and then the soleus was sampled at 0, 12, 24, 48, and 72 h after exercise. Protein expression levels of microtubule-associated protein 1 light chain 3 (LC3), p62/sequestosome 1 (p62), α-tubulin, and acetylated α-tubulin (AcK40 α-tubulin) were detected by Western blotting. Alpha-tubulin was costained with AcK40 α-tubulin or cytoplasmic dynein intermediate chain in a single muscle fiber, and LC3 was costained with lysosomal-associated membrane protein 1 in cryosections. To assess autophagic flux in vivo, colchicine or vehicle was injected intraperitoneally 3 d before the exercise experiment, and the protein levels of LC3 and p62 were measured by Western blotting. RESULTS Downhill running induced a significant increase in the protein levels of LC3-II and p62, whereas the level and proportion of AcK40 α-tubulin were markedly decreased. Furthermore, the amount of dynein on α-tubulin was decreased after downhill running, and autophagosomes accumulated in the middle of myofibrils. Importantly, LC3-II flux was decreased after downhill running compared with that in the control group. CONCLUSIONS A bout of downhill running decreases microtubule acetylation, which may impair dynein recruitment and autophagosome transportation, causing blocked autophagic flux.
Collapse
Affiliation(s)
- Zhen Wang
- School of Sports Science, Beijing Sport University, Beijing, CHINA
| | | | - Bingkai Lei
- School of Sports Science, Beijing Sport University, Beijing, CHINA
| | - Liang Yu
- School of Sports Science, Beijing Sport University, Beijing, CHINA
| | - Zhifei Ke
- School of Sports Science, Beijing Sport University, Beijing, CHINA
| | - Chunxia Cao
- School of Sports Science, Beijing Sport University, Beijing, CHINA
| | - Ruiyuan Wang
- School of Sports Science, Beijing Sport University, Beijing, CHINA
| | - Junping Li
- School of Sports Science, Beijing Sport University, Beijing, CHINA
| |
Collapse
|
46
|
Abstract
PURPOSE OF REVIEW The practice of time-restricted feeding (TRF) has received fervent interest in recent years as a strategy to mitigate obesity and metabolic disease. We sought to review the implications of TRF for skeletal muscle health and function in aging. RECENT FINDINGS TRF has high adherence and can promote body weight loss in older populations. Body weight reductions favor fat mass in the young, however, there is also the potential for undesirable losses in lean mass. There is currently no evidence to support TRF for skeletal muscle function and metabolism in older persons, and only tentative findings in the young. With a narrow eating window of 6-8 h and a prolonged fasting period to minimize daily insulin exposure, TRF may contradict recommended dietary practices for optimizing skeletal muscle anabolism in older people. SUMMARY TRF might represent a promising intervention to address obesity and its associated metabolic diseases, however, at present there is insufficient evidence for optimizing skeletal muscle mass or health in older individuals. Further research is needed to: (1) ascertain the impact of TRF on body composition, skeletal muscle anabolism, and autophagy in aging, and; (2) delineate the potentially myoprotective roles of dietary protein and exercise within the framework of TRF in older persons.
Collapse
Affiliation(s)
- Matthew J Lees
- Faculty of Kinesiology and Physical Education, University of Toronto, Toronto, Ontario, Canada
| | | | | |
Collapse
|
47
|
Redox Signaling and Sarcopenia: Searching for the Primary Suspect. Int J Mol Sci 2021; 22:ijms22169045. [PMID: 34445751 PMCID: PMC8396474 DOI: 10.3390/ijms22169045] [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: 07/26/2021] [Revised: 08/17/2021] [Accepted: 08/19/2021] [Indexed: 12/16/2022] Open
Abstract
Sarcopenia, the age-related decline in muscle mass and function, derives from multiple etiological mechanisms. Accumulative research suggests that reactive oxygen species (ROS) generation plays a critical role in the development of this pathophysiological disorder. In this communication, we review the various signaling pathways that control muscle metabolic and functional integrity such as protein turnover, cell death and regeneration, inflammation, organismic damage, and metabolic functions. Although no single pathway can be identified as the most crucial factor that causes sarcopenia, age-associated dysregulation of redox signaling appears to underlie many deteriorations at physiological, subcellular, and molecular levels. Furthermore, discord of mitochondrial homeostasis with aging affects most observed problems and requires our attention. The search for the primary suspect of the fundamental mechanism for sarcopenia will likely take more intense research for the secret of this health hazard to the elderly to be unlocked.
Collapse
|
48
|
Leduc-Gaudet JP, Hussain SNA, Barreiro E, Gouspillou G. Mitochondrial Dynamics and Mitophagy in Skeletal Muscle Health and Aging. Int J Mol Sci 2021; 22:ijms22158179. [PMID: 34360946 PMCID: PMC8348122 DOI: 10.3390/ijms22158179] [Citation(s) in RCA: 137] [Impact Index Per Article: 34.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 07/23/2021] [Accepted: 07/27/2021] [Indexed: 01/18/2023] Open
Abstract
The maintenance of mitochondrial integrity is critical for muscle health. Mitochondria, indeed, play vital roles in a wide range of cellular processes, including energy supply, Ca2+ homeostasis, retrograde signaling, cell death, and many others. All mitochondria-containing cells, including skeletal muscle cells, dispose of several pathways to maintain mitochondrial health, including mitochondrial biogenesis, mitochondrial-derived vesicles, mitochondrial dynamics (fusion and fission process shaping mitochondrial morphology), and mitophagy—the process in charge of the removal of mitochondria though autophagy. The loss of skeletal muscle mass (atrophy) is a major health problem worldwide, especially in older people. Currently, there is no treatment to counteract the progressive decline in skeletal muscle mass and strength that occurs with aging, a process termed sarcopenia. There is increasing data, including our own, suggesting that accumulation of dysfunctional mitochondria contributes to the development of sarcopenia. Impairments in mitochondrial dynamics and mitophagy were recently proposed to contribute to sarcopenia. This review summarizes the current state of knowledge on the role played by mitochondrial dynamics and mitophagy in skeletal muscle health and in the development of sarcopenia. We also highlight recent studies showing that enhancing mitophagy in skeletal muscle is a promising therapeutic target to prevent or even treat skeletal muscle dysfunction in the elderly.
Collapse
Affiliation(s)
- Jean-Philippe Leduc-Gaudet
- Research Institute of the McGill University Health Centre, Department of Critical Care, Montréal, QC H4A 3J1, Canada; (S.N.A.H.); (G.G.)
- Meakins-Christie Laboratories, Department of Medicine, McGill University, Montréal, QC H4A 3J1, Canada
- Département des Sciences de l’activité Physique, Faculté des Sciences, UQAM, Montréal, QC H2X 1Y4, Canada
- Correspondence: ; Tel.: +1-514-476-6688
| | - Sabah N. A. Hussain
- Research Institute of the McGill University Health Centre, Department of Critical Care, Montréal, QC H4A 3J1, Canada; (S.N.A.H.); (G.G.)
- Meakins-Christie Laboratories, Department of Medicine, McGill University, Montréal, QC H4A 3J1, Canada
| | - Esther Barreiro
- Pulmonology Department-Muscle Wasting & Cachexia in Chronic Respiratory Diseases & Lung Cancer Research Group, IMIM-Hospital del Mar, Parc de Salut Mar, Biomedical Research Park (PRBB), C/Dr. Aiguader, 88, 08003 Barcelona, Spain;
- Centro de Investigación en Red de Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos III (ISCIII), 28029 Madrid, Spain
- Health and Experimental Sciences Department (CEXS), Pompeu Fabra University (UPF), Biomedical Research Park (PRBB), C/Dr. Aiguader, 88, 08003 Barcelona, Spain
| | - Gilles Gouspillou
- Research Institute of the McGill University Health Centre, Department of Critical Care, Montréal, QC H4A 3J1, Canada; (S.N.A.H.); (G.G.)
- Meakins-Christie Laboratories, Department of Medicine, McGill University, Montréal, QC H4A 3J1, Canada
- Département des Sciences de l’activité Physique, Faculté des Sciences, UQAM, Montréal, QC H2X 1Y4, Canada
- Centre de Recherche de l’Institut Universitaire de Gériatrie de Montréal, Montréal, QC H3W 1W5, Canada
| |
Collapse
|
49
|
Cen X, Zhang M, Zhou M, Ye L, Xia H. Mitophagy Regulates Neurodegenerative Diseases. Cells 2021; 10:1876. [PMID: 34440645 PMCID: PMC8392649 DOI: 10.3390/cells10081876] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 07/19/2021] [Accepted: 07/21/2021] [Indexed: 12/12/2022] Open
Abstract
Mitochondria play an essential role in supplying energy for the health and survival of neurons. Mitophagy is a metabolic process that removes dysfunctional or redundant mitochondria. This process preserves mitochondrial health. However, defective mitophagy triggers the accumulation of damaged mitochondria, causing major neurodegenerative disorders. This review introduces molecular mechanisms and signaling pathways behind mitophagy regulation. Furthermore, we focus on the recent advances in understanding the potential role of mitophagy in the pathogenesis of major neurodegenerative diseases (Parkinson's, Alzheimer's, Huntington's, etc.) and aging. The findings will help identify the potential interventions of mitophagy regulation and treatment strategies of neurodegenerative diseases.
Collapse
Affiliation(s)
- Xufeng Cen
- Department of Biochemistry & Research Center of Clinical Pharmacy of The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China; (X.C.); (M.Z.); (M.Z.); (L.Y.)
- Liangzhu Laboratory, Zhejiang University Medical Center, 1369 West Wenyi Road, Hangzhou 311121, China
| | - Manke Zhang
- Department of Biochemistry & Research Center of Clinical Pharmacy of The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China; (X.C.); (M.Z.); (M.Z.); (L.Y.)
| | - Mengxin Zhou
- Department of Biochemistry & Research Center of Clinical Pharmacy of The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China; (X.C.); (M.Z.); (M.Z.); (L.Y.)
| | - Lingzhi Ye
- Department of Biochemistry & Research Center of Clinical Pharmacy of The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China; (X.C.); (M.Z.); (M.Z.); (L.Y.)
- Liangzhu Laboratory, Zhejiang University Medical Center, 1369 West Wenyi Road, Hangzhou 311121, China
| | - Hongguang Xia
- Department of Biochemistry & Research Center of Clinical Pharmacy of The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China; (X.C.); (M.Z.); (M.Z.); (L.Y.)
- Liangzhu Laboratory, Zhejiang University Medical Center, 1369 West Wenyi Road, Hangzhou 311121, China
| |
Collapse
|
50
|
Philp AM, Saner NJ, Lazarou M, Ganley IG, Philp A. The influence of aerobic exercise on mitochondrial quality control in skeletal muscle. J Physiol 2021; 599:3463-3476. [PMID: 33369731 DOI: 10.1113/jp279411] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Accepted: 12/17/2020] [Indexed: 01/12/2023] Open
Abstract
Mitochondria are dynamic organelles, intricately designed to meet cellular energy requirements. To accommodate alterations in energy demand, mitochondria have a high degree of plasticity, changing in response to transient activation of numerous stress-related pathways. This adaptive response is particularly relevant in highly metabolic tissues such as skeletal muscle, where mitochondria support numerous biological processes related to metabolism, growth and regeneration. Aerobic exercise is a potent stimulus for skeletal muscle remodelling, leading to alterations in substrate utilisation, fibre-type composition and performance. Underlying these physiological responses is a change in mitochondrial quality control (MQC), a term encompassing the co-ordination of mitochondrial synthesis (biogenesis), remodelling (dynamics) and degradation (mitophagy) pathways. Understanding of MQC in skeletal muscle and the regulatory role of aerobic exercise of this process are rapidly advancing, as are the molecular techniques allowing the study of MQC in vivo. Given the emerging link between MQC and the onset of numerous non-communicable diseases, understanding the molecular regulation of MQC, and the role of aerobic exercise in this process, will have substantial future impact on therapeutic approaches to manipulate MQC and maintain mitochondrial function across health span.
Collapse
Affiliation(s)
- Ashleigh M Philp
- Healthy Ageing Research Theme, Garvan Institute of Medical Research, 384 Victoria Street, Sydney, New South Wales, 2010, Australia
- St Vincent's Medical School, UNSW Medicine, UNSW Sydney, Sydney, New South Wales, 2010, Australia
| | - Nicholas J Saner
- Sports Cardiology, Baker Heart and Diabetes Institute, Melbourne, Australia
| | - Michael Lazarou
- Biochemistry and Molecular Biology, Monash Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| | - Ian G Ganley
- Medical Research Council Protein Phosphorylation and Ubiquitylation Unit, School of Life Sciences, University of Dundee, Dundee, UK
| | - Andrew Philp
- Healthy Ageing Research Theme, Garvan Institute of Medical Research, 384 Victoria Street, Sydney, New South Wales, 2010, Australia
- St Vincent's Medical School, UNSW Medicine, UNSW Sydney, Sydney, New South Wales, 2010, Australia
| |
Collapse
|