1
|
Lin KH, Hibbert JE, Flynn CG, Lemens JL, Torbey MM, Steinert ND, Flejsierowicz PM, Melka KM, Lindley GT, Lares M, Setaluri V, Wagers AJ, Hornberger TA. Satellite cell-derived TRIM28 is pivotal for mechanical load- and injury-induced myogenesis. EMBO Rep 2024; 25:3812-3841. [PMID: 39143258 PMCID: PMC11387408 DOI: 10.1038/s44319-024-00227-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Revised: 07/19/2024] [Accepted: 07/26/2024] [Indexed: 08/16/2024] Open
Abstract
Satellite cells are skeletal muscle stem cells that contribute to postnatal muscle growth, and they endow skeletal muscle with the ability to regenerate after a severe injury. Here we discover that this myogenic potential of satellite cells requires a protein called tripartite motif-containing 28 (TRIM28). Interestingly, different from the role reported in a previous study based on C2C12 myoblasts, multiple lines of both in vitro and in vivo evidence reveal that the myogenic function of TRIM28 is not dependent on changes in the phosphorylation of its serine 473 residue. Moreover, the functions of TRIM28 are not mediated through the regulation of satellite cell proliferation or differentiation. Instead, our findings indicate that TRIM28 regulates the ability of satellite cells to progress through the process of fusion. Specifically, we discover that TRIM28 controls the expression of a fusogenic protein called myomixer and concomitant fusion pore formation. Collectively, the outcomes of this study expose the framework of a novel regulatory pathway that is essential for myogenesis.
Collapse
Affiliation(s)
- Kuan-Hung Lin
- Department of Comparative Biosciences, University of Wisconsin - Madison, Madison, WI, USA
- School of Veterinary Medicine, University of Wisconsin - Madison, Madison, WI, USA
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
| | - Jamie E Hibbert
- Department of Comparative Biosciences, University of Wisconsin - Madison, Madison, WI, USA
- School of Veterinary Medicine, University of Wisconsin - Madison, Madison, WI, USA
| | - Corey Gk Flynn
- Department of Comparative Biosciences, University of Wisconsin - Madison, Madison, WI, USA
- School of Veterinary Medicine, University of Wisconsin - Madison, Madison, WI, USA
| | - Jake L Lemens
- Department of Comparative Biosciences, University of Wisconsin - Madison, Madison, WI, USA
- School of Veterinary Medicine, University of Wisconsin - Madison, Madison, WI, USA
| | - Melissa M Torbey
- Department of Comparative Biosciences, University of Wisconsin - Madison, Madison, WI, USA
- School of Veterinary Medicine, University of Wisconsin - Madison, Madison, WI, USA
| | - Nathaniel D Steinert
- Department of Comparative Biosciences, University of Wisconsin - Madison, Madison, WI, USA
- School of Veterinary Medicine, University of Wisconsin - Madison, Madison, WI, USA
| | - Philip M Flejsierowicz
- Department of Comparative Biosciences, University of Wisconsin - Madison, Madison, WI, USA
- School of Veterinary Medicine, University of Wisconsin - Madison, Madison, WI, USA
| | - Kiley M Melka
- Department of Comparative Biosciences, University of Wisconsin - Madison, Madison, WI, USA
- School of Veterinary Medicine, University of Wisconsin - Madison, Madison, WI, USA
| | - Garrison T Lindley
- Department of Comparative Biosciences, University of Wisconsin - Madison, Madison, WI, USA
- School of Veterinary Medicine, University of Wisconsin - Madison, Madison, WI, USA
| | - Marcos Lares
- Department of Dermatology, University of Wisconsin - Madison, Madison, WI, USA
| | | | - Amy J Wagers
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
- Harvard Stem Cell Institute, Cambridge, MA, USA
- Joslin Diabetes Center, Boston, MA, USA
| | - Troy A Hornberger
- Department of Comparative Biosciences, University of Wisconsin - Madison, Madison, WI, USA.
- School of Veterinary Medicine, University of Wisconsin - Madison, Madison, WI, USA.
| |
Collapse
|
2
|
Endo Y, Zhu C, Giunta E, Guo C, Koh DJ, Sinha I. The Role of Hypoxia and Hypoxia Signaling in Skeletal Muscle Physiology. Adv Biol (Weinh) 2024; 8:e2200300. [PMID: 37817370 DOI: 10.1002/adbi.202200300] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 04/06/2023] [Indexed: 10/12/2023]
Abstract
Hypoxia and hypoxia signaling play an integral role in regulating skeletal muscle physiology. Environmental hypoxia and tissue hypoxia in muscles cue for their appropriate physiological response and adaptation, and cause an array of cellular and metabolic changes. In addition, muscle stem cells (satellite cells), exist in a hypoxic state, and this intrinsic hypoxic state correlates with their quiescence and stemness. The mechanisms of hypoxia-mediated regulation of satellite cells and myogenesis are yet to be characterized, and their seemingly contradicting effects reported leave their exact roles somewhat perplexing. This review summarizes the recent findings on the effect of hypoxia and hypoxia signaling on the key aspects of muscle physiology, namely, stem cell maintenance and myogenesis with a particular attention given to distinguish the intrinsic versus local hypoxia in an attempt to better understand their respective regulatory roles and how their relationship affects the overall response. This review further describes their mechanistic links and their possible implications on the relevant pathologies and therapeutics.
Collapse
Affiliation(s)
- Yori Endo
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Brigham and Women's Hospital, Harvard University, Boston, MA, 02115, USA
| | - Christina Zhu
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Brigham and Women's Hospital, Harvard University, Boston, MA, 02115, USA
- Texas Tech University Health Sciences Center School of Medicine, Lubbock, TX, 79430, USA
| | - Elena Giunta
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Brigham and Women's Hospital, Harvard University, Boston, MA, 02115, USA
- Ludwig-Maximilians-Universität München, Geschwister-Scholl-Platz 1, 80539, München, Germany
| | - Cynthia Guo
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Brigham and Women's Hospital, Harvard University, Boston, MA, 02115, USA
- Warren Alpert Medical School, Brown University, Providence, RI, 02903, USA
| | - Daniel J Koh
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Brigham and Women's Hospital, Harvard University, Boston, MA, 02115, USA
| | - Indranil Sinha
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Brigham and Women's Hospital, Harvard University, Boston, MA, 02115, USA
| |
Collapse
|
3
|
Going nuclear: Molecular adaptations to exercise mediated by myonuclei. SPORTS MEDICINE AND HEALTH SCIENCE 2022; 5:2-9. [PMID: 36994170 PMCID: PMC10040379 DOI: 10.1016/j.smhs.2022.11.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 11/23/2022] [Accepted: 11/30/2022] [Indexed: 12/13/2022] Open
Abstract
Muscle fibers are multinucleated, and muscle fiber nuclei (myonuclei) are believed to be post-mitotic and are typically situated near the periphery of the myofiber. Due to the unique organization of muscle fibers and their nuclei, the cellular and molecular mechanisms regulating myofiber homeostasis in unstressed and stressed conditions (e.g., exercise) are unique. A key role myonuclei play in regulating muscle during exercise is gene transcription. Only recently have investigators had the capability to identify molecular changes at high resolution exclusively in myonuclei in response to perturbations in vivo. The purpose of this review is to describe how myonuclei modulate their transcriptome, epigenetic status, mobility and shape, and microRNA expression in response to exercise in vivo. Given the relative paucity of high-fidelity information on myonucleus-specific contributions to exercise adaptation, we identify specific gaps in knowledge and provide perspectives on future directions of research.
Collapse
|
4
|
Wang X, Liu F, An Q, Wang W, Cheng Z, Dai Y, Meng Q, Zhang Y. Lactoferrin Deficiency Impairs Proliferation of Satellite Cells via Downregulating the ERK1/2 Signaling Pathway. Int J Mol Sci 2022; 23:ijms23137478. [PMID: 35806481 PMCID: PMC9267821 DOI: 10.3390/ijms23137478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Revised: 06/30/2022] [Accepted: 07/01/2022] [Indexed: 11/23/2022] Open
Abstract
Lactoferrin (Ltf), a naturally active glycoprotein, possesses anti-inflammatory, anti-microbial, anti-tumor, and immunomodulatory activities. Many published studies have indicated that Ltf modulates the proliferation of stem cells. However, the role of Ltf in the proliferation of satellite cells, an important cell type in muscle regeneration, has not yet been reported. Here, by using Ltf systemic knockout mice, we illustrate the role of Ltf in skeletal muscle. Results shows that Ltf deficiency impaired proliferation of satellite cells (SCs) and the regenerative capability of skeletal muscle. Mechanistic studies showed that ERK1/2 phosphorylation was significantly downregulated after Ltf deletion in SCs. Simultaneously, the cell cycle-related proteins cyclin D and CDK4 were significantly downregulated. Intervention with exogenous recombinant lactoferrin (R-Ltf) at a concentration of 1000 μg/mL promoted proliferation of SCs. In addition, intraperitoneal injection of Ltf effectively ameliorated the skeletal muscle of mice injured by 1.2% BaCl2 solution. Our results suggest a protective effect of Ltf in the repair of skeletal muscle damage. Ltf holds promise as a novel therapeutic agent for skeletal muscle injuries.
Collapse
Affiliation(s)
- Xiong Wang
- College of Food Science and Nutritional Engineering, China Agricultural University, Tsing Hua Road No. 17, Haidian District, Beijing 100083, China; (X.W.); (Q.A.); (W.W.); (Z.C.)
| | - Fan Liu
- State Key Laboratory for Agrobiotechnology, College of Biological Sciences, China Agricultural University, Yuanmingyuan West Road No. 2, Haidian District, Beijing 100193, China; (F.L.); (Y.D.); (Q.M.)
| | - Qin An
- College of Food Science and Nutritional Engineering, China Agricultural University, Tsing Hua Road No. 17, Haidian District, Beijing 100083, China; (X.W.); (Q.A.); (W.W.); (Z.C.)
| | - Wenli Wang
- College of Food Science and Nutritional Engineering, China Agricultural University, Tsing Hua Road No. 17, Haidian District, Beijing 100083, China; (X.W.); (Q.A.); (W.W.); (Z.C.)
| | - Zhimei Cheng
- College of Food Science and Nutritional Engineering, China Agricultural University, Tsing Hua Road No. 17, Haidian District, Beijing 100083, China; (X.W.); (Q.A.); (W.W.); (Z.C.)
| | - Yunping Dai
- State Key Laboratory for Agrobiotechnology, College of Biological Sciences, China Agricultural University, Yuanmingyuan West Road No. 2, Haidian District, Beijing 100193, China; (F.L.); (Y.D.); (Q.M.)
| | - Qingyong Meng
- State Key Laboratory for Agrobiotechnology, College of Biological Sciences, China Agricultural University, Yuanmingyuan West Road No. 2, Haidian District, Beijing 100193, China; (F.L.); (Y.D.); (Q.M.)
| | - Yali Zhang
- College of Food Science and Nutritional Engineering, China Agricultural University, Tsing Hua Road No. 17, Haidian District, Beijing 100083, China; (X.W.); (Q.A.); (W.W.); (Z.C.)
- Correspondence: ; Tel.: +86-010-6273-7465
| |
Collapse
|
5
|
Rostami S, Salehizadeh R, Shamloo S, Fayazmilani R. The Effect of Voluntary Physical Activity in an Enriched Environment and Combined Exercise Training on the Satellite Cell Pool in Developing Rats. Front Physiol 2022; 13:899234. [PMID: 35694391 PMCID: PMC9174454 DOI: 10.3389/fphys.2022.899234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Accepted: 04/22/2022] [Indexed: 11/21/2022] Open
Abstract
Aim: Postnatal skeletal muscle growth is strongly associated with a satellite cell pool. Early adolescence might be a crucial period when different exercise training interventions have specific consequence on satellite cells. Pax7 and MyoD have been suggested as the leading indicators of satellite cell activation. Methods: In this study, pre-adolescent male rats (n = 18) were either subjected to an enriched environment that facilitated physical activities or combined training or control for three weeks. The flexor hallucis longus muscle was removed for biochemical and histochemical analysis. Results: Findings demonstrated that exercise trained rats displayed high levels of serum IGF-1 (p <0.05). There was an increase in Pax7 (p <0.05) and MyoD (p <0.001) mRNA expression. A significant increase in the mean fiber area (p <0.01), satellite cell (p <0.001), and myonuclear numbers (p <0.01) were also observed in both intervention groups. Importantly, enriched rats showed lower corticosterone levels (p <0.05) compared to training ones. Regarding performance, trained and enriched rats had significant improvement in forelimb grip strength (p <0.01) and load-carrying capacity (p <0.05). Conclusion: Type of physical exercise is an essential part in changing satellite cells pool. Different and frequent physical activities in an enriched environment can be effective for muscle development.
Collapse
|
6
|
Viggars MR, Wen Y, Peterson CA, Jarvis JC. Automated cross-sectional analysis of trained, severely atrophied and recovering rat skeletal muscles using MyoVision 2.0. J Appl Physiol (1985) 2022; 132:593-610. [PMID: 35050795 DOI: 10.1152/japplphysiol.00491.2021] [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] Open
Abstract
The number of myonuclei within a muscle fiber is an important factor in muscle growth, but its regulation during muscle adaptation is not well understood. We aimed to elucidate the timecourse of myonuclear dynamics during endurance training, loaded and concentric resistance training, and nerve silencing-induced disuse atrophy with subsequent recovery. We modified tibialis anterior muscle activity in free-living rats with electrical stimulation from implantable pulse generators, or with implantable osmotic pumps delivering tetrodotoxin (TTX) to silence the motor nerve without transection. We used the updated, automated software MyoVision to measure fiber type-specific responses in whole tibialis anterior cross-sections (~8000 fibers each). Seven days of continuous low frequency stimulation (CLFS) reduced muscle mass (-12%), increased slower myosin isoforms and reduced IIX/IIB fibers (-32%) and substantially increased myonuclei especially in IIX/IIB fibers (55.5%). High load resistance training (Spillover), produced greater hypertrophy (~16%) in muscle mass and fiber cross-sectional area (CSA) than low load resistance training (concentric, ~6%) and was associated with myonuclear addition in all fiber types (35-46%). TTX-induced nerve silencing resulted in progressive loss in muscle mass, fiber CSA, and myonuclei per fiber cross-section (-50.7%, -53.7%, -40.7%, respectively at 14 days). Myonuclear loss occurred in a fiber type-independent manner, but subsequent recovery during voluntary habitual activity suggested that type IIX/IIB fibers contained more new myonuclei during recovery from severe atrophy. This study demonstrates the power and accuracy provided by the updated MyoVision software and introduces new models for studying myonuclear dynamics in training, detraining, retraining, repeated disuse, and recovery.
Collapse
Affiliation(s)
- Mark Robert Viggars
- Research Institute for Sport & Exercise Sciences, grid.4425.7Liverpool John Moores University, Liverpool, United Kingdom.,Department of Physiology and Functional Genomics, University of Florida, Gainesville, Florida, United States.,Myology Institute, University of Florida, Gainesville, Florida, United States
| | - Yuan Wen
- Department of Physiology, College of Medicine, University of Kentucky, Lexington, Kentucky, United States.,Center for Muscle Biology, University of Kentucky, Lexington, Kentucky, United States.,MyoAnalytics, LLC, Lexington, Kentucky, United States
| | - Charlotte A Peterson
- Department of Physiology, College of Medicine, University of Kentucky, Lexington, Kentucky, United States.,Center for Muscle Biology, University of Kentucky, Lexington, Kentucky, United States
| | - Jonathan C Jarvis
- Research Institute for Sport & Exercise Sciences, grid.4425.7Liverpool John Moores University, Liverpool, United Kingdom
| |
Collapse
|
7
|
Minari ALA, Thomatieli-Santos RV. From skeletal muscle damage and regeneration to the hypertrophy induced by exercise: What is the role of different macrophages subsets? Am J Physiol Regul Integr Comp Physiol 2021; 322:R41-R54. [PMID: 34786967 DOI: 10.1152/ajpregu.00038.2021] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Macrophages are one of the top players when considering immune cells involved with tissue homeostasis. Recently, increasing evidence has demonstrated that these macrophages could also present two major subsets during tissue healing; proliferative macrophages (M1-like), which are responsible for increasing myogenic cell proliferation, and restorative macrophages (M2-like), which are accountable for the end of the mature muscle myogenesis. The participation and characterization of these macrophage subsets is critical during myogenesis, not only to understand the inflammatory role of macrophages during muscle recovery but also to create supportive strategies that can improve mass muscle maintenance. Indeed, most of our knowledge about macrophage subsets comes from skeletal muscle damage protocols, and we still do not know how these subsets can contribute to skeletal muscle adaptation. This narrative review aims to collect and discuss studies demonstrating the involvement of different macrophage subsets during the skeletal muscle damage/regeneration process, showcasing an essential role of these macrophage subsets during muscle adaptation induced by acute and chronic exercise programs.
Collapse
Affiliation(s)
- André Luis Araujo Minari
- Universidade estadual Paulista, Campus Presidente Prudente, Brazil.,Universidade Federal de São Paulo, Psicobiologia, Brazil
| | - Ronaldo V Thomatieli-Santos
- Universidade Federal de São Paulo, Campus Baixada Santista, Brazil.,Universidade Federal de São Paulo, Psicobiologia, Brazil
| |
Collapse
|
8
|
Murach KA, Fry CS, Dupont-Versteegden EE, McCarthy JJ, Peterson CA. Fusion and beyond: Satellite cell contributions to loading-induced skeletal muscle adaptation. FASEB J 2021; 35:e21893. [PMID: 34480776 PMCID: PMC9293230 DOI: 10.1096/fj.202101096r] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2021] [Revised: 08/13/2021] [Accepted: 08/17/2021] [Indexed: 12/14/2022]
Abstract
Satellite cells support adult skeletal muscle fiber adaptations to loading in numerous ways. The fusion of satellite cells, driven by cell-autonomous and/or extrinsic factors, contributes new myonuclei to muscle fibers, associates with load-induced hypertrophy, and may support focal membrane damage repair and long-term myonuclear transcriptional output. Recent studies have also revealed that satellite cells communicate within their niche to mediate muscle remodeling in response to resistance exercise, regulating the activity of numerous cell types through various mechanisms such as secretory signaling and cell-cell contact. Muscular adaptation to resistance and endurance activity can be initiated and sustained for a period of time in the absence of satellite cells, but satellite cell participation is ultimately required to achieve full adaptive potential, be it growth, function, or proprioceptive coordination. While significant progress has been made in understanding the roles of satellite cells in adult muscle over the last few decades, many conclusions have been extrapolated from regeneration studies. This review highlights our current understanding of satellite cell behavior and contributions to adaptation outside of regeneration in adult muscle, as well as the roles of satellite cells beyond fusion and myonuclear accretion, which are gaining broader recognition.
Collapse
Affiliation(s)
- Kevin A Murach
- The Center for Muscle Biology, University of Kentucky, Lexington, Kentucky, USA.,Molecular Muscle Mass Regulation Laboratory, Exercise Science Research Center, Department of Health, Human Performance, and Recreation, University of Arkansas, Fayetteville, Arkansas, USA.,Cell and Molecular Biology Program, University of Arkansas, Fayetteville, Arkansas, USA
| | - Christopher S Fry
- The Center for Muscle Biology, University of Kentucky, Lexington, Kentucky, USA.,Department of Athletic Training and Clinical Nutrition, College of Health Sciences, University of Kentucky, Lexington, Kentucky, USA
| | - Esther E Dupont-Versteegden
- The Center for Muscle Biology, University of Kentucky, Lexington, Kentucky, USA.,Department of Physical Therapy, College of Health Sciences, University of Kentucky, Lexington, Kentucky, USA
| | - John J McCarthy
- The Center for Muscle Biology, University of Kentucky, Lexington, Kentucky, USA.,Department of Physiology, College of Medicine, University of Kentucky, Lexington, Kentucky, USA
| | - Charlotte A Peterson
- The Center for Muscle Biology, University of Kentucky, Lexington, Kentucky, USA.,Department of Physical Therapy, College of Health Sciences, University of Kentucky, Lexington, Kentucky, USA.,Department of Physiology, College of Medicine, University of Kentucky, Lexington, Kentucky, USA
| |
Collapse
|
9
|
Solsona R, Pavlin L, Bernardi H, Sanchez AMJ. Molecular Regulation of Skeletal Muscle Growth and Organelle Biosynthesis: Practical Recommendations for Exercise Training. Int J Mol Sci 2021; 22:2741. [PMID: 33800501 PMCID: PMC7962973 DOI: 10.3390/ijms22052741] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 03/04/2021] [Accepted: 03/04/2021] [Indexed: 12/18/2022] Open
Abstract
The regulation of skeletal muscle mass and organelle homeostasis is dependent on the capacity of cells to produce proteins and to recycle cytosolic portions. In this investigation, the mechanisms involved in skeletal muscle mass regulation-especially those associated with proteosynthesis and with the production of new organelles-are presented. Thus, the critical roles of mammalian/mechanistic target of rapamycin complex 1 (mTORC1) pathway and its regulators are reviewed. In addition, the importance of ribosome biogenesis, satellite cells involvement, myonuclear accretion, and some major epigenetic modifications related to protein synthesis are discussed. Furthermore, several studies conducted on the topic of exercise training have recognized the central role of both endurance and resistance exercise to reorganize sarcomeric proteins and to improve the capacity of cells to build efficient organelles. The molecular mechanisms underlying these adaptations to exercise training are presented throughout this review and practical recommendations for exercise prescription are provided. A better understanding of the aforementioned cellular pathways is essential for both healthy and sick people to avoid inefficient prescriptions and to improve muscle function with emergent strategies (e.g., hypoxic training). Finally, current limitations in the literature and further perspectives, notably on epigenetic mechanisms, are provided to encourage additional investigations on this topic.
Collapse
Affiliation(s)
- Robert Solsona
- Laboratoire Interdisciplinaire Performance Santé Environnement de Montagne (LIPSEM), Faculty of Sports Sciences, University of Perpignan Via Domitia, UR 4640, 7 Avenue Pierre de Coubertin, 66120 Font-Romeu, France;
| | - Laura Pavlin
- DMEM, University of Montpellier, INRAE UMR866, 2 Place Pierre Viala, 34060 Montpellier, France; (L.P.); (H.B.)
| | - Henri Bernardi
- DMEM, University of Montpellier, INRAE UMR866, 2 Place Pierre Viala, 34060 Montpellier, France; (L.P.); (H.B.)
| | - Anthony MJ Sanchez
- Laboratoire Interdisciplinaire Performance Santé Environnement de Montagne (LIPSEM), Faculty of Sports Sciences, University of Perpignan Via Domitia, UR 4640, 7 Avenue Pierre de Coubertin, 66120 Font-Romeu, France;
| |
Collapse
|
10
|
Murach KA, Mobley CB, Zdunek CJ, Frick KK, Jones SR, McCarthy JJ, Peterson CA, Dungan CM. Muscle memory: myonuclear accretion, maintenance, morphology, and miRNA levels with training and detraining in adult mice. J Cachexia Sarcopenia Muscle 2020; 11:1705-1722. [PMID: 32881361 PMCID: PMC7749570 DOI: 10.1002/jcsm.12617] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Revised: 08/06/2020] [Accepted: 08/10/2020] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND In the context of mass regulation, 'muscle memory' can be defined as long-lasting cellular adaptations to hypertrophic exercise training that persist during detraining-induced atrophy and may facilitate future adaptation. The cellular basis of muscle memory is not clearly defined but may be related to myonuclear number and/or epigenetic changes within muscle fibres. METHODS Utilizing progressive weighted wheel running (PoWeR), a novel murine exercise training model, we explored myonuclear dynamics and skeletal muscle miRNA levels with training and detraining utilizing immunohistochemistry, single fibre myonuclear analysis, and quantitative analysis of miRNAs. We also used a genetically inducible mouse model of fluorescent myonuclear labelling to study myonuclear adaptations early during exercise. RESULTS In the soleus, oxidative type 2a fibres were larger after 2 months of PoWeR (P = 0.02), but muscle fibre size and myonuclear number did not return to untrained levels after 6 months of detraining. Soleus type 1 fibres were not larger after PoWeR but had significantly more myonuclei, as well as central nuclei (P < 0.0001), the latter from satellite cell-derived or resident myonuclei, appearing early during training and remaining with detraining. In the gastrocnemius muscle, oxidative type 2a fibres of the deep region were larger and contained more myonuclei after PoWeR (P < 0.003), both of which returned to untrained levels after detraining. In the gastrocnemius and plantaris, two muscles where myonuclear number was comparable with untrained levels after 6 months of detraining, myonuclei were significantly elongated with detraining (P < 0.0001). In the gastrocnemius, miR-1 was lower with training and remained lower after detraining (P < 0.002). CONCLUSIONS This study found that (i) myonuclei gained during hypertrophy are lost with detraining across muscles, even in oxidative fibres; (ii) complete reversal of muscle adaptations, including myonuclear number, to untrained levels occurs within 6 months in the plantaris and gastrocnemius; (iii) the murine soleus is resistant to detraining; (iv) myonuclear accretion occurs early with wheel running and can be uncoupled from muscle fibre hypertrophy; (v) resident (non-satellite cell-derived) myonuclei can adopt a central location; (vi) myonuclei change shape with training and detraining; and (vii) miR-1 levels may reflect a memory of previous adaptation that facilitates future growth.
Collapse
Affiliation(s)
- Kevin A. Murach
- Department of Physical TherapyUniversity of KentuckyLexingtonKYUSA
- Center for Muscle BiologyUniversity of KentuckyLexingtonKYUSA
| | - C. Brooks Mobley
- Department of PhysiologyUniversity of KentuckyLexingtonKYUSA
- Center for Muscle BiologyUniversity of KentuckyLexingtonKYUSA
| | | | | | | | - John J. McCarthy
- Department of PhysiologyUniversity of KentuckyLexingtonKYUSA
- Center for Muscle BiologyUniversity of KentuckyLexingtonKYUSA
| | - Charlotte A. Peterson
- Department of Physical TherapyUniversity of KentuckyLexingtonKYUSA
- Center for Muscle BiologyUniversity of KentuckyLexingtonKYUSA
| | - Cory M. Dungan
- Department of Physical TherapyUniversity of KentuckyLexingtonKYUSA
- Center for Muscle BiologyUniversity of KentuckyLexingtonKYUSA
- Sanders‐Brown Center on AgingUniversity of KentuckyLexingtonKYUSA
| |
Collapse
|
11
|
Xing HY, Liu N, Zhou MW. Satellite cell proliferation and myofiber cross-section area increase after electrical stimulation following sciatic nerve crush injury in rats. Chin Med J (Engl) 2020; 133:1952-1960. [PMID: 32826459 PMCID: PMC7462209 DOI: 10.1097/cm9.0000000000000822] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Indexed: 11/26/2022] Open
Abstract
BACKGROUND Electrical stimulation has been recommended as an effective therapy to prevent muscle atrophy after nerve injury. However, the effect of electrical stimulation on the proliferation of satellite cells in denervated muscles has not yet been fully elucidated. This study was aimed to evaluate the changes in satellite cell proliferation after electrical stimulation in nerve injury and to determine whether these changes are related to the restoration of myofiber cross-section area (CSA). METHODS Sciatic nerve crush injury was performed in 48 male Sprague-Dawley rats. In half (24/48) of the rats, the gastrocnemius was electrically stimulated transcutaneously on a daily basis after injury, while the other half were not stimulated. Another group of 24 male Sprague-Dawley rats were used as sham operation controls without injury or stimulation. The rats were euthanized 2, 4, and 6 weeks later. After 5-bromo-2'-deoxyuridine (BrdU) labeling, the gastrocnemia were harvested for the detection of paired box protein 7 (Pax7), BrdU, myofiber CSA, and myonuclei number per fiber. All data were analyzed using two-way analysis of variance and Bonferroni post-hoc test. RESULTS The percentages of Pax7-positive nuclei (10.81 ± 0.56%) and BrdU-positive nuclei (34.29 ± 3.87%) in stimulated muscles were significantly higher compared to those in non-stimulated muscles (2.58 ± 0.33% and 1.30 ± 0.09%, respectively, Bonferroni t = 15.91 and 18.14, P < 0.05). The numbers of myonuclei per fiber (2.19 ± 0.24) and myofiber CSA (1906.86 ± 116.51 μm) were also increased in the stimulated muscles (Bonferroni t = 3.57 and 2.73, P < 0.05), and both were positively correlated with the Pax7-positive satellite cell content (R = 0.52 and 0.60, P < 0.01). There was no significant difference in the ratio of myofiber CSA/myonuclei number per fiber among the three groups. CONCLUSIONS Our results indicate that satellite cell proliferation is promoted by electrical stimulation after nerve injury, which may be correlated with an increase in myonuclei number and myofiber CSA.
Collapse
Affiliation(s)
- Hua-Yi Xing
- Department of Rehabilitation Medicine, Peking University Third Hospital, Beijing 100191, China
| | | | | |
Collapse
|
12
|
Fukada SI, Akimoto T, Sotiropoulos A. Role of damage and management in muscle hypertrophy: Different behaviors of muscle stem cells in regeneration and hypertrophy. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2020; 1867:118742. [PMID: 32417255 DOI: 10.1016/j.bbamcr.2020.118742] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 05/07/2020] [Accepted: 05/12/2020] [Indexed: 12/12/2022]
Abstract
Skeletal muscle is a dynamic tissue with two unique abilities; one is its excellent regenerative ability, due to the activity of skeletal muscle-resident stem cells named muscle satellite cells (MuSCs); and the other is the adaptation of myofiber size in response to external stimulation, intrinsic factors, or physical activity, which is known as plasticity. Low physical activity and some disease conditions lead to the reduction of myofiber size, called atrophy, whereas hypertrophy refers to the increase in myofiber size induced by high physical activity or anabolic hormones/drugs. MuSCs are essential for generating new myofibers during regeneration and the increase in new myonuclei during hypertrophy; however, there has been little investigation of the molecular mechanisms underlying MuSC activation, proliferation, and differentiation during hypertrophy compared to those of regeneration. One reason is that 'degenerative damage' to myofibers during muscle injury or upon hypertrophy (especially overloaded muscle) is believed to trigger similar activation/proliferation of MuSCs. However, evidence suggests that degenerative damage of myofibers is not necessary for MuSC activation/proliferation during hypertrophy. When considering MuSC-based therapy for atrophy, including sarcopenia, it will be indispensable to elucidate MuSC behaviors in muscles that exhibit non-degenerative damage, because degenerated myofibers are not present in the atrophied muscles. In this review, we summarize recent findings concerning the relationship between MuSCs and hypertrophy, and discuss what remains to be discovered to inform the development and application of relevant treatments for muscle atrophy.
Collapse
Affiliation(s)
- So-Ichiro Fukada
- Project for Muscle Stem Cell Biology, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan.
| | | | - Athanassia Sotiropoulos
- Institut National de la Santé et de la Recherche Médicale U1016, Institut Cochin, Paris, France
| |
Collapse
|
13
|
Roussel MP, Morin M, Girardin M, Fortin AM, Leone M, Mathieu J, Gagnon C, Duchesne E. Training program-induced skeletal muscle adaptations in two men with myotonic dystrophy type 1. BMC Res Notes 2019; 12:526. [PMID: 31429798 PMCID: PMC6700834 DOI: 10.1186/s13104-019-4554-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Accepted: 08/10/2019] [Indexed: 01/03/2023] Open
Abstract
Objective The purpose of this side product of another unpublished research project, was to address the effects of a training program on skeletal muscle adaptations of people with myotonic dystrophy type 1 (DM1), under a multifaceted perspective. The objective of this study was to look at training induced muscular adaptations by evaluating changes in muscle strength, myofiber cross-sectional area (CSA), proportion of myofiber types and with indirect markers of muscle growth [proportion of centrally nucleated fibers (CNF) and density of neutrophils and macrophages]. Two men with DM1 underwent a 12-week strength/endurance training program (18 sessions). Two muscle biopsies were obtained pre- and post-training program. Results Muscular adaptations occurred only in Patient 1, who attended 72% of the training sessions compared to 39% for Patient 2. These adaptations included increase in the CSA of type I and II myofibers and changes in their proportion. No changes were observed in the percentage of CNF, infiltration of neutrophils and macrophages and muscle strength. These results illustrate the capacity of skeletal muscle cells to undergo adaptations linked to muscle growth in DM1 patients. Also, these adaptations seem to be dependent on the attendance. Trial registration Clinicaltrials.gov NCT04001920 retrospectively registered on June 26th, 2019
Collapse
Affiliation(s)
- Marie-Pier Roussel
- Département des sciences fondamentales, Université du Québec à Chicoutimi, Saguenay, QC, Canada.,Groupe de recherche interdisciplinaire sur les maladies neuromusculaires, Centre intégré universitaire de santé et de services sociaux du Saguenay-Lac-St-Jean, Installations de Jonquière, Saguenay, QC, Canada.,Centre de recherche-Hôpital Charles-Le Moyne - Saguenay-Lac-Saint-Jean sur les innovations en santé, Saguenay, QC, Canada
| | - Marika Morin
- Département des sciences de la santé, Université du Québec à Chicoutimi, Saguenay, QC, Canada
| | - Mélina Girardin
- Département des sciences de la santé, Université du Québec à Chicoutimi, Saguenay, QC, Canada
| | - Anne-Marie Fortin
- Groupe de recherche interdisciplinaire sur les maladies neuromusculaires, Centre intégré universitaire de santé et de services sociaux du Saguenay-Lac-St-Jean, Installations de Jonquière, Saguenay, QC, Canada
| | - Mario Leone
- Groupe de recherche interdisciplinaire sur les maladies neuromusculaires, Centre intégré universitaire de santé et de services sociaux du Saguenay-Lac-St-Jean, Installations de Jonquière, Saguenay, QC, Canada.,Département des sciences de la santé, Université du Québec à Chicoutimi, Saguenay, QC, Canada
| | - Jean Mathieu
- Groupe de recherche interdisciplinaire sur les maladies neuromusculaires, Centre intégré universitaire de santé et de services sociaux du Saguenay-Lac-St-Jean, Installations de Jonquière, Saguenay, QC, Canada.,Centre de recherche-Hôpital Charles-Le Moyne - Saguenay-Lac-Saint-Jean sur les innovations en santé, Saguenay, QC, Canada.,Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Cynthia Gagnon
- Groupe de recherche interdisciplinaire sur les maladies neuromusculaires, Centre intégré universitaire de santé et de services sociaux du Saguenay-Lac-St-Jean, Installations de Jonquière, Saguenay, QC, Canada.,Centre de recherche-Hôpital Charles-Le Moyne - Saguenay-Lac-Saint-Jean sur les innovations en santé, Saguenay, QC, Canada.,Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Elise Duchesne
- Groupe de recherche interdisciplinaire sur les maladies neuromusculaires, Centre intégré universitaire de santé et de services sociaux du Saguenay-Lac-St-Jean, Installations de Jonquière, Saguenay, QC, Canada. .,Centre de recherche-Hôpital Charles-Le Moyne - Saguenay-Lac-Saint-Jean sur les innovations en santé, Saguenay, QC, Canada. .,Département des sciences de la santé, Université du Québec à Chicoutimi, Saguenay, QC, Canada. .,Unité d'enseignement en physiothérapie, Département des sciences de la santé, Université du Québec à Chicoutimi, 555, boulevard de l'Université, Saguenay, G7H 2B1, Quebec, Canada.
| |
Collapse
|
14
|
Wang F, Zhang QB, Zhou Y, Chen S, Huang PP, Liu Y, Xu YH. The mechanisms and treatments of muscular pathological changes in immobilization-induced joint contracture: A literature review. Chin J Traumatol 2019; 22:93-98. [PMID: 30928194 PMCID: PMC6488749 DOI: 10.1016/j.cjtee.2019.02.001] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Revised: 10/15/2018] [Accepted: 01/26/2019] [Indexed: 02/04/2023] Open
Abstract
The clinical treatment of joint contracture due to immobilization remains difficult. The pathological changes of muscle tissue caused by immobilization-induced joint contracture include disuse skeletal muscle atrophy and skeletal muscle tissue fibrosis. The proteolytic pathways involved in disuse muscle atrophy include the ubiquitin-proteasome-dependent pathway, caspase system pathway, matrix metalloproteinase pathway, Ca2+-dependent pathway and autophagy-lysosomal pathway. The important biological processes involved in skeletal muscle fibrosis include intermuscular connective tissue thickening caused by transforming growth factor-β1 and an anaerobic environment within the skeletal muscle leading to the induction of hypoxia-inducible factor-1α. This article reviews the progress made in understanding the pathological processes involved in immobilization-induced muscle contracture and the currently available treatments. Understanding the mechanisms involved in immobilization-induced contracture of muscle tissue should facilitate the development of more effective treatment measures for the different mechanisms in the future.
Collapse
Affiliation(s)
- Feng Wang
- Department of Rehabilitation Medicine, The Second Hospital of Anhui Medical University, Hefei, 230601, China
| | - Quan-Bing Zhang
- Department of Rehabilitation Medicine, The Second Hospital of Anhui Medical University, Hefei, 230601, China
| | - Yun Zhou
- Department of Rehabilitation Medicine, The Second Hospital of Anhui Medical University, Hefei, 230601, China,Corresponding author.
| | - Shuang Chen
- Department of Rehabilitation Medicine, The Second Hospital of Anhui Medical University, Hefei, 230601, China
| | - Peng-Peng Huang
- Department of Rehabilitation Medicine, The Second Hospital of Anhui Medical University, Hefei, 230601, China
| | - Yi Liu
- Department of Rehabilitation Medicine, The Second Hospital of Anhui Medical University, Hefei, 230601, China
| | - Yuan-Hong Xu
- Department of Clinical Laboratory, The First Hospital of Anhui Medical University, Hefei, 230601, China
| |
Collapse
|
15
|
Formicola L, Pannérec A, Correra RM, Gayraud-Morel B, Ollitrault D, Besson V, Tajbakhsh S, Lachey J, Seehra JS, Marazzi G, Sassoon DA. Inhibition of the Activin Receptor Type-2B Pathway Restores Regenerative Capacity in Satellite Cell-Depleted Skeletal Muscle. Front Physiol 2018; 9:515. [PMID: 29881353 PMCID: PMC5978452 DOI: 10.3389/fphys.2018.00515] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2018] [Accepted: 04/20/2018] [Indexed: 12/11/2022] Open
Abstract
Degenerative myopathies typically display a decline in satellite cells coupled with a replacement of muscle fibers by fat and fibrosis. During this pathological remodeling, satellite cells are present at lower numbers and do not display a proper regenerative function. Whether a decline in satellite cells directly contributes to disease progression or is a secondary result is unknown. In order to dissect these processes, we used a genetic model to reduce the satellite cell population by ~70–80% which leads to a nearly complete loss of regenerative potential. We observe that while no overt tissue damage is observed following satellite cell depletion, muscle fibers atrophy accompanied by changes in the stem cell niche cellular composition. Treatment of these mice with an Activin receptor type-2B (AcvR2B) pathway blocker reverses muscle fiber atrophy as expected, but also restores regenerative potential of the remaining satellite cells. These findings demonstrate that in addition to controlling fiber size, the AcvR2B pathway acts to regulate the muscle stem cell niche providing a more favorable environment for muscle regeneration.
Collapse
Affiliation(s)
- Luigi Formicola
- UMR S 1166 French National Institute of Health and Medical Research, France and the Institute of Cardiometabolism and Nutrition, Stem Cells and Regenerative Medicine, University of Pierre and Marie Curie Paris VI, Paris, France
| | - Alice Pannérec
- UMR S 1166 French National Institute of Health and Medical Research, France and the Institute of Cardiometabolism and Nutrition, Stem Cells and Regenerative Medicine, University of Pierre and Marie Curie Paris VI, Paris, France
| | - Rosa Maria Correra
- UMR S 1166 French National Institute of Health and Medical Research, France and the Institute of Cardiometabolism and Nutrition, Stem Cells and Regenerative Medicine, University of Pierre and Marie Curie Paris VI, Paris, France
| | - Barbara Gayraud-Morel
- Centre National de la Recherche Scientifique URA 2578, Institut Pasteur, Stem Cells and Development, Paris, France
| | - David Ollitrault
- UMR S 1166 French National Institute of Health and Medical Research, France and the Institute of Cardiometabolism and Nutrition, Stem Cells and Regenerative Medicine, University of Pierre and Marie Curie Paris VI, Paris, France
| | - Vanessa Besson
- UMR S 1166 French National Institute of Health and Medical Research, France and the Institute of Cardiometabolism and Nutrition, Stem Cells and Regenerative Medicine, University of Pierre and Marie Curie Paris VI, Paris, France
| | - Shahragim Tajbakhsh
- Centre National de la Recherche Scientifique URA 2578, Institut Pasteur, Stem Cells and Development, Paris, France
| | - Jennifer Lachey
- Acceleron Pharma, Cambridge, MA, United States.,Ember Therapeutics, Watertown, MA, United States
| | - Jasbir S Seehra
- Acceleron Pharma, Cambridge, MA, United States.,Ember Therapeutics, Watertown, MA, United States
| | - Giovanna Marazzi
- UMR S 1166 French National Institute of Health and Medical Research, France and the Institute of Cardiometabolism and Nutrition, Stem Cells and Regenerative Medicine, University of Pierre and Marie Curie Paris VI, Paris, France
| | - David A Sassoon
- UMR S 1166 French National Institute of Health and Medical Research, France and the Institute of Cardiometabolism and Nutrition, Stem Cells and Regenerative Medicine, University of Pierre and Marie Curie Paris VI, Paris, France
| |
Collapse
|
16
|
Diabetes-Induced Dysfunction of Mitochondria and Stem Cells in Skeletal Muscle and the Nervous System. Int J Mol Sci 2017; 18:ijms18102147. [PMID: 29036909 PMCID: PMC5666829 DOI: 10.3390/ijms18102147] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Accepted: 10/11/2017] [Indexed: 12/21/2022] Open
Abstract
Diabetes mellitus is one of the most common metabolic diseases spread all over the world, which results in hyperglycemia caused by the breakdown of insulin secretion or insulin action or both. Diabetes has been reported to disrupt the functions and dynamics of mitochondria, which play a fundamental role in regulating metabolic pathways and are crucial to maintain appropriate energy balance. Similar to mitochondria, the functions and the abilities of stem cells are attenuated under diabetic condition in several tissues. In recent years, several studies have suggested that the regulation of mitochondria functions and dynamics is critical for the precise differentiation of stem cells. Importantly, physical exercise is very useful for preventing the diabetic alteration by improving the functions of both mitochondria and stem cells. In the present review, we provide an overview of the diabetic alterations of mitochondria and stem cells and the preventive effects of physical exercise on diabetes, focused on skeletal muscle and the nervous system. We propose physical exercise as a countermeasure for the dysfunction of mitochondria and stem cells in several target tissues under diabetes complication and to improve the physiological function of patients with diabetes, resulting in their quality of life being maintained.
Collapse
|
17
|
Alharby E, Albalawi A, Nasir A, Alhijji S, Mahmood A, Ramzan K, Abdusamad F, Aljohani A, Abdelsalam O, Eldardear A, Basit S. A homozygous potentially pathogenic variant in thePAXBP1gene in a large family with global developmental delay and myopathic hypotonia. Clin Genet 2017; 92:579-586. [DOI: 10.1111/cge.13051] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Revised: 04/26/2017] [Accepted: 05/05/2017] [Indexed: 11/30/2022]
Affiliation(s)
- E. Alharby
- Center for Genetics and Inherited Diseases; Taibah University; Almadinah Almunawwarah Saudi Arabia
| | - A.M. Albalawi
- Center for Genetics and Inherited Diseases; Taibah University; Almadinah Almunawwarah Saudi Arabia
| | - A. Nasir
- Synthetic Protein Engineering Laboratory (SPEL); Ajou University; Suwon Korea
| | - S.A. Alhijji
- Paediatric Neurology Department; King Abdullah Medical City, Madinah Maternity and Children Hospital; Almadinah Almunawwarah Saudi Arabia
| | - A. Mahmood
- Stem Cells Unit, Department of Anatomy; King Khalid University Hospital, King Saud University; Riyadh Saudi Arabia
| | - K. Ramzan
- Department of Genetics, Research Centre; King Faisal Specialist Hospital and Research Centre; Riyadh Saudi Arabia
| | - F. Abdusamad
- Center for Genetics and Inherited Diseases; Taibah University; Almadinah Almunawwarah Saudi Arabia
| | - A. Aljohani
- College of Applied Medical Sciences; Taibah University; Almadinah Almunawwarah Saudi Arabia
| | | | - A. Eldardear
- College of Medicine; Taibah University; Almadinah Almunawwarah Saudi Arabia
| | - S. Basit
- Center for Genetics and Inherited Diseases; Taibah University; Almadinah Almunawwarah Saudi Arabia
| |
Collapse
|
18
|
Non-linear actions of physiological agents: Finite disarrangements elicit fitness benefits. Redox Biol 2017; 13:235-243. [PMID: 28595161 PMCID: PMC5460745 DOI: 10.1016/j.redox.2017.05.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Accepted: 05/11/2017] [Indexed: 12/16/2022] Open
Abstract
Finite disarrangements of important (vital) physiological agents and nutrients can induce plethora of beneficial effects, exceeding mere attenuation of the specific stress. Such response to disrupted homeostasis appears to be universally conserved among species. The underlying mechanism of improved fitness and longevity, when physiological agents act outside their normal range is similar to hormesis, a phenomenon whereby toxins elicit beneficial effects at low doses. Due to similarity with such non-linear response to toxins described with J-shaped curve, we have coined a new term “mirror J-shaped curves” for non-linear response to finite disarrangement of physiological agents. Examples from the clinical trials and basic research are provided, along with the unifying mechanisms that tie classical non-linear response to toxins with the non-linear response to physiological agents (glucose, oxygen, osmolarity, thermal energy, calcium, body mass, calorie intake and exercise). Reactive oxygen species and cytosolic calcium seem to be common triggers of signaling pathways that result in these beneficial effects. Awareness of such phenomena and exploring underlying mechanisms can help physicians in their everyday practice. It can also benefit researchers when designing studies and interpreting growing number of scientific data showing non-linear responses to physiological agents.
Collapse
|
19
|
Roles of Peroxisome Proliferator-Activated Receptor β/δ in skeletal muscle physiology. Biochimie 2016; 136:42-48. [PMID: 27916646 DOI: 10.1016/j.biochi.2016.11.010] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Accepted: 11/21/2016] [Indexed: 02/07/2023]
Abstract
More than two decades of studying Peroxisome Proliferator-Activated Receptors (PPARs) has led to an understanding of their implications in various physiological processes that are key for health and disease. All three PPAR isotypes, PPARα, PPARβ/δ, and PPARγ, are activated by a variety of molecules, including fatty acids, eicosanoids and phospholipids, and regulate a spectrum of genes involved in development, lipid and carbohydrate metabolism, inflammation, and proliferation and differentiation of many cell types in different tissues. The hypolipidemic and antidiabetic functions of PPARα and PPARγ in response to fibrate and thiazolidinedione treatment, respectively, are well documented. However, until more recently the functions of PPARβ/δ were less well defined, but are now becoming more recognized in fatty acid metabolism, energy expenditure, and tissue repair. Skeletal muscle is an active metabolic organ with high plasticity for adaptive responses to varying conditions such as fasting or physical exercise. It is the major site of energy expenditure resulting from lipid and glucose catabolism. Here, we review the multifaceted roles of PPARβ/δ in skeletal muscle physiology.
Collapse
|
20
|
Abreu P, Mendes SVD, Ceccatto VM, Hirabara SM. Satellite cell activation induced by aerobic muscle adaptation in response to endurance exercise in humans and rodents. Life Sci 2016; 170:33-40. [PMID: 27888112 DOI: 10.1016/j.lfs.2016.11.016] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Revised: 10/31/2016] [Accepted: 11/21/2016] [Indexed: 11/15/2022]
Abstract
Although the requirement of satellite cells activation and expansion following injury, mechanical load or growth stimulus provoked by resistance exercise has been well established, their function in response to aerobic exercise adaptation remains unclear. A clear relationship between satellite cell expansion in fiber-type specific myosin heavy chain and aerobic performance has been related, independent of myonuclear accretion or muscle growth. However, the trigger for this activation process is not fully understood yet and it seems to be a multi-faceted and well-orchestrated process. Emerging in vitro studies suggest a role for metabolic pathways and oxygen availability for satellite cell activation, modulating the self-renewal potential and cell fate control. The goal of this review is to describe and discuss the current knowledge about the satellite cell activation and expansion in response to aerobic exercise adaptation in human and rodent models. Additionally, findings about the in vitro metabolic control, which seems be involved in the satellite cell activation and cell fate control, are presented and discussed.
Collapse
Affiliation(s)
- Phablo Abreu
- Institute of Biomedical Sciences, University of Sao Paulo, SP, Brazil; Institute of Biomedical Sciences, State University of Ceará, CE, Brazil.
| | | | | | - Sandro Massao Hirabara
- Institute of Biomedical Sciences, University of Sao Paulo, SP, Brazil; Institute of Physical Activity Sciences and Sports, Cruzeiro do Sul University, Sao Paulo, SP, Brazil
| |
Collapse
|
21
|
Itoh Y, Murakami T, Mori T, Agata N, Kimura N, Inoue-Miyazu M, Hayakawa K, Hirano T, Sokabe M, Kawakami K. Training at non-damaging intensities facilitates recovery from muscle atrophy. Muscle Nerve 2016; 55:243-253. [PMID: 27301985 DOI: 10.1002/mus.25218] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/09/2016] [Indexed: 11/07/2022]
Abstract
INTRODUCTION Resistance training promotes recovery from muscle atrophy, but optimum training programs have not been established. We aimed to determine the optimum training intensity for muscle atrophy. METHODS Mice recovering from atrophied muscles after 2 weeks of tail suspension underwent repeated isometric training with varying joint torques 50 times per day. RESULTS Muscle recovery assessed by maximal isometric contraction and myofiber cross-sectional areas (CSAs) were facilitated at 40% and 60% maximum contraction strength (MC), but at not at 10% and 90% MC. At 60% and 90% MC, damaged and contained smaller diameter fibers were observed. Activation of myogenic satellite cells and a marked increase in myonuclei were observed at 40%, 60%, and 90% MC. CONCLUSIONS The increases in myofiber CSAs were likely caused by increased myonuclei formed through fusion of resistance-induced myofibers with myogenic satellite cells. These data indicate that resistance training without muscle damage facilitates efficient recovery from atrophy. Muscle Nerve 55: 243-253, 2017.
Collapse
Affiliation(s)
- Yuta Itoh
- Physical and Occupational Therapy Program, Nagoya University Graduate School of Medicine, Nagoya, Japan
- Faculty of Rehabilitation Science, Nagoya Gakuin University, Seto, Japan
| | - Taro Murakami
- Faculty of Wellness, Shigakkan University, Ohbu, Japan
| | - Tomohiro Mori
- Physical and Occupational Therapy Program, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Nobuhide Agata
- Physical and Occupational Therapy Program, Nagoya University Graduate School of Medicine, Nagoya, Japan
- Faculty of Health and Medical Sciences, Tokoha University, Hamamatsu, Japan
| | - Nahoko Kimura
- Aiche Medical College for Physical and Occupational Therapy, Kiyosu, Japan
| | | | - Kimihide Hayakawa
- Mechanobiology Laboratory, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Takayuki Hirano
- Faculty of Rehabilitation Science, Nagoya Gakuin University, Seto, Japan
| | - Masahiro Sokabe
- Mechanobiology Laboratory, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Keisuke Kawakami
- Physical and Occupational Therapy Program, Nagoya University Graduate School of Medicine, Nagoya, Japan
- Faculty of Welfare and Health Sciences, Oita University, Dannoharu 700, Oita City, 870-1192, Japan
| |
Collapse
|
22
|
Bazgir B, Fathi R, Rezazadeh Valojerdi M, Mozdziak P, Asgari A. Satellite Cells Contribution to Exercise Mediated Muscle Hypertrophy and Repair. CELL JOURNAL 2016; 18:473-484. [PMID: 28042532 PMCID: PMC5086326 DOI: 10.22074/cellj.2016.4714] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/05/2016] [Accepted: 05/26/2016] [Indexed: 12/20/2022]
Abstract
Satellite cells (SCs) are the most abundant skeletal muscle stem cells. They are widely recognized for their contributions to maintenance of muscle mass, regeneration and hypertrophy during the human life span. These cells are good candidates for cell therapy due to their self-renewal capabilities and presence in an undifferentiated form. Presently, a significant gap exists between our knowledge of SCs behavior and their application as a means for human skeletal muscle tissue repair and regeneration. Both physiological and pathological stimuli potentially affect SCs activation, proliferation, and terminal differentiation the former category being the focus of this article. Activation of SCs occurs following exercise, post-training micro-injuries, and electrical stimulation. Exercise, as a potent and natural stimulus, is at the center of numerous studies on SC activation and relevant fields. According to research, different exercise modalities end with various effects. This review article attempts to picture the state of the art of the SCs life span and their engagement in muscle regeneration and hypertrophy in exercise.
Collapse
Affiliation(s)
- Behzad Bazgir
- Exercise Physiology Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
- Department of Embryology, Reproductive Biomedicine Research Center, Royan Institute for Reproductive
Biomedicine, ACECR, Tehran, Iran
| | - Rouhollah Fathi
- Department of Embryology, Reproductive Biomedicine Research Center, Royan Institute for Reproductive
Biomedicine, ACECR, Tehran, Iran
| | - Mojtaba Rezazadeh Valojerdi
- Department of Embryology, Reproductive Biomedicine Research Center, Royan Institute for Reproductive
Biomedicine, ACECR, Tehran, Iran
- Department of Anatomy, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Paul Mozdziak
- Physiology Graduate Program, North Carolina State University, Raleigh, NC, USA
| | - Alireza Asgari
- Exercise Physiology Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
- Aerospace and Subaquatic Medicine Faculty, Aerospace Medicine Research Center, AJA Medical Sciences
University, Tehran, Iran
| |
Collapse
|
23
|
Fujimaki S, Wakabayashi T, Asashima M, Takemasa T, Kuwabara T. Treadmill running induces satellite cell activation in diabetic mice. Biochem Biophys Rep 2016; 8:6-13. [PMID: 28955935 PMCID: PMC5613654 DOI: 10.1016/j.bbrep.2016.07.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2016] [Revised: 07/01/2016] [Accepted: 07/08/2016] [Indexed: 11/19/2022] Open
Abstract
Skeletal muscle-derived stem cells, termed as satellite cells, play essential roles in regeneration after muscle injury in adult skeletal muscle. Diabetes mellitus (DM), one of the most common metabolic diseases, causes impairments of satellite cell function. However, the studies of the countermeasures for the DM-induced dysfunction of satellite cells have been poor. Here, we investigated the effects of chronic running exercise on satellite cell activation in diabetic mice focused on the molecular mechanism including Notch and Wnt signaling, which are contribute to the fate determination of satellite cells. Male C57BL/6 mice 4 weeks of age were injected with streptozotocin and were randomly divided into runner group and control group. Runner group mice were performed treadmill running for 4 weeks. DM attenuated satellite cell activation and the expressions of the components of Notch and Wnt signaling. However, chronic running resulted in activation of satellite cells in diabetic mice and salvaged the inactivity of Wnt signaling but not Notch signaling. Our results suggest that chronic running induces satellite cell activation via upregulation of Wnt signaling in diabetic as well as normal mice. Diabetes attenuates satellite cell activation. Diabetes downregulates the activities of Notch and Wnt signaling. Treadmill running activates satellite cells in diabetic mice. Treadmill running can salvage diabetes-induced downregulation of Wnt signaling.
Collapse
Affiliation(s)
- Shin Fujimaki
- Stem Cell Engineering Research Group, Biotechnology Research Institute for Drug Discovery, Department of Life Science and Biotechnology, National Institute of Advanced Industrial Science and Technology (AIST), Central 5, 1-1-1 Higashi, Tsukuba, Ibaraki 305-8565, Japan
- Physical Education, Health and Sport Sciences, Graduate School of Comprehensive Human Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8574, Japan
| | - Tamami Wakabayashi
- Stem Cell Engineering Research Group, Biotechnology Research Institute for Drug Discovery, Department of Life Science and Biotechnology, National Institute of Advanced Industrial Science and Technology (AIST), Central 5, 1-1-1 Higashi, Tsukuba, Ibaraki 305-8565, Japan
| | - Makoto Asashima
- Stem Cell Engineering Research Group, Biotechnology Research Institute for Drug Discovery, Department of Life Science and Biotechnology, National Institute of Advanced Industrial Science and Technology (AIST), Central 5, 1-1-1 Higashi, Tsukuba, Ibaraki 305-8565, Japan
| | - Tohru Takemasa
- Physical Education, Health and Sport Sciences, Graduate School of Comprehensive Human Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8574, Japan
- Corresponding authors.
| | - Tomoko Kuwabara
- Stem Cell Engineering Research Group, Biotechnology Research Institute for Drug Discovery, Department of Life Science and Biotechnology, National Institute of Advanced Industrial Science and Technology (AIST), Central 5, 1-1-1 Higashi, Tsukuba, Ibaraki 305-8565, Japan
- Corresponding authors.
| |
Collapse
|
24
|
Functional Overload Enhances Satellite Cell Properties in Skeletal Muscle. Stem Cells Int 2015; 2016:7619418. [PMID: 26779264 PMCID: PMC4686724 DOI: 10.1155/2016/7619418] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2015] [Accepted: 07/29/2015] [Indexed: 12/24/2022] Open
Abstract
Skeletal muscle represents a plentiful and accessible source of adult stem cells. Skeletal-muscle-derived stem cells, termed satellite cells, play essential roles in postnatal growth, maintenance, repair, and regeneration of skeletal muscle. Although it is well known that the number of satellite cells increases following physical exercise, functional alterations in satellite cells such as proliferative capacity and differentiation efficiency following exercise and their molecular mechanisms remain unclear. Here, we found that functional overload, which is widely used to model resistance exercise, causes skeletal muscle hypertrophy and converts satellite cells from quiescent state to activated state. Our analysis showed that functional overload induces the expression of MyoD in satellite cells and enhances the proliferative capacity and differentiation potential of these cells. The changes in satellite cell properties coincided with the inactivation of Notch signaling and the activation of Wnt signaling and likely involve modulation by transcription factors of the Sox family. These results indicate the effects of resistance exercise on the regulation of satellite cells and provide insight into the molecular mechanism of satellite cell activation following physical exercise.
Collapse
|
25
|
Tanaka S, Kawahara E, Nakagawa T. Myogenic cell response to muscle contraction with short electrical stimulation. J Phys Ther Sci 2015; 27:2349-52. [PMID: 26311981 PMCID: PMC4540879 DOI: 10.1589/jpts.27.2349] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2015] [Accepted: 04/16/2015] [Indexed: 01/16/2023] Open
Abstract
[Purpose] The present study aimed to determine the effects of short muscle strength exercise on hepatocyte growth factor expression and satellite cell activation. [Subjects] The study included 72 2-12-week-old male Sprague-Dawley rats. [Methods] The rat plantaris muscle was contracted with a 5-min electrical stimulation of the sciatic nerve, and then, the mRNA expressions of hepatocyte growth factor and myogenic regulatory factors in the plantaris muscle were determined, and the phosphorylation of the hepatocyte growth factor receptor (c-Met) was examined. [Results] The mRNA expressions of hepatocyte growth factor and myogenic regulatory factors increased after a short muscle contraction compared to that un-contraction. Immunofluorescence analysis showed the expression of hepatocyte growth factor protein and the possibility that downstream biological changes occurred in the hepatocyte growth factor-bound c-Met. [Conclusion] Our results demonstrated that activation of satellite cells induced hepatocyte growth factor expression during muscle contraction with a short 5-min electrical stimulation, which simulates short muscle strength exercise in physical therapy. The present study provides evidence for the use of short muscle strength exercise in physical therapy.
Collapse
Affiliation(s)
- Shoji Tanaka
- Faculty of Health Sciences, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Japan
| | - Ei Kawahara
- Faculty of Health Sciences, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Japan
| | - Takao Nakagawa
- Faculty of Health Sciences, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Japan
| |
Collapse
|
26
|
Isanejad A, Saraf ZH, Mahdavi M, Gharakhanlou R, Shamsi MM, Paulsen G. The effect of endurance training and downhill running on the expression of IL-1β, IL-6, and TNF-α and HSP72 in rat skeletal muscle. Cytokine 2015; 73:302-8. [DOI: 10.1016/j.cyto.2015.03.013] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2014] [Revised: 03/02/2015] [Accepted: 03/12/2015] [Indexed: 11/27/2022]
|
27
|
Diabetes and stem cell function. BIOMED RESEARCH INTERNATIONAL 2015; 2015:592915. [PMID: 26075247 PMCID: PMC4449886 DOI: 10.1155/2015/592915] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/10/2014] [Accepted: 12/01/2014] [Indexed: 12/30/2022]
Abstract
Diabetes mellitus is one of the most common serious metabolic diseases that results in hyperglycemia due to defects of insulin secretion or insulin action or both. The present review focuses on the alterations to the diabetic neuronal tissues and skeletal muscle, including stem cells in both tissues, and the preventive effects of physical activity on diabetes. Diabetes is associated with various nervous disorders, such as cognitive deficits, depression, and Alzheimer's disease, and that may be caused by neural stem cell dysfunction. Additionally, diabetes induces skeletal muscle atrophy, the impairment of energy metabolism, and muscle weakness. Similar to neural stem cells, the proliferation and differentiation are attenuated in skeletal muscle stem cells, termed satellite cells. However, physical activity is very useful for preventing the diabetic alteration to the neuronal tissues and skeletal muscle. Physical activity improves neurogenic capacity of neural stem cells and the proliferative and differentiative abilities of satellite cells. The present review proposes physical activity as a useful measure for the patients in diabetes to improve the physiological functions and to maintain their quality of life. It further discusses the use of stem cell-based approaches in the context of diabetes treatment.
Collapse
|
28
|
Xing H, Zhou M, Assinck P, Liu N. Electrical stimulation influences satellite cell differentiation after sciatic nerve crush injury in rats. Muscle Nerve 2015; 51:400-11. [PMID: 24947716 DOI: 10.1002/mus.24322] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/16/2014] [Indexed: 12/13/2022]
Abstract
INTRODUCTION Electrical stimulation is often used to prevent muscle atrophy and preserve contractile function, but its effects on the satellite cell population after nerve injury are not well understood. In this study we aimed to determine whether satellite cell differentiation is affected by electrical stimulation after nerve crush. METHODS The sciatic nerves of Sprague-Dawley (SD) rats were crushed. Half of the injured rats received daily electrical stimulation of the gastrocnemius muscle, and the others did not. Tests for detecting paired box protein 7 (Pax7), myogenic differentiation antigen (MyoD), embryonic myosin heavy chain (eMyHC), and force production were performed 2, 4, and 6 weeks after injury. RESULTS More Pax7+/MyoD+ nuclei in stimulated muscles were observed than in non-stimulated muscles. eMyHC expression was elevated in stimulated muscles and correlated positively with enhanced force production. CONCLUSIONS Increased satellite cell differentiation is correlated with preserved muscle function in response to electrical stimulation after nerve injury.
Collapse
Affiliation(s)
- Huayi Xing
- Department of Rehabilitation Medicine, Peking University Third Hospital, 49 North Garden Road, Beijing, 100191, PR China
| | | | | | | |
Collapse
|
29
|
Graham ZA, Touchberry CD, Gupte AA, Bomhoff GL, Geiger PC, Gallagher PM. Changes in α7β1 integrin signaling after eccentric exercise in heat-shocked rat soleus. Muscle Nerve 2015; 51:562-8. [PMID: 24956997 DOI: 10.1002/mus.24324] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/18/2014] [Indexed: 12/16/2022]
Abstract
INTRODUCTION α7β1 integrin links the extracellular matrix to the focal adhesion (FA) in skeletal muscle and serves as a stabilizing and signal relayer. Heat shock (HS) induces expression of proteins that interact with the FA. METHODS Male Wistar rats were assigned to 1 of 3 groups: control (CON); eccentric exercise (EE); or EE+HS (HS). Soleus muscle was analyzed at 2 h and 48 h post-exercise. RESULTS The 120-kDa α7 integrin decreased in the EE and HS groups, and the 70-kDa peptide decreased in the EE group at 2 h post-exercise. Total expression of focal adhesion kinase (FAK) and RhoA were decreased in EE and HS at 2 h post-exercise. Expression of phosphorylated FAK(397) decreased in the EE group but not the HS group at 2 h post-exercise. CONCLUSIONS Long-duration EE may cause alterations in the FA in rat soleus muscle through the α7 integrin subunit and FAK.
Collapse
Affiliation(s)
- Zachary A Graham
- Applied Physiology Laboratory, Department of Health, Sport, and Exercise Sciences, University of Kansas, 101DJ Robinson Center, 1301 Sunnyside Avenue, Lawrence, Kansas, 66045, USA
| | | | | | | | | | | |
Collapse
|
30
|
Itoh Y, Hayakawa K, Mori T, Agata N, Inoue-Miyazu M, Murakami T, Sokabe M, Kawakami K. Stand-up exercise training facilitates muscle recovery from disuse atrophy by stimulating myogenic satellite cell proliferation in mice. Physiol Rep 2014; 2:2/11/e12185. [PMID: 25367692 PMCID: PMC4255801 DOI: 10.14814/phy2.12185] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Determining the cellular and molecular recovery processes in inactivity – or unloading –induced atrophied muscles should improve rehabilitation strategies. We assessed the effects of stand‐up exercise (SE) training on the recovery of atrophied skeletal muscles in male mice. Mice were trained to stand up and press an elevated lever in response to a light‐tone cue preceding an electric foot shock and then subjected to tail suspension (TS) for 2 weeks to induce disuse atrophy in hind limb muscles. After release from TS, mice were divided into SE‐trained (SE cues: 25 times per set, two sets per day) and non‐SE‐trained groups. Seven days after the training, average myofiber cross‐sectional area (CSA) of the soleus muscle was significantly greater in the SE‐trained group than in the non‐SE‐trained group (1843 ± 194 μm2 vs. 1315 ± 153 μm2). Mean soleus muscle CSA in the SE trained group was not different from that in the CON group subjected to neither TS nor SE training (2005 ± 196 μm2), indicating that SE training caused nearly complete recovery from muscle atrophy. The number of myonuclei per myofiber was increased by ~60% in the SE‐trained group compared with the non‐SE‐trained and CON groups (0.92 ± 0.03 vs. 0.57 ± 0.03 and 0.56 ± 0.11, respectively). The number of proliferating myonuclei, identified by 5‐ethynyl‐2′‐deoxyuridine staining, increased within the first few days of SE training. Thus, it is highly likely that myogenic satellite cells proliferated rapidly in atrophied muscles in response to SE training and fused with existing myofibers to reestablish muscle mass. Stand‐up exercise (SE) training facilitates recovery of myofiber size within 7 days and increases the number of myonuclei during atrophied muscle recovery. This rapid increase in the number of myonuclei derived from myogenic satellite cells may account for the rapid histological changes in atrophied muscles induced by SE training.
Collapse
Affiliation(s)
- Yuta Itoh
- Physical and Occupational Therapy Program, Nagoya University Graduate School of Medicine, Nagoya, Japan Faculty of Rehabilitation Science, Nagoya Gakuin University, Seto, Japan
| | - Kimihide Hayakawa
- Mechanobiology Laboratory, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Tomohiro Mori
- Physical and Occupational Therapy Program, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Nobuhide Agata
- Physical and Occupational Therapy Program, Nagoya University Graduate School of Medicine, Nagoya, Japan Faculty of Health and Medical Sciences, Tokoha University, Hamamatsu, Japan
| | | | - Taro Murakami
- Faculty of Wellness, Sigakkan University, Ohbu, Japan
| | - Masahiro Sokabe
- Mechanobiology Laboratory, Nagoya University Graduate School of Medicine, Nagoya, Japan Department of Physiology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Keisuke Kawakami
- Physical and Occupational Therapy Program, Nagoya University Graduate School of Medicine, Nagoya, Japan
| |
Collapse
|
31
|
Neels JG, Grimaldi PA. Physiological functions of peroxisome proliferator-activated receptor β. Physiol Rev 2014; 94:795-858. [PMID: 24987006 DOI: 10.1152/physrev.00027.2013] [Citation(s) in RCA: 124] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The peroxisome proliferator-activated receptors, PPARα, PPARβ, and PPARγ, are a family of transcription factors activated by a diversity of molecules including fatty acids and fatty acid metabolites. PPARs regulate the transcription of a large variety of genes implicated in metabolism, inflammation, proliferation, and differentiation in different cell types. These transcriptional regulations involve both direct transactivation and interaction with other transcriptional regulatory pathways. The functions of PPARα and PPARγ have been extensively documented mainly because these isoforms are activated by molecules clinically used as hypolipidemic and antidiabetic compounds. The physiological functions of PPARβ remained for a while less investigated, but the finding that specific synthetic agonists exert beneficial actions in obese subjects uplifted the studies aimed to elucidate the roles of this PPAR isoform. Intensive work based on pharmacological and genetic approaches and on the use of both in vitro and in vivo models has considerably improved our knowledge on the physiological roles of PPARβ in various cell types. This review will summarize the accumulated evidence for the implication of PPARβ in the regulation of development, metabolism, and inflammation in several tissues, including skeletal muscle, heart, skin, and intestine. Some of these findings indicate that pharmacological activation of PPARβ could be envisioned as a therapeutic option for the correction of metabolic disorders and a variety of inflammatory conditions. However, other experimental data suggesting that activation of PPARβ could result in serious adverse effects, such as carcinogenesis and psoriasis, raise concerns about the clinical use of potent PPARβ agonists.
Collapse
Affiliation(s)
- Jaap G Neels
- Institut National de la Santé et de la Recherche Médicale U 1065, Mediterranean Center of Molecular Medicine (C3M), Team "Adaptive Responses to Immuno-metabolic Dysregulations," Nice, France; and Faculty of Medicine, University of Nice Sophia-Antipolis, Nice, France
| | - Paul A Grimaldi
- Institut National de la Santé et de la Recherche Médicale U 1065, Mediterranean Center of Molecular Medicine (C3M), Team "Adaptive Responses to Immuno-metabolic Dysregulations," Nice, France; and Faculty of Medicine, University of Nice Sophia-Antipolis, Nice, France
| |
Collapse
|
32
|
Fujimaki S, Hidaka R, Asashima M, Takemasa T, Kuwabara T. Wnt protein-mediated satellite cell conversion in adult and aged mice following voluntary wheel running. J Biol Chem 2014; 289:7399-412. [PMID: 24482229 DOI: 10.1074/jbc.m113.539247] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Muscle represents an abundant, accessible, and replenishable source of adult stem cells. Skeletal muscle-derived stem cells, called satellite cells, play essential roles in regeneration after muscle injury in adult skeletal muscle. Although the molecular mechanism of muscle regeneration process after an injury has been extensively investigated, the regulation of satellite cells under steady state during the adult stage, including the reaction to exercise stimuli, is relatively unknown. Here, we show that voluntary wheel running exercise, which is a low stress exercise, converts satellite cells to the activated state due to accelerated Wnt signaling. Our analysis showed that up-regulated canonical Wnt/β-catenin signaling directly modulated chromatin structures of both MyoD and Myf5 genes, resulting in increases in the mRNA expression of Myf5 and MyoD and the number of proliferative Pax7(+)Myf5(+) and Pax7(+) MyoD(+) cells in skeletal muscle. The effect of Wnt signaling on the activation of satellite cells, rather than Wnt-mediated fibrosis, was observed in both adult and aged mice. The association of β-catenin, T-cell factor, and lymphoid enhancer transcription factors of multiple T-cell factor/lymphoid enhancer factor regulatory elements, conserved in mouse, rat, and human species, with the promoters of both the Myf5 and MyoD genes drives the de novo myogenesis in satellite cells even in aged muscle. These results indicate that exercise-stimulated extracellular Wnts play a critical role in the regulation of satellite cells in adult and aged skeletal muscle.
Collapse
Affiliation(s)
- Shin Fujimaki
- From the Research Center for Stem Cell Engineering, National Institute of Advanced Industrial Science and Technology, Tsukuba, Ibaraki 305-0046 and
| | | | | | | | | |
Collapse
|
33
|
Intrinsic ability of adult stem cell in skeletal muscle: an effective and replenishable resource to the establishment of pluripotent stem cells. Stem Cells Int 2013; 2013:420164. [PMID: 23818907 PMCID: PMC3684130 DOI: 10.1155/2013/420164] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2012] [Revised: 04/03/2013] [Accepted: 05/07/2013] [Indexed: 02/06/2023] Open
Abstract
Adult stem cells play an essential role in mammalian organ maintenance and repair throughout adulthood since they ensure that organs retain their ability to regenerate. The choice of cell fate by adult stem cells for cellular proliferation, self-renewal, and differentiation into multiple lineages is critically important for the homeostasis and biological function of individual organs. Responses of stem cells to stress, injury, or environmental change are precisely regulated by intercellular and intracellular signaling networks, and these molecular events cooperatively define the ability of stem cell throughout life. Skeletal muscle tissue represents an abundant, accessible, and replenishable source of adult stem cells. Skeletal muscle contains myogenic satellite cells and muscle-derived stem cells that retain multipotent differentiation abilities. These stem cell populations have the capacity for long-term proliferation and high self-renewal. The molecular mechanisms associated with deficits in skeletal muscle and stem cell function have been extensively studied. Muscle-derived stem cells are an obvious, readily available cell resource that offers promise for cell-based therapy and various applications in the field of tissue engineering. This review describes the strategies commonly used to identify and functionally characterize adult stem cells, focusing especially on satellite cells, and discusses their potential applications.
Collapse
|
34
|
Qun Z, Xinkai Y, Jing W. Effects of eccentric exercise on branched-chain amino acid profiles in rat serum and skeletal muscle. J Anim Physiol Anim Nutr (Berl) 2013; 98:215-22. [DOI: 10.1111/jpn.12062] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2012] [Accepted: 02/03/2013] [Indexed: 12/20/2022]
Affiliation(s)
- Z. Qun
- School of Kinesiology; Shanghai University of Sport; Shanghai China
| | - Y. Xinkai
- School of Kinesiology; Shanghai University of Sport; Shanghai China
| | - W. Jing
- Laboratory of Sports Medicine and Physiology; Tianjin University of Sport; Tianjin China
| |
Collapse
|
35
|
Touchberry CD, Gupte AA, Bomhoff GL, Graham ZA, Geiger PC, Gallagher PM. Acute heat stress prior to downhill running may enhance skeletal muscle remodeling. Cell Stress Chaperones 2012; 17:693-705. [PMID: 22589083 PMCID: PMC3468678 DOI: 10.1007/s12192-012-0343-5] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2011] [Revised: 04/16/2012] [Accepted: 04/23/2012] [Indexed: 02/07/2023] Open
Abstract
Heat shock proteins (HSPs) are chaperones that are known to have important roles in facilitating protein synthesis, protein assembly and cellular protection. While HSPs are known to be induced by damaging exercise, little is known about how HSPs actually mediate skeletal muscle adaption to exercise. The purpose of this study was to determine the effects of a heat shock pretreatment and the ensuing increase in HSP expression on early remodeling and signaling (2 and 48 h) events of the soleus (Sol) muscle following a bout of downhill running. Male Wistar rats (10 weeks old) were randomly assigned to control, eccentric exercise (EE; downhill running) or heat shock + eccentric exercise (HS; 41°C for 20 min, 48 h prior to exercise) groups. Markers of muscle damage, muscle regeneration and intracellular signaling were assessed. The phosphorylation (p) of HSP25, Akt, p70s6k, ERK1/2 and JNK proteins was also performed. As expected, following exercise the EE group had increased creatine kinase (CK; 2 h) and mononuclear cell infiltration (48 h) compared to controls. The EE group had an increase in p-HSP25, but there was no change in HSP72 expression, total protein concentration, or neonatal MHC content. Additionally, the EE group had increased p-p70s6k, p-ERK1/2, and p-JNK (2 h) compared to controls; however no changes in p-Akt were seen. In contrast, the HS group had reduced CK (2 h) and mononuclear cell infiltration (48 h) compared to EE. Moreover, the HS group had increased HSP72 content (2 and 48 h), total protein concentration (48 h), neonatal MHC content (2 and 48 h), p-HSP25 and p-p70s6k (2 h). Lastly, the HS group had reduced p-Akt (48 h) and p-ERK1/2 (2 h). These data suggest that heat shock pretreatment and/or the ensuing HSP72 response may protect against muscle damage, and enhance increases in total protein and neonatal MHC content following exercise. These changes appear to be independent of Akt and MAPK signaling pathways.
Collapse
Affiliation(s)
- Chad D. Touchberry
- University of Missouri-Kansas City, School of Medicine - Basic Medical Science, Health Sciences Building, 2464 Charlotte Street, Room 2211, Kansas City, MO 64108 USA
| | - Anisha A. Gupte
- Muscle Physiology Laboratory, University of Kansas Medical Center, Kansas City, KS 66160 USA
| | - Gregory L. Bomhoff
- Muscle Physiology Laboratory, University of Kansas Medical Center, Kansas City, KS 66160 USA
| | - Zachary A. Graham
- Applied Physiology Laboratory, University of Kansas, 1301 Sunnyside Avenue, Lawrence, KS 66045 USA
| | - Paige C. Geiger
- Muscle Physiology Laboratory, University of Kansas Medical Center, Kansas City, KS 66160 USA
| | - Philip M. Gallagher
- Applied Physiology Laboratory, University of Kansas, 1301 Sunnyside Avenue, Lawrence, KS 66045 USA
| |
Collapse
|
36
|
Erythropoietin enhances the regeneration of traumatized tissue after combined muscle-nerve injury. J Trauma Acute Care Surg 2012; 72:1567-75. [DOI: 10.1097/ta.0b013e318246498f] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
37
|
Yeghiazaryan M, Żybura-Broda K, Cabaj A, Włodarczyk J, Sławińska U, Rylski M, Wilczyński GM. Fine-structural distribution of MMP-2 and MMP-9 activities in the rat skeletal muscle upon training: a study by high-resolution in situ zymography. Histochem Cell Biol 2012; 138:75-87. [PMID: 22419075 PMCID: PMC3374103 DOI: 10.1007/s00418-012-0940-5] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/20/2012] [Indexed: 12/25/2022]
Abstract
Matrix metalloproteinases (MMPs) are key regulators of extracellular matrix remodeling, but have also important intracellular targets. The purpose of this study was to examine the activity and subcellular localization of the gelatinases MMP-2 and MMP-9 in skeletal muscle of control and physically trained rats. In control hind limb muscle, the activity of the gelatinases was barely detectable. In contrast, after 5 days of intense exercise, in Soleus (Sol), but not Extensor digitorum longus (EDL) muscle, significant upregulation of gelatinolytic activity in myofibers was observed mainly in the nuclei, as assessed by high resolution in situ zymography. The nuclei of quiescent satellite cells did not contain the activity. Within the myonuclei, the gelatinolytic activity colocalized with an activated RNA Polymerase II. Also in Sol, but not in EDL, there were few foci of mononuclear cells with strongly positive cytoplasm, associated with apparent necrotic myofibers. These cells were identified as activated satellite cells/myoblasts. No extracellular gelatinase activity was observed. Gel zymography combined with subcellular fractionation revealed training-related upregulation of active MMP-2 in the nuclear fraction, and increase of active MMP-9 in the cytoplasmic fraction of Sol. Using RT-PCR, selective increase in MMP-9 mRNA was observed. We conclude that training activates nuclear MMP-2, and increases expression and activity of cytoplasmic MMP-9 in Sol, but not in EDL. Our results suggest that the gelatinases are involved in muscle adaptation to training, and that MMP-2 may play a novel role in myonuclear functions.
Collapse
Affiliation(s)
- Marine Yeghiazaryan
- Nencki Institute of Experimental Biology, Polish Academy of Sciences, Pasteura 3, 02-093 Warsaw, Poland
| | - Katarzyna Żybura-Broda
- Nencki Institute of Experimental Biology, Polish Academy of Sciences, Pasteura 3, 02-093 Warsaw, Poland
| | - Anna Cabaj
- Nencki Institute of Experimental Biology, Polish Academy of Sciences, Pasteura 3, 02-093 Warsaw, Poland
- Institute of Biocybernetics and Biomedical Engineering, Trojdena 4, 02-109 Warsaw, Poland
| | - Jakub Włodarczyk
- Nencki Institute of Experimental Biology, Polish Academy of Sciences, Pasteura 3, 02-093 Warsaw, Poland
| | - Urszula Sławińska
- Nencki Institute of Experimental Biology, Polish Academy of Sciences, Pasteura 3, 02-093 Warsaw, Poland
| | - Marcin Rylski
- The Medical Center of Postgraduate Education, Marymoncka 99/103, 01-813 Warsaw, Poland
| | - Grzegorz M. Wilczyński
- Nencki Institute of Experimental Biology, Polish Academy of Sciences, Pasteura 3, 02-093 Warsaw, Poland
| |
Collapse
|
38
|
Smith HK, Merry TL. Voluntary resistance wheel exercise during post-natal growth in rats enhances skeletal muscle satellite cell and myonuclear content at adulthood. Acta Physiol (Oxf) 2012; 204:393-402. [PMID: 21854550 DOI: 10.1111/j.1748-1716.2011.02350.x] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
AIM To determine whether voluntary free wheel (FW) or resistance wheel (RW) exercise or reduced muscle activity would influence maturational increases in muscle mass and the number of satellite cells (SCs) and myonuclei (MN) accrued by adulthood. METHODS Hind limb muscles of male rats housed with, or without, FWs from 4 to 5, 7 or 10 weeks of age, and rats housed with RWs from 4 to 10 week of age, were evaluated. To assess the effect of reduced muscle activity, gastrocnemius muscles of 4-week-old rats were injected with botulinum toxin (Btx) and collected at 7 weeks of age. Muscle fibre size and the frequency of Pax7-positive SCs and MN were determined in 7- and 10-week-old muscles via immunohistochemical methods. RESULTS Free wheel exercise enhanced muscle growth and the frequency of SCs in the medial gastrocnemius (MG) (threefold) and vastus lateralis (VL) (twofold) of rats at 10 week of age. Resistance wheel exercise increased the number of SCs and MN (22-30%), with more muscle fibre nuclei being associated with larger fibre size, in the soleus, MG and VL muscles. Btx impaired the normal increases in muscle fibre size and the accrual of MN but not SCs. CONCLUSION A greater volume of exercise during maturational growth was important for enhancing SC numbers, whereas their conversion to MN required higher-intensity exercise. The enhanced muscle fibre nuclear populations may influence the capacity of the muscle to adapt to exercise, injury or disuse in later adulthood.
Collapse
Affiliation(s)
- H K Smith
- Department of Sport and Exercise Science, University of Auckland, New Zealand.
| | | |
Collapse
|
39
|
Skovgaard D, Bayer ML, Mackey AL, Madsen J, Kjaer M, Kjaer A. Increased cellular proliferation in rat skeletal muscle and tendon in response to exercise: use of FLT and PET/CT. Mol Imaging Biol 2011; 12:626-34. [PMID: 20379786 DOI: 10.1007/s11307-010-0316-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
PURPOSE The purpose of this study is to investigate exercise-induced cellular proliferation in rat skeletal muscle/tendon with the use of 3'-[F-18]fluoro-3'deoxythymidine (FLT) and to quantitatively study concomitant changes in the proliferation-associated factor, Ki67. PROCEDURES Wistar rats (n = 13) performed 3 days of treadmill running. Cellular proliferation was investigated 3 days before and 48 h after the running exercise with the use of FLT and positron emission tomography/computed tomography (PET/CT). Results were compared to a sedentary control group (n = 10). Image-derived standardized uptake values were calculated for Achilles tendons and calf muscles and compared to gene expression and immunohistochemical evaluations of Ki67. RESULTS Treadmill running induced increased uptake of FLT uptake in calf muscles (30%; p < 0.001) and in Achilles tendon (21%, p < 0.001). The image-derived results were supported by a correlation in calf muscle to Ki67 (protein and mRNA level), while this coherence was not found in tendon. CONCLUSION FLT-PET seems to be a promising tool for imaging of exercise-induced cellular proliferation in musculo-tendinous tissue.
Collapse
Affiliation(s)
- Dorthe Skovgaard
- Institute of Sports Medicine Copenhagen, Bispebjerg Hospital and Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark.
| | | | | | | | | | | |
Collapse
|
40
|
Laziz I, Ferry A, Armand AS, Gallien CL, Gaspera BD, Charbonnier F, Chanoine C. Eccentric stimulation reveals an involvement of FGF6 in muscle resistance to mechanical stress. Eur J Appl Physiol 2010; 111:1507-15. [PMID: 21188410 DOI: 10.1007/s00421-010-1784-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2010] [Accepted: 12/10/2010] [Indexed: 11/26/2022]
Abstract
The objective of this report was to analyse a potential role for FGF6 in muscle resistance to mechanical stress. Normal or regenerating muscles of FGF6 (-/-) mice versus wild-type mice were submitted to different protocols of damaging eccentric contractions (eccentric electrostimulation and intermittent downhill exercise). Then muscular structural properties were analysed by histological and immunochemistry techniques to evaluate the post-injury muscle recovery; their muscle contractile parameters (maximal tetanic force, kinetics properties and fatigue resistance) were assessed. The absence of FGF6 causes (1) a fast-to-slow myofibre type switch in adult control and regenerating Tibialis anterior (TA) muscle; (2) muscle weakness in regenerating muscles in animals submitted to eccentric exercise protocols due to aberrant extensive necrotic zones. These observations point out a crucial and unexpected role for FGF6 in muscle integrity and muscle protection against mechanical stress.
Collapse
Affiliation(s)
- Iman Laziz
- UMR 8194 CNRS, Centre d'Etude de Sensori-Motricité, Centre Universitaire des Saints-Pères, Université Paris Descartes, 45 rue des Saints-Pères, 75270 Paris Cedex 06, France
| | | | | | | | | | | | | |
Collapse
|
41
|
Khattak MJ, Ahmad T, Rehman R, Umer M, Hasan SH, Ahmed M. Muscle healing and nerve regeneration in a muscle contusion model in the rat. ACTA ACUST UNITED AC 2010; 92:894-9. [PMID: 20513892 DOI: 10.1302/0301-620x.92b6.22819] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The nervous system is known to be involved in inflammation and repair. We aimed to determine the effect of physical activity on the healing of a muscle injury and to examine the pattern of innervation. Using a drop-ball technique, a contusion was produced in the gastrocnemius in 20 rats. In ten the limb was immobilised in a plaster cast and the remaining ten had mobilisation on a running wheel. The muscle and the corresponding dorsal-root ganglia were studied by histological and immunohistochemical methods. In the mobilisation group, there was a significant reduction in lymphocytes (p = 0.016), macrophages (p = 0.008) and myotubules (p = 0.008) between three and 21 days. The formation of myotubules and the density of nerve fibres was significantly higher (both p = 0.016) compared with those in the immobilisation group at three days, while the density of CGRP-positive fibres was significantly lower (p = 0.016) after 21 days. Mobilisation after contusional injury to the muscle resulted in early and increased formation of myotubules, early nerve regeneration and progressive reduction in inflammation, suggesting that it promoted a better healing response.
Collapse
Affiliation(s)
- M J Khattak
- University College London Hospital, London NW1 2BU, UK
| | | | | | | | | | | |
Collapse
|
42
|
Tsivitse S. Notch and Wnt signaling, physiological stimuli and postnatal myogenesis. Int J Biol Sci 2010; 6:268-81. [PMID: 20567496 PMCID: PMC2878172 DOI: 10.7150/ijbs.6.268] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2010] [Accepted: 05/13/2010] [Indexed: 12/18/2022] Open
Abstract
Adult skeletal muscle stem cells, termed satellite cells are imperative to muscle regeneration. Much work has been performed on satellite cell identification and the subsequent activation of the myogenic response but the regulation of satellite cells including its activation is not well elucidated. The purpose of this review article is to synthesize what the literature reveals in regards to the current understanding of satellite cells including their contribution to muscle repair and growth following physiological stimuli. In addition, this review article will describe the recent findings on the roles of the classic developmental signaling pathways, Notch and Wnt, to the myogenic response in various muscle injury models. This purpose of this summary is to bring awareness of the impact that muscle contraction models have on the local and systemic environment of adult muscle stem cells which will be beneficial for comprehending and treatment development for muscle -associated ailments and other organ diseases.
Collapse
Affiliation(s)
- Susan Tsivitse
- Department of Kinesiology, Exercise Physiology Laboratory, University North Carolina-Charlotte, NC 28223, USA.
| |
Collapse
|
43
|
Abstract
As women enter menopause, the concentration of estrogen and other female hormones declines. This hormonal decrease has been associated with a number of negative outcomes, including a greater incidence of injury as well as a delay in recovery from these injuries. Over the past two decades, our understanding of the protective effects of estrogen against various types of injury and disease states has grown immensely. In skeletal muscle, studies with animals have demonstrated that sex and estrogen may potentially influence muscle contractile properties and attenuate indices of post-exercise muscle damage, including the release of creatine kinase into the bloodstream and activity of the intramuscular lysosomal acid hydrolase, beta-glucuronidase. Furthermore, numerous studies have revealed an estrogen-mediated attenuation of infiltration of inflammatory cells such as neutrophils and macrophages into the skeletal muscles of rats following exercise or injury. Estrogen has also been shown to play a significant role in stimulating muscle repair and regenerative processes, including the activation and proliferation of satellite cells. Although the mechanisms by which estrogen exerts its influence upon indices of skeletal muscle damage, inflammation and repair have not been fully elucidated, it is thought that estrogen may potentially exert its protective effects by: (i) acting as an antioxidant, thus limiting oxidative damage; (ii) acting as a membrane stabilizer by intercalating within membrane phospholipids; and (iii) binding to estrogen receptors, thus governing the regulation of a number of downstream genes and molecular targets. In contrast to animal studies, studies with humans have not as clearly delineated an effect of estrogen on muscle contractile function or on indices of post-exercise muscle damage and inflammation. These inconsistencies have been attributed to a number of factors, including age and fitness level of subjects, the type and intensity of exercise protocols, and a focus on sex differences that typically involve factors and hormones in addition to estrogen. In recent years, hormone replacement therapy (HRT) or estrogen combined with exercise have been proposed as potentially therapeutic agents for postmenopausal women, as these agents may potentially limit muscle damage and inflammation and stimulate repair in this population. While the benefits and potential health risks of long-term HRT use have been widely debated, controlled studies using short-term HRT or other estrogen agonists may provide future new and valuable insights into understanding the effects of estrogen on skeletal muscle, and greatly benefit the aging female population. Recent studies with older females have begun to demonstrate their benefits.
Collapse
Affiliation(s)
- Deborah L Enns
- Department of Kinesiology and Physical Education, Faculty of Science, Wilfrid Laurier University, Waterloo, Ontario, Canada
| | | |
Collapse
|
44
|
Yamada M, Tatsumi R, Yamanouchi K, Hosoyama T, Shiratsuchi SI, Sato A, Mizunoya W, Ikeuchi Y, Furuse M, Allen RE. High concentrations of HGF inhibit skeletal muscle satellite cell proliferation in vitro by inducing expression of myostatin: a possible mechanism for reestablishing satellite cell quiescence in vivo. Am J Physiol Cell Physiol 2009; 298:C465-76. [PMID: 20007454 DOI: 10.1152/ajpcell.00449.2009] [Citation(s) in RCA: 73] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Skeletal muscle regeneration and work-induced hypertrophy rely on molecular events responsible for activation and quiescence of resident myogenic stem cells, satellite cells. Recent studies demonstrated that hepatocyte growth factor (HGF) triggers activation and entry into the cell cycle in response to mechanical perturbation, and that subsequent expression of myostatin may signal a return to cell quiescence. However, mechanisms responsible for coordinating expression of myostatin after an appropriate time lag following activation and proliferation are not clear. Here we address the possible role of HGF in quiescence through its concentration-dependent negative-feedback mechanism following satellite cell activation and proliferation. When activated/proliferating satellite cell cultures were treated for 24 h beginning 48-h postplating with 10-500 ng/ml HGF, the percentage of bromodeoxyuridine-incorporating cells decreased down to a baseline level comparable to 24-h control cultures in a HGF dose-dependent manner. The high level HGF treatment did not impair the cell viability and differentiation levels, and cells could be reactivated by lowering HGF concentrations to 2.5 ng/ml, a concentration that has been shown to optimally stimulate activation of satellite cells in culture. Coaddition of antimyostatin neutralizing antibody could prevent deactivation and abolish upregulation of cyclin-dependent kinase (Cdk) inhibitor p21. Myostatin mRNA expression was upregulated with high concentrations of HGF, as demonstrated by RT-PCR, and enhanced myostatin protein expression and secretion were revealed by Western blots of the cell lysates and conditioned media. These results indicate that HGF could induce satellite cell quiescence by stimulating myostatin expression. The HGF concentration required (over 10-50 ng/ml), however, is much higher than that for activation, which is initiated by rapid release of HGF from its extracellular association. Considering that HGF is produced by satellite cells and spleen and liver cells in response to muscle damage, local concentrations of HGF bathing satellite cells may reach a threshold sufficient to induce myostatin expression. This time lag may delay action of the quiescence signaling program in proliferating satellite cells during initial phases of muscle regeneration followed by induction of quiescence in a subset of cells during later phases.
Collapse
Affiliation(s)
- Michiko Yamada
- Dept. of Bioscience and Biotechnology, Graduate School of Agriculture, Kyushu University, Hakozaki 6-10-1, Higashi-ku, Fukuoka 812-8581, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Tatsumi R, Sankoda Y, Anderson JE, Sato Y, Mizunoya W, Shimizu N, Suzuki T, Yamada M, Rhoads RP, Ikeuchi Y, Allen RE. Possible implication of satellite cells in regenerative motoneuritogenesis: HGF upregulates neural chemorepellent Sema3A during myogenic differentiation. Am J Physiol Cell Physiol 2009; 297:C238-52. [DOI: 10.1152/ajpcell.00161.2009] [Citation(s) in RCA: 73] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Regenerative coordination and remodeling of the intramuscular motoneuron network and neuromuscular connections are critical for restoring skeletal muscle function and physiological properties. The regulatory mechanisms of such coordination remain unclear, although both attractive and repulsive axon guidance molecules may be involved in the signaling pathway. Here we show that expression of a neural secreted chemorepellent semaphorin 3A (Sema3A) is remarkably upregulated in satellite cells of resident myogenic stem cells that are positioned beneath the basal lamina of mature muscle fibers, when treated with hepatocyte growth factor (HGF), established as an essential cue in muscle fiber growth and regeneration. When satellite cells were treated with HGF in primary cultures of cells or muscle fibers, Sema3A message and protein were upregulated as revealed by reverse transcription-polymerase chain reaction and immunochemical studies. Other growth factors had no inductive effect except for a slight effect of epidermal growth factor treatment. Sema3A upregulation was HGF dose dependent with a maximum (about 7- to 8-fold units relative to the control) at 10–25 ng/ml and occurred exclusively at the early-differentiation stage, as characterized by the level of myogenin expression and proliferation (bromodeoxyuridine incorporation) of the cells. Neutralizing antibody to the HGF-specific receptor, c-met, did not abolish the HGF response, indicating that c-met may not mediate the Sema3A expression signaling. Finally, in vivo Sema3A was upregulated in the differentiation phase of satellite cells isolated from muscle regenerating following crush injury. Overall, the data highlight a heretofore unexplored and active role for satellite cells as a key source of Sema3A expression triggered by HGF, hence suggesting that regenerative activity toward motor innervation may importantly reside in satellite cells and could be a crucial contributor during postnatal myogenesis.
Collapse
|
46
|
Kumar D, Shadrach JL, Wagers AJ, Lassar AB. Id3 is a direct transcriptional target of Pax7 in quiescent satellite cells. Mol Biol Cell 2009; 20:3170-7. [PMID: 19458195 DOI: 10.1091/mbc.e08-12-1185] [Citation(s) in RCA: 79] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
Pax7 is a key regulator of skeletal muscle stem cells and is required along with Pax3 to generate skeletal muscle precursors. We have identified a collection of genes induced by either Pax3 or Pax7 in C2C12 muscle cells. Two notable Pax3/7 targets are the inhibitory helix-loop-helix (HLH) proteins inhibitor of DNA binding (Id) 2 and Id3, both of which are coordinately expressed with Pax7 in quiescent satellite cells and are induced in quiescent C2C12 myogenic cells after ectopic expression of either Pax3 or Pax7. Ectopic Pax7 activates expression of a luciferase reporter driven by the Id3 promoter, and maximal induction of this reporter requires a conserved Pax7 binding site located upstream of the Id3 gene. Chromatin immunoprecipitation indicated that Pax7 is bound upstream of the Id3 promoter in quiescent satellite cells. In addition, short hairpin RNA-mediated knockdown of Pax7 expression in cultured satellite cells coordinately decreased both Id2 and Id3 expression. Together, these findings indicate that Id3 is a direct transcriptional target for Pax7 in quiescent satellite cells, and they suggest that Pax7 acts to block premature differentiation of quiescent satellite cells by inducing the expression of Id2 and Id3, which in turn may act to block either the precocious induction of myogenic basic (b)HLH proteins, the activity of myogenic bHLH proteins, or both.
Collapse
Affiliation(s)
- Deepak Kumar
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA
| | | | | | | |
Collapse
|
47
|
Peroxisome proliferator-activated receptor beta activation promotes myonuclear accretion in skeletal muscle of adult and aged mice. Pflugers Arch 2009; 458:901-13. [PMID: 19415321 PMCID: PMC2719750 DOI: 10.1007/s00424-009-0676-9] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2009] [Revised: 04/01/2009] [Accepted: 04/21/2009] [Indexed: 11/01/2022]
Abstract
We reported recently that peroxisome proliferator-activated receptor beta (PPARbeta) activation promotes a calcineurin-dependent exercise-like remodelling characterised by increased numbers of oxidative fibres and capillaries. As physical exercise also induces myonuclear accretion, we investigated whether PPARbeta activation alters myonuclear density. Transgenic muscle-specific PPARbeta over-expression induced 14% increase of myonuclear density. Pharmacological PPARbeta activation promoted rapid and massive myonuclear accretion (20% increase after 48 h), which is dependent upon calcineurin/nuclear factor of activated T cells signalling pathway. In vivo bromodeoxyuridine labelling and proliferating cell nuclear antigen immunodetection revealed that PPARbeta activation did not promote cell proliferation, suggesting that the PPARbeta-promoted myonuclear accretion involves fusion of pre-existing muscle precursor cells to myofibres rather than cell division. Finally, we showed that in skeletal muscle, ageing led to a down-regulation of PPARbeta accompanied by decrease of both oxidative fibre number and myonuclear density. PPARbeta pharmacological activation counteracts, at least in part, the ageing-driven muscle remodelling.
Collapse
|
48
|
Dietary carbohydrate level affects transcription factor expression that regulates skeletal muscle myogenesis in rainbow trout. Comp Biochem Physiol B Biochem Mol Biol 2009; 153:66-72. [PMID: 19416696 DOI: 10.1016/j.cbpb.2009.01.013] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2008] [Revised: 01/26/2009] [Accepted: 01/26/2009] [Indexed: 11/20/2022]
Abstract
Understanding the effects of dietary carbohydrates on transcription factors that regulate myogenesis provides insight into the role of nutrient sensing by satellite cells towards myocyte differentiation. We evaluated the influence of dietary carbohydrate level (0, 15, 25 or 35%) on the temporal mRNA expression patterns (4, 8 or 12 weeks) of transcription factors that regulate satellite cell myocyte addition (MA) in rainbow trout (Oncorhynchus mykiss), a vertebrate with indeterminate growth. Relative to the 0% carbohydrate (NC) diet, 15 (IC-15) and 25% (IC-25) carbohydrate containing diets significantly up-regulate MyoD and Myf5, but not Pax7, after 12 weeks of feeding. Simultaneously, the Pax7/MyoD mRNA expression ratio declined significantly with both the IC diets. Myogenin mRNA expression also increased in rainbow trout (RBT) fed the IC-15 diet. The high carbohydrate (HC) diet (35%) attenuated the increased mRNA expression of these transcription factors. It is of note that the 4 and 8 week samples lacked the promyogenic expression patterns. The myogenic gene expression in fish fed the IC-15 diet for 12 weeks indicate a transcriptional signature that reflects increased satellite cell myogenesis. Our results suggest a potential role for satellite cells in the nutrient sensing ability of a vertebrate with indeterminate skeletal muscle growth.
Collapse
|
49
|
Miyata T, Tanaka S, Tachino K. MyoD and Myogenin mRNA Levels after Single Session of Treadmill Exercise in Rat Skeletal Muscle. J Phys Ther Sci 2009. [DOI: 10.1589/jpts.21.81] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Affiliation(s)
- Takuya Miyata
- Department of Rehabilitation, Tsurugi Hospital
- Department of Impairment Study, Graduate Course of Rehabilitation Science, Division of Health Sciences, Graduate School of Medical Science, Kanazawa University
| | - Shoji Tanaka
- Department of Impairment Study, Graduate Course of Rehabilitation Science, Division of Health Sciences, Graduate School of Medical Science, Kanazawa University
| | - Katsuhiko Tachino
- Department of Impairment Study, Graduate Course of Rehabilitation Science, Division of Health Sciences, Graduate School of Medical Science, Kanazawa University
| |
Collapse
|
50
|
Tanaka S, Miyata T, Fujita T, Kawahara E, Tachino K, Funakoshi H, Nakamura T. Differing Responses of Satellite Cell Activity to Exercise Training in Rat Skeletal Muscle. J Phys Ther Sci 2009. [DOI: 10.1589/jpts.21.141] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Affiliation(s)
- Shoji Tanaka
- Institute of Medical, Pharmaceutical and Health Science, Kanazawa University
| | - Takuya Miyata
- Institute of Medical, Pharmaceutical and Health Science, Kanazawa University
- Department of Rehabilitation, Tsurugi Hospital
| | | | - Ei Kawahara
- Institute of Medical, Pharmaceutical and Health Science, Kanazawa University
| | - Katsuhiko Tachino
- Institute of Medical, Pharmaceutical and Health Science, Kanazawa University
| | - Hiroshi Funakoshi
- Division of Molecular Regenerative Medicine, Department of Biochemistry and Molecular Biology, Osaka University Graduate School of Medicine
| | - Toshikazu Nakamura
- Kringle Pharma Joint Research Division for Regenerative Drug Discovery, Center for Advanced Science and Innovation, Osaka University
| |
Collapse
|