1
|
Abdel-Halim NHM, Farrag EAE, Hammad MO, Habotta OA, Hassan HM. Probiotics Attenuate Myopathic Changes in Aging Rats via Activation of the Myogenic Stellate Cells. Probiotics Antimicrob Proteins 2025; 17:1308-1319. [PMID: 38112993 PMCID: PMC12055950 DOI: 10.1007/s12602-023-10202-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/05/2023] [Indexed: 12/21/2023]
Abstract
Aging represents a complex biological process associated with decline in skeletal muscle functions. Aging impairs satellite cells that serve as muscle progenitor cells. Probiotic supplementation may have many beneficial effects via various mechanisms. We examined the possible effects of probiotics in stimulating the proliferation of myogenic stellate cells in aging rats. Twenty-four male albino Sprague-Dawley rats were classified equally into four groups: adult control, old control, adult + probiotics, and old + probiotics. Probiotics (Lactobacillus LB) were administered gavage at a dose of 1 ml (1 × 109 CFU/ml/day) for 4 weeks. A significant increase in the relative gastrocnemius weight ratio and improvement of contractile parameters was detected in the old + probiotics group (0.6 ± 0.01) compared to the old control group (0.47 ± 0.01; P < 0.001). Probiotics significantly upregulated the activities of GSH, while NO and MDA were markedly decreased compared to control groups (P ≤ 0.001). Also, probiotics increased the mRNA and protein expressions of myogenin and CD34 (P < 0.05) as determined by real-time PCR and immunohistochemistry. Moreover, the old + probiotics group showed apparent restoration of the connective tissue spaces, reflecting the all-beneficial effects of probiotics. Our findings indicated that probiotics attenuated myopathic changes in aging rats probably through activation of the myogenic stellate cells. Probiotics improved the muscle weight, function, antioxidant activity, and myogenic transcription factors of the skeletal muscle.
Collapse
Affiliation(s)
- Nehal H M Abdel-Halim
- Physiology Department, Faculty of Medicine, Mansoura University, Mansoura, 35511, Egypt
| | - Eman A E Farrag
- Clinical Pharmacology Department, Faculty of Medicine, Mansoura University, Mansoura, 35511, Egypt.
| | - Maha O Hammad
- Medical Biochemistry and Molecular Biology Department, Faculty of Medicine, Mansoura University, Mansoura, 35511, Egypt
| | - Ola Ali Habotta
- Forensic and Toxicology Department, Faculty of Veterinary Medicine, Mansoura University, Mansoura, 35511, Egypt
| | - Hend M Hassan
- Human Anatomy and Embryology Department, Faculty of Medicine, Mansoura University, Mansoura, 35511, Egypt
| |
Collapse
|
2
|
Yang H, Zheng Y, Yu T, Wu B, Liu Z, Liu S, Sun X, Zhou L. A functional role for myostatin in muscle hyperplasia and hypertrophy revealed by comparative transcriptomics in Yesso scallop Patinopecten yessoensis. Int J Biol Macromol 2025; 307:142308. [PMID: 40118415 DOI: 10.1016/j.ijbiomac.2025.142308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Revised: 03/03/2025] [Accepted: 03/18/2025] [Indexed: 03/23/2025]
Abstract
Elucidating the molecular regulatory mechanisms underlying muscle growth and development is of profound significance in aquaculture. Yesso scallop is a cold-water bivalve of considerable economic importance, having its primary edible component of adductor muscle. In this study, comparative transcriptomics and histological analysis at different sampling times after Myostatin (MSTN) interference were performed to identify the potential candidate genes potentially involved in muscle growth and development. The comparative transcriptomics revealed that growth factors and cytokines, extracellular matrix proteins and ubiquitin-proteasome system are potentially involved in muscle hypertrophy and hyperplasia. After MSTN interference, striated adductor muscle displays significant muscle hypertrophy (51.77 % increase on day 7 and 59.83 % increase on day 21) and muscle hyperplasia (59.36 % increase on day 7 and 61.83 % increase on day 21). WGCNA identifies the key darkolivegreen module, which may play crucial roles in muscle hyperplasia and hypertrophy within the striated muscle of the scallop. Five key transcription factors (zf-CCCH, zf-C2H2, PPP1R10, LRRFIP2, and Gon4) are identified by analyzing the co-expression patterns of core genes within the module. These findings will aid in understanding the regulatory mechanisms of muscle growth in scallops and provide a basis for genetic improvement in shellfish aquaculture.
Collapse
Affiliation(s)
- Hongsu Yang
- State Key Laboratory of Mariculture Biobreeding and Sustainable Goods, Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Qingdao, Shandong 266071, China; Laboratory for Marine Fisheries Science and Food Production Processes, Laoshan Laboratory, Qingdao, Shandong 266237, China; Fisheries College, Zhejiang Ocean University, Zhoushan 316022, China
| | - Yanxin Zheng
- Changdao Enhancement and Experiment Station, Chinese Academy of Fishery Sciences, Changdao, China
| | - Tao Yu
- Changdao Enhancement and Experiment Station, Chinese Academy of Fishery Sciences, Changdao, China
| | - Biao Wu
- State Key Laboratory of Mariculture Biobreeding and Sustainable Goods, Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Qingdao, Shandong 266071, China; Laboratory for Marine Fisheries Science and Food Production Processes, Laoshan Laboratory, Qingdao, Shandong 266237, China
| | - Zhihong Liu
- State Key Laboratory of Mariculture Biobreeding and Sustainable Goods, Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Qingdao, Shandong 266071, China; Laboratory for Marine Fisheries Science and Food Production Processes, Laoshan Laboratory, Qingdao, Shandong 266237, China
| | - Shufang Liu
- State Key Laboratory of Mariculture Biobreeding and Sustainable Goods, Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Qingdao, Shandong 266071, China; Laboratory for Marine Fisheries Science and Food Production Processes, Laoshan Laboratory, Qingdao, Shandong 266237, China
| | - Xiujun Sun
- State Key Laboratory of Mariculture Biobreeding and Sustainable Goods, Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Qingdao, Shandong 266071, China; Laboratory for Marine Fisheries Science and Food Production Processes, Laoshan Laboratory, Qingdao, Shandong 266237, China.
| | - Liqing Zhou
- State Key Laboratory of Mariculture Biobreeding and Sustainable Goods, Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Qingdao, Shandong 266071, China; Laboratory for Marine Fisheries Science and Food Production Processes, Laoshan Laboratory, Qingdao, Shandong 266237, China
| |
Collapse
|
3
|
Wageh M, Fortino SA, Pontello R, Maklad A, McGlory C, Kumbhare D, Phillips SM, Parise G. The Effect of Multi-Ingredient Protein versus Collagen Supplementation on Satellite Cell Properties in Males and Females. Med Sci Sports Exerc 2024; 56:2125-2134. [PMID: 39475860 DOI: 10.1249/mss.0000000000003505] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2024]
Abstract
INTRODUCTION Skeletal muscle satellite cells (SC) contribute to the adaptive process of resistance exercise training (RET) and may be influenced by nutritional supplementation. However, little research exists on the impact of multi-ingredient supplementation on the SC response to RET. PURPOSE We tested the effect of a multi-ingredient supplement (MIS) including whey protein, creatine, leucine, calcium citrate, and vitamin D on SC content and activity as well as myonuclear accretion, SC and myonuclear domain compared with a collagen control (COL) throughout a 10-wk RET program. METHODS Twenty-six participants underwent a 10-wk linear RET program while consuming either the MIS or COL supplement twice daily. Muscle biopsies were taken from the vastus lateralis at baseline and 48 h after a bout of damaging exercise, before and after RET. Muscle tissue was analyzed for SC and myonuclear content, domain, acute SC activation, and fiber cross-sectional area (fCSA). RESULTS MIS resulted in a greater increase in type II fCSA following 10 wk of RET (effect size (ES) = 0.89) but not myonuclear accretion or SC content. Change in myonuclei per fiber was positively correlated with type I and II and total fiber hypertrophy in the COL group only, indicating a robust independent effect of MIS on fCSA. Myonuclear domain increased similarly in both groups, whereas SC domain remained unchanged following RET. SC activation was similar between groups for all fiber types in the untrained state but showed a trend toward greater increases with MIS after RET (ES = 0.70). CONCLUSIONS SC responses to acute damaging exercise and long-term RET are predominantly similar in MIS and COL groups. However, MIS can induce greater increases in type II fCSA with RET and potentially SC activation following damage in the trained state.
Collapse
Affiliation(s)
- Mai Wageh
- Department of Kinesiology, McMaster University, Hamilton, Ontario, CANADA
| | - Stephen A Fortino
- Department of Kinesiology, McMaster University, Hamilton, Ontario, CANADA
| | - Riley Pontello
- Department of Kinesiology, McMaster University, Hamilton, Ontario, CANADA
| | - Ahmed Maklad
- Department of Kinesiology, McMaster University, Hamilton, Ontario, CANADA
| | - Chris McGlory
- Department of Kinesiology, McMaster University, Hamilton, Ontario, CANADA
| | | | - Stuart M Phillips
- Department of Kinesiology, McMaster University, Hamilton, Ontario, CANADA
| | - Gianni Parise
- Department of Kinesiology, McMaster University, Hamilton, Ontario, CANADA
| |
Collapse
|
4
|
Furrer R, Handschin C. Molecular aspects of the exercise response and training adaptation in skeletal muscle. Free Radic Biol Med 2024; 223:53-68. [PMID: 39059515 PMCID: PMC7617583 DOI: 10.1016/j.freeradbiomed.2024.07.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 07/13/2024] [Accepted: 07/21/2024] [Indexed: 07/28/2024]
Abstract
Skeletal muscle plasticity enables an enormous potential to adapt to various internal and external stimuli and perturbations. Most notably, changes in contractile activity evoke a massive remodeling of biochemical, metabolic and force-generating properties. In recent years, a large number of signals, sensors, regulators and effectors have been implicated in these adaptive processes. Nevertheless, our understanding of the molecular underpinnings of training adaptation remains rudimentary. Specifically, the mechanisms that underlie signal integration, output coordination, functional redundancy and other complex traits of muscle adaptation are unknown. In fact, it is even unclear how stimulus-dependent specification is brought about in endurance or resistance exercise. In this review, we will provide an overview on the events that describe the acute perturbations in single endurance and resistance exercise bouts. Furthermore, we will provide insights into the molecular principles of long-term training adaptation. Finally, current gaps in knowledge will be identified, and strategies for a multi-omic and -cellular analyses of the molecular mechanisms of skeletal muscle plasticity that are engaged in individual, acute exercise bouts and chronic training adaptation discussed.
Collapse
Affiliation(s)
- Regula Furrer
- Biozentrum, University of Basel, Spitalstrasse 41, 4056, Basel, Switzerland.
| | - Christoph Handschin
- Biozentrum, University of Basel, Spitalstrasse 41, 4056, Basel, Switzerland.
| |
Collapse
|
5
|
Traversa C. Skeletal Muscle Memory: An Update From the Antidoping Perspective. Drug Test Anal 2024. [PMID: 39317641 DOI: 10.1002/dta.3804] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 08/30/2024] [Accepted: 09/02/2024] [Indexed: 09/26/2024]
Abstract
This narrative review explores the concept of muscle memory, focusing on the physiological and biochemical mechanisms underlying information retention in skeletal muscle tissue as it relates to antidoping. The discussion encompasses the role of satellite cells (SCs) in myonuclei recruitment, resulting in increased myonuclear density and heightened muscle protein turnover. The myonuclear domain theory suggests that myonuclei acquired during hypertrophy may persist, contributing to enhanced muscle protein synthesis (MPS) and potential benefits of muscle memory. The impact of sustained training, protein intake, and resistance exercise on muscle memory, especially in elite athletes, is considered. The review also delves into the influence of anabolic androgenic steroids (AAS) on muscle tissue, highlighting their role in elevating the performance threshold and supporting recovery during intense training through increased muscle protein turnover rates. Additionally, genetic and epigenetic modifications, such as DNA methylation, are explored as potential contributors to muscle memory. The complex interplay of continuous training, AAS use, and genetic factors offers avenues for further research, especially in the context of antidoping efforts. The understanding of muscle memory has implications for maintaining performance gains and addressing ethical challenges in sports.
Collapse
Affiliation(s)
- Claire Traversa
- World Anti-Doping Agency (WADA), Montreal, Quebec, Canada
- Department of Kinesiology and Physical Education, McGill University, Montreal, Quebec, Canada
| |
Collapse
|
6
|
Wen Y, Tian J, Li J, Na X, Yu Z, Zhou W. Developing engineered muscle tissues utilizing standard cell culture plates and mesenchymal stem cell-conditioned medium. Regen Ther 2024; 26:683-692. [PMID: 39286640 PMCID: PMC11403061 DOI: 10.1016/j.reth.2024.08.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 08/08/2024] [Accepted: 08/18/2024] [Indexed: 09/19/2024] Open
Abstract
The construction of engineered muscle tissues that resemble the function and microstructure of human muscles holds significant promise for various applications, including disease modeling, regenerative medicine, and biological machines. However, current muscle tissue engineering approaches often rely on complex equipment which may limit their accessibility and practicality. Herein, we present a convenient approach using a standard 24-well cell culture plate to construct a platform to facilitate engineered muscle tissues formation and culture. Using this platform, engineered muscle tissue with differentiation characteristics can be manufactured in large quantities. Additionally, the mesenchymal stem cell conditioned medium was utilized to promote the formation and functionality of the engineered muscle tissues. The resulting tissues comprised a higher cell density and a better differentiation effect in the tissues. Taken together, this study provides a simple, convenient, and effective platform for studying muscle tissue engineering.
Collapse
Affiliation(s)
- Yihao Wen
- State Key Laboratory of Materials-Oriented Chemical Engineering, College of Chemical Engineering, Nanjing Tech University, Nanjing 211816, China
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China
- Key Laboratory of Biopharmaceutical Preparation and Delivery, Chinese Academy of Sciences, Institute of Process Engineering, Beijing 100190, China
| | - Jia Tian
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China
- Key Laboratory of Biopharmaceutical Preparation and Delivery, Chinese Academy of Sciences, Institute of Process Engineering, Beijing 100190, China
- College of Chemical Engineering, University of the Chinese Academy of Sciences, Beijing 101408, China
| | - Juan Li
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China
- Key Laboratory of Biopharmaceutical Preparation and Delivery, Chinese Academy of Sciences, Institute of Process Engineering, Beijing 100190, China
| | - Xiangming Na
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China
- Key Laboratory of Biopharmaceutical Preparation and Delivery, Chinese Academy of Sciences, Institute of Process Engineering, Beijing 100190, China
| | - Ziyi Yu
- State Key Laboratory of Materials-Oriented Chemical Engineering, College of Chemical Engineering, Nanjing Tech University, Nanjing 211816, China
| | - Weiqing Zhou
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China
- Key Laboratory of Biopharmaceutical Preparation and Delivery, Chinese Academy of Sciences, Institute of Process Engineering, Beijing 100190, China
- College of Chemical Engineering, University of the Chinese Academy of Sciences, Beijing 101408, China
| |
Collapse
|
7
|
Jo SI, Park YS, Chang Y, Moon JH, Lee S, Lee H, Kim M, Kim DY, Bae S, Park SY, Yun H, You JE, Im M, Han HJ, Kim SY, Jin DH. Effects of the purified dry extract of fermented ginseng BST204 on muscle fiber regeneration. Biochem Biophys Rep 2023; 35:101525. [PMID: 37601455 PMCID: PMC10439383 DOI: 10.1016/j.bbrep.2023.101525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2023] [Revised: 07/27/2023] [Accepted: 07/28/2023] [Indexed: 08/22/2023] Open
Abstract
Background Sarcopenia and muscular dystrophy are two muscle diseases. In cancer patients, cancer cachexia induces continuous weight loss and muscle loss due to the disease itself or the use of anticancer drugs. Cachexia occurs in up to 80% of cancer patients. It is recognized as a direct cause of reduced quality of life, contributing to at least 20% of cancer-associated deaths and limiting therapeutic options for cancer patients. Cancer cachexia is associated with multiple chronic or end-stage conditions and develops similarly. There are various options for the treatment of cancer cachexia, but there are still many issues to be solved. Hence, to determine its potential to overcome the muscle wasting during cancer cachexia, we studied the effect of BST204, a refined dry ginseng extract, on muscle fiber regeneration. Experimental procedure We checked the muscle regeneration efficacy of BST204. First, BaCl2 and freeze injury models were selected to investigate muscle regeneration after BST204 administration. In addition, after inducing muscle differentiation of C2C12 cells, the efficacy of BST204 was analyzed. In this model, we analyzed the expression of the signal pathway (PI3K-AKT signal) by Western blot and imaging methods. Results and conclusion These results showed that BST204 induced muscle fiber regeneration in BaCl2 and freeze injury models. Also, we confirmed that BST204 could regulate the PI3K/AKT signaling pathway and regulate the differentiation of C2C12 cells. These results indicate that BST204 has the potential to facilitate the skeletal muscle regeneration during muscle wasting induced by various factors including cancer cachexia.
Collapse
Affiliation(s)
- Su In Jo
- Asan Institute for Life Science, Asan Medical Center, Seoul, Republic of Korea
| | - Yoon Sun Park
- Asan Institute for Life Science, Asan Medical Center, Seoul, Republic of Korea
- Department of Medical Science, AMIST, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Yeeun Chang
- Asan Institute for Life Science, Asan Medical Center, Seoul, Republic of Korea
| | - Jai-Hee Moon
- Asan Institute for Life Science, Asan Medical Center, Seoul, Republic of Korea
| | - Slee Lee
- Asan Institute for Life Science, Asan Medical Center, Seoul, Republic of Korea
| | - Hyejin Lee
- Asan Institute for Life Science, Asan Medical Center, Seoul, Republic of Korea
| | - MiYeon Kim
- Department of Medical Science, AMIST, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Do Yeon Kim
- Asan Institute for Life Science, Asan Medical Center, Seoul, Republic of Korea
- Department of Medical Science, AMIST, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - SangMun Bae
- Asan Institute for Life Science, Asan Medical Center, Seoul, Republic of Korea
| | - Se Yeong Park
- Asan Institute for Life Science, Asan Medical Center, Seoul, Republic of Korea
| | - Hyeseon Yun
- Asan Institute for Life Science, Asan Medical Center, Seoul, Republic of Korea
- Department of Medical Science, AMIST, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Ji-Eun You
- Asan Institute for Life Science, Asan Medical Center, Seoul, Republic of Korea
- Department of Medical Science, AMIST, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Minju Im
- Research Institute, GREENCROSS WellBeing Co., Ltd., Seoul, Republic of Korea
| | - Hae-Jung Han
- Research Institute, GREENCROSS WellBeing Co., Ltd., Seoul, Republic of Korea
| | - Sang-Yeob Kim
- Asan Institute for Life Science, Asan Medical Center, Seoul, Republic of Korea
| | - Dong-Hoon Jin
- Asan Institute for Life Science, Asan Medical Center, Seoul, Republic of Korea
- Department of Pharmacology, AMIST, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
- Department of Convergence Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
8
|
Bersiner K, Park SY, Schaaf K, Yang WH, Theis C, Jacko D, Gehlert S. Resistance exercise: a mighty tool that adapts, destroys, rebuilds and modulates the molecular and structural environment of skeletal muscle. Phys Act Nutr 2023; 27:78-95. [PMID: 37583075 PMCID: PMC10440184 DOI: 10.20463/pan.2023.0021] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 06/29/2023] [Accepted: 06/30/2023] [Indexed: 08/17/2023] Open
Abstract
PURPOSE Skeletal muscle regulates health and performance by maintaining or increasing strength and muscle mass. Although the molecular mechanisms in response to resistance exercise (RE) significantly target the activation of protein synthesis, a plethora of other mechanisms and structures must be involved in orchestrating the communication, repair, and restoration of homeostasis after RE stimulation. In practice, RE can be modulated by variations in intensity, continuity and volume, which affect molecular responses and skeletal muscle adaptation. Knowledge of these aspects is important with respect to planning of training programs and assessing the impact of RE training on skeletal muscle. METHODS In this narrative review, we introduce general aspects of skeletal muscle substructures that adapt in response to RE. We further highlighted the molecular mechanisms that control human skeletal muscle anabolism, degradation, repair and memory in response to acute and repeated RE and linked these aspects to major training variables. RESULTS Although RE is a key stimulus for the activation of skeletal muscle anabolism, it also induces myofibrillar damage. Nevertheless, to increase muscle mass accompanied by a corresponding adaptation of the essential substructures of the sarcomeric environment, RE must be continuously repeated. This requires the permanent engagement of molecular mechanisms that re-establish skeletal muscle integrity after each RE-induced muscle damage. CONCLUSION Various molecular regulators coordinately control the adaptation of skeletal muscle after acute and repeated RE and expand their actions far beyond muscle growth. Variations of key resistance training variables likely affect these mechanisms without affecting muscle growth.
Collapse
Affiliation(s)
- Käthe Bersiner
- Department for Biosciences of Sports, Institute of Sports Science, University of Hildesheim, Hildesheim, Germany
| | - So-Young Park
- Graduate School of Sports Medicine, CHA University, Pocheon, Republic of Korea
| | - Kirill Schaaf
- Department of Molecular and Cellular Sports Medicine, Institute of Cardiovascular Research and Sports Medicine, German Sport University Cologne, Cologne, Germany
| | - Woo-Hwi Yang
- Graduate School of Sports Medicine, CHA University, Pocheon, Republic of Korea
- Department of Medicine, General Graduate School, CHA University, Pocheon, Republic of Korea
| | - Christian Theis
- Center for Anaesthesiology, Helios University Hospital Wuppertal, Wuppertal, Germany
| | - Daniel Jacko
- Department of Molecular and Cellular Sports Medicine, Institute of Cardiovascular Research and Sports Medicine, German Sport University Cologne, Cologne, Germany
| | - Sebastian Gehlert
- Department for Biosciences of Sports, Institute of Sports Science, University of Hildesheim, Hildesheim, Germany
| |
Collapse
|
9
|
Thomas ACQ, Brown A, Hatt AA, Manta K, Costa-Parke A, Kamal M, Joanisse S, McGlory C, Phillips SM, Kumbhare D, Parise G. Short-term aerobic conditioning prior to resistance training augments muscle hypertrophy and satellite cell content in healthy young men and women. FASEB J 2022; 36:e22500. [PMID: 35971745 DOI: 10.1096/fj.202200398rr] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 07/22/2022] [Accepted: 08/02/2022] [Indexed: 11/11/2022]
Abstract
Factors influencing inter-individual variability of responses to resistance training (RT) remain to be fully elucidated. We have proposed the importance of capillarization in skeletal muscle for the satellite cell (SC) response to RT-induced muscle hypertrophy, and hypothesized that aerobic conditioning (AC) would augment RT-induced adaptations. Fourteen healthy young (22 ± 2 years) men and women underwent AC via 6 weeks of unilateral cycling followed by 10 weeks of bilateral RT to investigate how AC alters SC content, activity, and muscle hypertrophy following RT. Muscle biopsies were taken at baseline (unilateral), post AC (bilateral), and post RT (bilateral) in the aerobically conditioned (AC + RT) and unconditioned (RT) legs. Immunofluorescence was used to determine muscle capillarization, fiber size, SC content, and activity. Type I and type II fiber cross-sectional area (CSA) increased following RT, and when legs were analyzed independently, AC + RT increased type I, type II, and mixed-fiber CSA, where the RT leg tended to increase type II (p = .05), but not type I or mixed-fiber CSA. SC content, activation, and differentiation increased with RT, where type I total and quiescent SC content was greater in AC + RT compared to the RT leg. Those with the greatest capillary-to-fiber perimeter exchange index before RT had the greatest change in CSA following RT and a significant relationship was observed between type II fiber capillarization and the change in type II-fiber CSA with RT (r = 0.35). This study demonstrates that AC prior to RT can augment RT-induced muscle adaptions and that these differences are associated with increases in capillarization.
Collapse
Affiliation(s)
- Aaron C Q Thomas
- Exercise Metabolism Research Group, Department of Kinesiology, McMaster University, Hamilton, Ontario, Canada
| | - Alex Brown
- Exercise Metabolism Research Group, Department of Kinesiology, McMaster University, Hamilton, Ontario, Canada
| | - Aidan A Hatt
- Exercise Metabolism Research Group, Department of Kinesiology, McMaster University, Hamilton, Ontario, Canada
| | - Katherine Manta
- Exercise Metabolism Research Group, Department of Kinesiology, McMaster University, Hamilton, Ontario, Canada
| | - Anamaria Costa-Parke
- Exercise Metabolism Research Group, Department of Kinesiology, McMaster University, Hamilton, Ontario, Canada
| | - Michael Kamal
- Exercise Metabolism Research Group, Department of Kinesiology, McMaster University, Hamilton, Ontario, Canada
| | - Sophie Joanisse
- Exercise Metabolism Research Group, Department of Kinesiology, McMaster University, Hamilton, Ontario, Canada.,Musculoskeletal Sciences and Sport Medicine Research Centre, Department of Sport and Exercise Sciences, Faculty of Science and Engineering, Manchester Metropolitan University, Manchester, UK.,Manchester Metropolitan University Institute of Sport, Manchester, UK
| | - Chris McGlory
- School of Kinesiology and Health Studies, Queen's University, Kingston, Ontario, Canada.,Department of Medicine, Queen's University, Kingston, Ontario, Canada
| | - Stuart M Phillips
- Exercise Metabolism Research Group, Department of Kinesiology, McMaster University, Hamilton, Ontario, Canada
| | | | - Gianni Parise
- Exercise Metabolism Research Group, Department of Kinesiology, McMaster University, Hamilton, Ontario, Canada
| |
Collapse
|
10
|
Shi LL, Zhu KC, Wang HL. Characterization of myogenic regulatory factors, myod and myf5 from Megalobrama amblycephala and the effect of lipopolysaccharide on satellite cells in skeletal muscle. Gene 2022; 834:146608. [PMID: 35659893 DOI: 10.1016/j.gene.2022.146608] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2021] [Revised: 04/27/2022] [Accepted: 05/18/2022] [Indexed: 11/16/2022]
Abstract
Myod and Myf5 are muscle-specific basic helix-loop-helix (bHLH) transcription factors that play essential roles in regulating skeletal muscle development and growth. In order to investigate potential function of myod and myf5 of Megalobrama amblycephala, an economically important freshwater fish species, in the present study, we characterized the sequences and expression profiles of M. amblycephala myod and myf5. The open reading frame (ORF) sequences of myod and myf5 encoded 275 and 240 amino acids, respectively, possessing analogous structure with the highly conserved domains, bHLH and C-terminal helix III domains. Spatio-temporal expression patterns revealed that myod and myf5 were predominant in skeletal muscle with the highest expression in white muscle, and the highest at 10 days post-hatching (dph) and the segmentation period, respectively. Furthermore, we evaluated the effects of lipopolysaccharide (LPS) on the expression of muscle-related genes in white and red muscle, and proliferation and differentiation of satellite cells. The myod, myf5 and pax-7 expression generally increased and then decreased with increase of LPS concentration and treatment time in red muscle, while these genes showed inconsistent expression patterns in white muscle. In addition, LPS administration caused the frequency increase of satellite cells in red and white muscle especially at 3 and 7 days after LPS-injection.
Collapse
Affiliation(s)
- Lin-Lin Shi
- Key Lab of Freshwater Animal Breeding, Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction, Ministry of Education, College of Fishery, Huazhong Agricultural University, 430070 Wuhan, PR China
| | - Ke-Cheng Zhu
- Key Laboratory of South China Sea Fishery Resources Exploitation and Utilization, Ministry of Agriculture and Rural Affairs, South China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, 510300 Guangzhou, Guangdong Province, PR China
| | - Huan-Ling Wang
- Key Lab of Freshwater Animal Breeding, Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction, Ministry of Education, College of Fishery, Huazhong Agricultural University, 430070 Wuhan, PR China.
| |
Collapse
|
11
|
Risha MA, Ali A, Siengdee P, Trakooljul N, Dannenberger D, Wimmers K, Ponsuksili S. Insights into molecular pathways and fatty acid membrane composition during the temperature stress response in the murine C2C12 cell model. THE SCIENCE OF THE TOTAL ENVIRONMENT 2022; 807:151019. [PMID: 34662617 DOI: 10.1016/j.scitotenv.2021.151019] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 10/11/2021] [Accepted: 10/12/2021] [Indexed: 06/13/2023]
Abstract
Daily and seasonal temperature fluctuations are inevitable due to climate change, which highlights the importance of studying the detrimental effects of temperature fluctuations on the health, productivity, and product quality of farm animals. Muscle membrane composition and the molecular signals are vital for muscle cell differentiation and muscle growth, but their response to temperature stress is not well characterized. Temperature changes can lead to modification of membrane components of the cell, which may affect its surroundings and intracellular signaling pathways. Using C2C12 myoblast cells as a model of skeletal muscle development, this study was designed to investigate the effects of high temperature (39 °C and 41 °C) and low temperature (35 °C) on molecular pathways in the cells as well as the cell membrane fatty acid composition. Our results show that several genes were differentially expressed in C2C12 cells cultured under heat or cold stress, and these genes were enriched important KEGG pathways including PI3K-Akt signaling pathway, lysosome and HIF- signaling pathway, Wnt signaling pathway and AMPK signaling pathway. Our analysis further reveals that several membrane transporters and genes involved in lipid metabolism and fatty acid elongation were also differentially expressed in C2C12 cells cultured under high or low temperature. Additionally, temperature stress shifts the fatty acid composition in the cell membranes, including the proportion of saturated, monounsaturated and polyunsaturated fatty acids. This study revealed an interference between fatty acid composition in the membranes and changing molecular pathways including lipid metabolism and fatty acids elongation mediated under thermal stress. These findings will reinforce a better understanding of the adaptive mechanisms in skeletal muscle under temperature stress.
Collapse
Affiliation(s)
- Marua Abu Risha
- Leibniz Institute for Farm Animal Biology (FBN), Institute of Genome Biology, Functional Genome Analysis Research Unit, Wilhelm-Stahl-Allee 2, D-18196 Dummerstorf, Germany
| | - Asghar Ali
- Leibniz Institute for Farm Animal Biology (FBN), Institute of Genome Biology, Functional Genome Analysis Research Unit, Wilhelm-Stahl-Allee 2, D-18196 Dummerstorf, Germany
| | - Puntita Siengdee
- Leibniz Institute for Farm Animal Biology (FBN), Institute of Genome Biology, Functional Genome Analysis Research Unit, Wilhelm-Stahl-Allee 2, D-18196 Dummerstorf, Germany
| | - Nares Trakooljul
- Leibniz Institute for Farm Animal Biology (FBN), Institute of Genome Biology, Genomics Research Unit, Wilhelm-Stahl-Allee 2, D-18196 Dummerstorf, Germany
| | - Dirk Dannenberger
- Leibniz Institute for Farm Animal Biology (FBN), Institute of Muscle Biology and Growth, Lipid metabolism and muscular adaptation workgroup, Wilhelm-Stahl-Allee 2, D-18196 Dummerstorf, Germany
| | - Klaus Wimmers
- Leibniz Institute for Farm Animal Biology (FBN), Institute of Genome Biology, Genomics Research Unit, Wilhelm-Stahl-Allee 2, D-18196 Dummerstorf, Germany; Faculty of Agricultural and Environmental Sciences, University Rostock, 18059 Rostock, Germany
| | - Siriluck Ponsuksili
- Leibniz Institute for Farm Animal Biology (FBN), Institute of Genome Biology, Functional Genome Analysis Research Unit, Wilhelm-Stahl-Allee 2, D-18196 Dummerstorf, Germany.
| |
Collapse
|
12
|
Huang H, Xing D, Zhang Q, Li H, Lin J, He Z, Lin J. LncRNAs as a new regulator of chronic musculoskeletal disorder. Cell Prolif 2021; 54:e13113. [PMID: 34498342 PMCID: PMC8488571 DOI: 10.1111/cpr.13113] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2021] [Revised: 06/15/2021] [Accepted: 07/30/2021] [Indexed: 01/15/2023] Open
Abstract
OBJECTIVES In recent years, long non-coding RNAs (lncRNAs) have been found to play a role in the occurrence, progression and prognosis of chronic musculoskeletal disorders. DESIGN AND METHODS Literature exploring on PubMed was conducted using the combination of keywords 'LncRNA' and each of the following: 'osteoarthritis', 'rheumatoid arthritis', 'osteoporosis', 'osteogenesis', 'osteoclastogenesis', 'gout arthritis', 'Kashin-Beck disease', 'ankylosing spondylitis', 'cervical spondylotic myelopathy', 'intervertebral disc degeneration', 'human muscle disease' and 'muscle hypertrophy and atrophy'. For each disorder, we focused on the publications in the last five years (5/1/2016-2021/5/1, except for Kashin-Beck disease). Finally, we excluded publications that had been reported in reviews of various musculoskeletal disorders during the last three years. Here, we summarized the progress of research on the role of lncRNA in multiple pathological processes during musculoskeletal disorders. RESULTS LncRNAs play a crucial role in regulating downstream gene expression and maintaining function and homeostasis of cells, especially in chondrocytes, synovial cells, osteoblasts, osteoclasts and skeletal muscle cells. CONCLUSIONS Understanding the mechanisms of lncRNAs in musculoskeletal disorders may provide promising strategies for clinical practice.
Collapse
Affiliation(s)
- Hesuyuan Huang
- Arthritis Clinic & Research CenterPeking University People's Hospital, Peking UniversityBeijingChina
- Arthritis InstitutePeking UniversityBeijingChina
| | - Dan Xing
- Arthritis Clinic & Research CenterPeking University People's Hospital, Peking UniversityBeijingChina
- Arthritis InstitutePeking UniversityBeijingChina
| | - Qingxi Zhang
- Arthritis Clinic & Research CenterPeking University People's Hospital, Peking UniversityBeijingChina
- Arthritis InstitutePeking UniversityBeijingChina
| | - Hui Li
- Arthritis Clinic & Research CenterPeking University People's Hospital, Peking UniversityBeijingChina
- Arthritis InstitutePeking UniversityBeijingChina
| | - Jianjing Lin
- Arthritis Clinic & Research CenterPeking University People's Hospital, Peking UniversityBeijingChina
- Arthritis InstitutePeking UniversityBeijingChina
| | - Zihao He
- Arthritis Clinic & Research CenterPeking University People's Hospital, Peking UniversityBeijingChina
- Arthritis InstitutePeking UniversityBeijingChina
| | - Jianhao Lin
- Arthritis Clinic & Research CenterPeking University People's Hospital, Peking UniversityBeijingChina
- Arthritis InstitutePeking UniversityBeijingChina
| |
Collapse
|
13
|
Skeletal muscle heme oxygenase-1 activity regulates aerobic capacity. Cell Rep 2021; 35:109018. [PMID: 33882313 PMCID: PMC8196422 DOI: 10.1016/j.celrep.2021.109018] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 03/08/2021] [Accepted: 03/30/2021] [Indexed: 11/23/2022] Open
Abstract
Physical exercise has profound effects on quality of life and susceptibility to chronic disease; however, the regulation of skeletal muscle function at the molecular level after exercise remains unclear. We tested the hypothesis that the benefits of exercise on muscle function are linked partly to microtraumatic events that result in accumulation of circulating heme. Effective metabolism of heme is controlled by Heme Oxygenase-1 (HO-1, Hmox1), and we find that mouse skeletal muscle-specific HO-1 deletion (Tam-Cre-HSA-Hmox1fl/fl) shifts the proportion of muscle fibers from type IIA to type IIB concomitant with a disruption in mitochondrial content and function. In addition to a significant impairment in running performance and response to exercise training, Tam-Cre-HSA-Hmox1fl/fl mice show remarkable muscle atrophy compared to Hmox1fl/fl controls. Collectively, these data define a role for heme and HO-1 as central regulators in the physiologic response of skeletal muscle to exercise.
Collapse
|
14
|
Wong A, Garcia SM, Tamaki S, Striedinger K, Barruet E, Hansen SL, Young DM, Pomerantz JH. Satellite cell activation and retention of muscle regenerative potential after long-term denervation. Stem Cells 2021; 39:331-344. [PMID: 33326654 DOI: 10.1002/stem.3316] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Accepted: 12/01/2020] [Indexed: 12/17/2022]
Abstract
Irreversible denervation atrophy remains an unsolved clinical problem, and the role of skeletal muscle stem cell (MuSC, satellite cell) depletion in this process is unclear. We investigated the ability of MuSCs to regenerate muscle in the context of denervation. Three to 12 months following sciatic denervation in mice, MuSC number, size, EdU uptake, rate of division, and mitochondrial activity were increased. Following acute myotoxin injury, denervated muscles formed new muscle fibers in situ. MuSCs isolated via flow cytometry from denervated mouse muscle, or from atrophic denervated gluteus maximus muscles of humans with complete spinal cord injuries two decades prior, formed new muscle fibers and reoccupied the anatomic niche after transplantation into uninjured muscle. Our results show unequivocally that, even after prolonged denervation, MuSCs retain intrinsic regenerative potential similar to that of uninjured MuSCs. Treatment of denervation atrophy will require elucidating the non-MuSC environmental changes in muscle that prevent functional regeneration.
Collapse
Affiliation(s)
- Alvin Wong
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Program in Craniofacial Biology, Eli and Edythe Broad Center of Regeneration Medicine, University of California, San Francisco, California, USA
| | - Steven M Garcia
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Program in Craniofacial Biology, Eli and Edythe Broad Center of Regeneration Medicine, University of California, San Francisco, California, USA
| | - Stanley Tamaki
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Program in Craniofacial Biology, Eli and Edythe Broad Center of Regeneration Medicine, University of California, San Francisco, California, USA
| | - Katharine Striedinger
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Program in Craniofacial Biology, Eli and Edythe Broad Center of Regeneration Medicine, University of California, San Francisco, California, USA
| | - Emilie Barruet
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Program in Craniofacial Biology, Eli and Edythe Broad Center of Regeneration Medicine, University of California, San Francisco, California, USA
| | - Scott L Hansen
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Program in Craniofacial Biology, Eli and Edythe Broad Center of Regeneration Medicine, University of California, San Francisco, California, USA
| | - David M Young
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Program in Craniofacial Biology, Eli and Edythe Broad Center of Regeneration Medicine, University of California, San Francisco, California, USA
| | - Jason H Pomerantz
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Program in Craniofacial Biology, Eli and Edythe Broad Center of Regeneration Medicine, University of California, San Francisco, California, USA
- Department of Orofacial Sciences, University of California, San Francisco, California, USA
| |
Collapse
|
15
|
Bass JJ, Nakhuda A, Deane CS, Brook MS, Wilkinson DJ, Phillips BE, Philp A, Tarum J, Kadi F, Andersen D, Garcia AM, Smith K, Gallagher IJ, Szewczyk NJ, Cleasby ME, Atherton PJ. Overexpression of the vitamin D receptor (VDR) induces skeletal muscle hypertrophy. Mol Metab 2020; 42:101059. [PMID: 32771696 PMCID: PMC7475200 DOI: 10.1016/j.molmet.2020.101059] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 07/23/2020] [Accepted: 07/28/2020] [Indexed: 12/30/2022] Open
Abstract
OBJECTIVE The Vitamin D receptor (VDR) has been positively associated with skeletal muscle mass, function and regeneration. Mechanistic studies have focused on the loss of the receptor, with in vivo whole-body knockout models demonstrating reduced myofibre size and function and impaired muscle development. To understand the mechanistic role upregulation of the VDR elicits in muscle mass/health, we studied the impact of VDR over-expression (OE) in vivo before exploring the importance of VDR expression upon muscle hypertrophy in humans. METHODS Wistar rats underwent in vivo electrotransfer (IVE) to overexpress the VDR in the Tibialis anterior (TA) muscle for 10 days, before comprehensive physiological and metabolic profiling to characterise the influence of VDR-OE on muscle protein synthesis (MPS), anabolic signalling and satellite cell activity. Stable isotope tracer (D2O) techniques were used to assess sub-fraction protein synthesis, alongside RNA-Seq analysis. Finally, human participants underwent 20 wks of resistance exercise training, with body composition and transcriptomic analysis. RESULTS Muscle VDR-OE yielded total protein and RNA accretion, manifesting in increased myofibre area, i.e., hypertrophy. The observed increases in MPS were associated with enhanced anabolic signalling, reflecting translational efficiency (e.g., mammalian target of rapamycin (mTOR-signalling), with no effects upon protein breakdown markers being observed. Additionally, RNA-Seq illustrated marked extracellular matrix (ECM) remodelling, while satellite cell content, markers of proliferation and associated cell-cycled related gene-sets were upregulated. Finally, induction of VDR mRNA correlated with muscle hypertrophy in humans following long-term resistance exercise type training. CONCLUSION VDR-OE stimulates muscle hypertrophy ostensibly via heightened protein synthesis, translational efficiency, ribosomal expansion and upregulation of ECM remodelling-related gene-sets. Furthermore, VDR expression is a robust marker of the hypertrophic response to resistance exercise in humans. The VDR is a viable target of muscle maintenance through testable Vitamin D molecules, as active molecules and analogues.
Collapse
Affiliation(s)
- Joseph J Bass
- MRC/ARUK Centre for Musculoskeletal Ageing Research and National Institute for Health Research (NIHR), Nottingham Biomedical Research Centre (BRC), School of Medicine, University of Nottingham, DE22 3DT, UK
| | - Asif Nakhuda
- MRC/ARUK Centre for Musculoskeletal Ageing Research and National Institute for Health Research (NIHR), Nottingham Biomedical Research Centre (BRC), School of Medicine, University of Nottingham, DE22 3DT, UK
| | - Colleen S Deane
- Department of Sport and Health Sciences, University of Exeter, EX1 2LU, UK
| | - Matthew S Brook
- MRC/ARUK Centre for Musculoskeletal Ageing Research and National Institute for Health Research (NIHR), Nottingham Biomedical Research Centre (BRC), School of Medicine, University of Nottingham, DE22 3DT, UK
| | - Daniel J Wilkinson
- MRC/ARUK Centre for Musculoskeletal Ageing Research and National Institute for Health Research (NIHR), Nottingham Biomedical Research Centre (BRC), School of Medicine, University of Nottingham, DE22 3DT, UK
| | - Bethan E Phillips
- MRC/ARUK Centre for Musculoskeletal Ageing Research and National Institute for Health Research (NIHR), Nottingham Biomedical Research Centre (BRC), School of Medicine, University of Nottingham, DE22 3DT, UK
| | - Andrew Philp
- Mitochondrial Metabolism and Ageing Laboratory, Diabetes and Metabolism Division, Garvan Institute of Medical Research, NSW, 2010, Australia; School of Sport, Exercise and Rehabilitation Sciences, University of Birmingham, B15 2TT, UK
| | - Janelle Tarum
- School of Health Sciences, Örebro University, 70182, Sweden
| | - Fawzi Kadi
- School of Health Sciences, Örebro University, 70182, Sweden
| | - Ditte Andersen
- Molecular Physiology of Diabetes Laboratory, Dept. of Comparative Biomedical Sciences, Royal Veterinary College, NW1 0TU, UK
| | - Amadeo Muñoz Garcia
- Institute of Metabolism and Systems Research, The University of Birmingham, Birmingham, UK; Department of Bioinformatics - BiGCaT, NUTRIM School of Nutrition and Metabolism in Translational Research, Maastricht University, Maastricht, the Netherlands
| | - Ken Smith
- MRC/ARUK Centre for Musculoskeletal Ageing Research and National Institute for Health Research (NIHR), Nottingham Biomedical Research Centre (BRC), School of Medicine, University of Nottingham, DE22 3DT, UK
| | - Iain J Gallagher
- Physiology, Exercise and Nutrition Research Group, Faculty of Health Sciences and Sport, University of Stirling, FK9 4LA, UK
| | - Nathaniel J Szewczyk
- MRC/ARUK Centre for Musculoskeletal Ageing Research and National Institute for Health Research (NIHR), Nottingham Biomedical Research Centre (BRC), School of Medicine, University of Nottingham, DE22 3DT, UK
| | - Mark E Cleasby
- Molecular Physiology of Diabetes Laboratory, Dept. of Comparative Biomedical Sciences, Royal Veterinary College, NW1 0TU, UK
| | - Philip J Atherton
- MRC/ARUK Centre for Musculoskeletal Ageing Research and National Institute for Health Research (NIHR), Nottingham Biomedical Research Centre (BRC), School of Medicine, University of Nottingham, DE22 3DT, UK.
| |
Collapse
|
16
|
Bodine SC. Edward F. Adolph Distinguished Lecture. Skeletal muscle atrophy: Multiple pathways leading to a common outcome. J Appl Physiol (1985) 2020; 129:272-282. [PMID: 32644910 PMCID: PMC7473948 DOI: 10.1152/japplphysiol.00381.2020] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 07/02/2020] [Accepted: 07/03/2020] [Indexed: 12/14/2022] Open
Abstract
Skeletal muscle atrophy continues to be a serious consequence of many diseases and conditions for which there is no treatment. Our understanding of the mechanisms regulating skeletal muscle mass has improved considerably over the past two decades. For many years it was known that skeletal muscle atrophy resulted from an imbalance between protein synthesis and protein breakdown, with the net balance shifting toward protein breakdown. However, the molecular and cellular mechanisms underlying the increased breakdown of myofibrils was unknown. Over the past two decades, numerous reports have identified novel genes and signaling pathways that are upregulated and activated in response to stimuli such as disuse, inflammation, metabolic stress, starvation and others that induce muscle atrophy. This review summarizes the discovery efforts performed in the identification of several pathways involved in the regulation of skeletal muscle mass: the mammalian target of rapamycin (mTORC1) and the ubiquitin proteasome pathway and the E3 ligases, MuRF1 and MAFbx. While muscle atrophy is a common outcome of many diseases, it is doubtful that a single gene or pathway initiates or mediates the breakdown of myofibrils. Interestingly, however, is the observation that upregulation of the E3 ligases, MuRF1 and MAFbx, is a common feature of many divergent atrophy conditions. The challenge for the field of muscle biology is to understand how all of the various molecules, transcription factors, and signaling pathways interact to produce muscle atrophy and to identify the critical factors for intervention.
Collapse
Affiliation(s)
- Sue C Bodine
- Department of Internal Medicine/Endocrinology and Metabolism, University of Iowa Carver College of Medicine, Iowa City, Iowa
| |
Collapse
|
17
|
Angulo J, El Assar M, Álvarez-Bustos A, Rodríguez-Mañas L. Physical activity and exercise: Strategies to manage frailty. Redox Biol 2020; 35:101513. [PMID: 32234291 PMCID: PMC7284931 DOI: 10.1016/j.redox.2020.101513] [Citation(s) in RCA: 366] [Impact Index Per Article: 73.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Revised: 03/13/2020] [Accepted: 03/17/2020] [Indexed: 12/25/2022] Open
Abstract
Frailty, a consequence of the interaction of the aging process and certain chronic diseases, compromises functional outcomes in the elderly and substantially increases their risk for developing disabilities and other adverse outcomes. Frailty follows from the combination of several impaired physiological mechanisms affecting multiple organs and systems. And, though frailty and sarcopenia are related, they are two different conditions. Thus, strategies to preserve or improve functional status should consider systemic function in addition to muscle conditioning. Physical activity/exercise is considered one of the main strategies to counteract frailty-related physical impairment in the elderly. Exercise reduces age-related oxidative damage and chronic inflammation, increases autophagy, and improves mitochondrial function, myokine profile, insulin-like growth factor-1 (IGF-1) signaling pathway, and insulin sensitivity. Exercise interventions target resistance (strength and power), aerobic, balance, and flexibility work. Each type improves different aspects of physical functioning, though they could be combined according to need and prescribed as a multicomponent intervention. Therefore, exercise intervention programs should be prescribed based on an individual's physical functioning and adapted to the ensuing response.
Collapse
Affiliation(s)
- Javier Angulo
- Servicio de Histología-Investigación, Unidad de Investigación Traslacional en Cardiología (IRYCIS-UFV), Hospital Universitario Ramón y Cajal, Madrid, Spain; Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, Madrid, Spain
| | - Mariam El Assar
- Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, Madrid, Spain; Fundación para la Investigación Biomédica del Hospital Universitario de Getafe, Getafe, Spain
| | | | - Leocadio Rodríguez-Mañas
- Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, Madrid, Spain; Servicio de Geriatría, Hospital Universitario de Getafe, Getafe, Spain.
| |
Collapse
|
18
|
Forcina L, Cosentino M, Musarò A. Mechanisms Regulating Muscle Regeneration: Insights into the Interrelated and Time-Dependent Phases of Tissue Healing. Cells 2020; 9:E1297. [PMID: 32456017 PMCID: PMC7290814 DOI: 10.3390/cells9051297] [Citation(s) in RCA: 130] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2020] [Revised: 05/18/2020] [Accepted: 05/20/2020] [Indexed: 12/12/2022] Open
Abstract
Despite a massive body of knowledge which has been produced related to the mechanisms guiding muscle regeneration, great interest still moves the scientific community toward the study of different aspects of skeletal muscle homeostasis, plasticity, and regeneration. Indeed, the lack of effective therapies for several physiopathologic conditions suggests that a comprehensive knowledge of the different aspects of cellular behavior and molecular pathways, regulating each regenerative stage, has to be still devised. Hence, it is important to perform even more focused studies, taking the advantage of robust markers, reliable techniques, and reproducible protocols. Here, we provide an overview about the general aspects of muscle regeneration and discuss the different approaches to study the interrelated and time-dependent phases of muscle healing.
Collapse
Affiliation(s)
| | | | - Antonio Musarò
- Laboratory affiliated to Istituto Pasteur Italia—Fondazione Cenci Bolognetti, DAHFMO-Unit of Histology and Medical Embryology, Sapienza University of Rome, Via Antonio Scarpa, 14, 00161 Rome, Italy; (L.F.); (M.C.)
| |
Collapse
|
19
|
Abstract
Individuals that maintain healthy skeletal tissue tend to live healthier, happier lives as proper muscle function enables maintenance of independence and actuation of autonomy. The onset of skeletal muscle decline begins around the age of 30, and muscle atrophy is associated with a number of serious morbidities and mortalities. Satellite cells are responsible for regeneration of skeletal muscle and enter a reversible non-dividing state of quiescence under homeostatic conditions. In response to injury, satellite cells are able to activate and re-enter the cell cycle, creating new cells to repair and create nascent muscle fibres while preserving a small population that can return to quiescence for future regenerative demands. However, in aged muscle, satellite cells that experience prolonged quiescence will undergo programmed cellular senescence, an irreversible non-dividing state that handicaps the regenerative capabilities of muscle. This review examines how periodic activation and cycling of satellite cells through exercise can mitigate senescence acquisition and myogenic decline.
Collapse
Affiliation(s)
- William Chen
- Sprott Centre for Stem Cell Research, Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada K1H 8L6.,Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada K1H 8M5
| | - David Datzkiw
- Sprott Centre for Stem Cell Research, Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada K1H 8L6.,Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada K1H 8M5
| | - Michael A Rudnicki
- Sprott Centre for Stem Cell Research, Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada K1H 8L6.,Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada K1H 8M5
| |
Collapse
|
20
|
Gomes MJ, Pagan LU, Lima ARR, Reyes DRA, Martinez PF, Damatto FC, Pontes THD, Rodrigues EA, Souza LM, Tosta IF, Fernandes AAH, Zornoff LAM, Okoshi K, Okoshi MP. Effects of aerobic and resistance exercise on cardiac remodelling and skeletal muscle oxidative stress of infarcted rats. J Cell Mol Med 2020; 24:5352-5362. [PMID: 32239667 PMCID: PMC7205792 DOI: 10.1111/jcmm.15191] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Revised: 01/21/2020] [Accepted: 02/22/2020] [Indexed: 12/22/2022] Open
Abstract
We compared the influence of aerobic and resistance exercise on cardiac remodelling, physical capacity and skeletal muscle oxidative stress in rats with MI‐induced heart failure. Three months after MI induction, Wistar rats were divided into four groups: Sham; sedentary MI (S‐MI); aerobic exercised MI (A‐MI); and resistance exercised MI (R‐MI). Exercised rats trained three times a week for 12 weeks on a treadmill or ladder. Statistical analysis was performed by ANOVA or Kruskal‐Wallis test. Functional aerobic capacity was greater in A‐MI and strength gain higher in R‐MI. Echocardiographic parameters did not differ between infarct groups. Reactive oxygen species production, evaluated by fluorescence, was higher in S‐MI than Sham, and lipid hydroperoxide concentration was lower in A‐MI than the other groups. Glutathione peroxidase activity was higher in A‐MI than S‐MI and R‐MI. Superoxide dismutase was lower in S‐MI than Sham and R‐MI. Gastrocnemius cross‐sectional area, satellite cell activation and expression of the ubiquitin‐proteasome system proteins did not differ between groups. In conclusion, aerobic exercise and resistance exercise improve functional capacity and maximum load carrying, respectively, without changing cardiac remodelling in infarcted rats. In the gastrocnemius, infarction increases oxidative stress and changes antioxidant enzyme activities. Aerobic exercise reduces oxidative stress and attenuates superoxide dismutase and glutathione peroxidase changes.
Collapse
Affiliation(s)
- Mariana J Gomes
- Botucatu Medical School, Sao Paulo State University, UNESP, Botucatu, Brazil
| | - Luana U Pagan
- Botucatu Medical School, Sao Paulo State University, UNESP, Botucatu, Brazil
| | - Aline R R Lima
- Botucatu Medical School, Sao Paulo State University, UNESP, Botucatu, Brazil
| | - David R A Reyes
- Botucatu Medical School, Sao Paulo State University, UNESP, Botucatu, Brazil
| | - Paula F Martinez
- School of Physical Therapy, Federal University of Mato Grosso do Sul, Campo Grande, Brazil
| | - Felipe C Damatto
- Botucatu Medical School, Sao Paulo State University, UNESP, Botucatu, Brazil
| | - Thierres H D Pontes
- Botucatu Medical School, Sao Paulo State University, UNESP, Botucatu, Brazil
| | - Eder A Rodrigues
- Botucatu Medical School, Sao Paulo State University, UNESP, Botucatu, Brazil
| | - Lidiane M Souza
- Botucatu Medical School, Sao Paulo State University, UNESP, Botucatu, Brazil
| | - Ingrid F Tosta
- Institute of Biosciences, Sao Paulo State University, UNESP, Botucatu, Brazil
| | - Ana A H Fernandes
- Institute of Biosciences, Sao Paulo State University, UNESP, Botucatu, Brazil
| | | | - Katashi Okoshi
- Botucatu Medical School, Sao Paulo State University, UNESP, Botucatu, Brazil
| | - Marina P Okoshi
- Botucatu Medical School, Sao Paulo State University, UNESP, Botucatu, Brazil
| |
Collapse
|
21
|
Beiter T, Nieß AM, Moser D. Transcriptional memory in skeletal muscle. Don't forget (to) exercise. J Cell Physiol 2020; 235:5476-5489. [PMID: 31967338 DOI: 10.1002/jcp.29535] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Accepted: 01/09/2020] [Indexed: 12/29/2022]
Abstract
Transcriptional memory describes an ancient and highly conserved form of cellular learning that enables cells to benefit from recent experience by retaining a mitotically inheritable but reversible memory of the initial transcriptional response when encountering an environmental or physiological stimulus. Herein, we will review recent progress made in the understanding of how cells can make use of diverse constituents of the epigenetic toolbox to retain a transcriptional memory of past states and perturbations. Specifically, we will outline how these mechanisms will help to improve our understanding of skeletal muscle plasticity in health and disease. We describe the epigenetic road map that allows skeletal muscle fibers to navigate through training-induced adaptation processes, and how epigenetic memory marks can preserve an autobiographical history of lifestyle behavior changes, pathological challenges, and aging. We will further consider some key findings in the field of exercise epigenomics to emphasize major challenges when interpreting dynamic changes in the chromatin landscape in response to acute exercise and training.
Collapse
Affiliation(s)
- Thomas Beiter
- Department of Sports Medicine, University Hospital Tübingen, Tübingen, Germany
| | - Andreas M Nieß
- Department of Sports Medicine, University Hospital Tübingen, Tübingen, Germany
| | - Dirk Moser
- Department of Genetic Psychology, Faculty of Psychology, Ruhr-University Bochum, Bochum, Germany
| |
Collapse
|
22
|
Chang EI, Rozance PJ, Wesolowski SR, Nguyen LM, Shaw SC, Sclafani RA, Bjorkman KK, Peter AK, Hay WW, Brown LD. Rates of myogenesis and myofiber numbers are reduced in late gestation IUGR fetal sheep. J Endocrinol 2019; 244:339-352. [PMID: 31751294 PMCID: PMC7192794 DOI: 10.1530/joe-19-0273] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Accepted: 11/12/2019] [Indexed: 12/14/2022]
Abstract
Intrauterine growth-restricted (IUGR) fetuses are born with reduced skeletal muscle mass. We hypothesized that reduced rates of myogenesis would contribute to fewer and smaller myofibers in IUGR fetal hindlimb muscle compared to the normally growing fetus. We tested this hypothesis in IUGR fetal sheep with progressive placental insufficiency produced by exposing pregnant ewes to elevated ambient temperatures from 38 to 116 days gestation (dGA; term = 147 dGA). Surgically catheterized control (CON, n = 8) and IUGR (n = 13) fetal sheep were injected with intravenous 5-bromo-2′-deoxyuridine (BrdU) prior to muscle collection (134 dGA). Rates of myogenesis, defined as the combined processes of myoblast proliferation, differentiation, and fusion into myofibers, were determined in biceps femoris (BF), tibialis anterior (TA), and flexor digitorum superficialis (FDS) muscles. Total myofiber number was determined for the entire cross-section of the FDS muscle. In IUGR fetuses, the number of BrdU+ myonuclei per myofiber cross-section was lower in BF, TA, and FDS (P < 0.05), total myonuclear number per myofiber cross-section was lower in BF and FDS (P < 0.05), and total myofiber number was lower in FDS (P < 0.005) compared to CON. mRNA expression levels of cyclins, cyclin-dependent protein kinases, and myogenic regulatory factors were lower (P < 0.05), and inhibitors of the cell cycle were higher (P < 0.05) in IUGR BF compared to CON. Markers of apoptosis were not different in IUGR BF muscle. These results show that in IUGR fetuses, reduced rates of myogenesis produce fewer numbers of myonuclei, which may limit hypertrophic myofiber growth. Fewer myofibers of smaller size contribute to smaller muscle mass in the IUGR fetus.
Collapse
Affiliation(s)
- Eileen I. Chang
- Department of Pediatrics, University of Colorado School of Medicine, Perinatal Research Center, Aurora, Colorado, USA
| | - Paul J. Rozance
- Department of Pediatrics, University of Colorado School of Medicine, Perinatal Research Center, Aurora, Colorado, USA
| | - Stephanie R. Wesolowski
- Department of Pediatrics, University of Colorado School of Medicine, Perinatal Research Center, Aurora, Colorado, USA
| | - Leanna M. Nguyen
- Department of Pediatrics, University of Colorado School of Medicine, Perinatal Research Center, Aurora, Colorado, USA
| | - Steven C. Shaw
- Department of Pediatrics, University of Colorado School of Medicine, Perinatal Research Center, Aurora, Colorado, USA
| | - Robert A. Sclafani
- Department of Biochemistry and Molecular Genetics, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Kristen K. Bjorkman
- Department of Molecular, Cellular and Developmental Biology and BioFrontiers Institute, University of Colorado Boulder, Boulder, Colorado, USA
| | - Angela K. Peter
- Department of Molecular, Cellular and Developmental Biology and BioFrontiers Institute, University of Colorado Boulder, Boulder, Colorado, USA
| | - William W. Hay
- Department of Pediatrics, University of Colorado School of Medicine, Perinatal Research Center, Aurora, Colorado, USA
| | - Laura D. Brown
- Department of Pediatrics, University of Colorado School of Medicine, Perinatal Research Center, Aurora, Colorado, USA
| |
Collapse
|
23
|
Brearley MC, Li C, Daniel ZC, Loughna PT, Parr T, Brameld JM. Changes in expression of serine biosynthesis and integrated stress response genes during myogenic differentiation of C2C12 cells. Biochem Biophys Rep 2019; 20:100694. [PMID: 31681859 PMCID: PMC6818154 DOI: 10.1016/j.bbrep.2019.100694] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Revised: 08/05/2019] [Accepted: 10/02/2019] [Indexed: 01/13/2023] Open
Abstract
Skeletal muscle is a highly metabolic and dynamic tissue that is formed through the complex and well-organised process of myogenesis. Although there is a good understanding about the role of the Muscle Regulatory Factors during myogenesis, little is known about the potential interplay of other metabolic proteins. The aim of this study was to determine the endogenous mRNA expression profile for a novel group of genes, recently associated with β2-adrenergic agonist (BA) induced muscle hypertrophy in pigs [1], during myogenic differentiation in C2C12 cells and their response to dibutyryl cyclic-AMP (dbcAMP). These genes included mitochondrial phosphoenolpyruvate carboxykinase (PCK2/PEPCK-M), genes involved in serine biosynthesis (Phosphoglycerate dehydrogenase, PHGDH; Phosphoserine aminotransferase-1, PSAT1; Phosphoserine phosphatase, PSPH) and those involved in an integrated stress response (Asparagine synthetase, ASNS; Sestrin-2, SESN2; and Activating transcription factor-5, ATF5). A coordinated peak in endogenous PCK2, PHGDH, PSAT1, PSPH, ASNS, ATF5 and SESN2 mRNA expression was observed at day 2 of differentiation (P < 0.001) in C2C12 cells, which coincided with the peak in myogenin mRNA. Myotube hypertrophy was induced with dbcAMP (1 mM) treatment from day 0, thereby mimicking the in vivo BA response. Although dbcAMP treatment from day 0 induced larger myotubes and increased both myosin heavy chain-IIB (MyHC-IIB) and pyruvate carboxylase (PC) mRNA, the expression of PCK2, PHGDH, PSAT1 and ASNS mRNA were all unaffected. Treatment with dbcAMP from day 4 increased MyHC-IIB mRNA, however this was less dramatic compared to the response observed following treatment from day 0, but there was no effect on PC mRNA. There was also no effect of dbcAMP treatment from day 4 on PCK2, PHGDH, PSAT1 and ASNS mRNA. To conclude, the coordinated day 2 peak in endogenous expression of PCK2, PHGDH, PSAT1, PSPH, ASNS, ATF5 and SESN2 mRNA may relate to a shift in biosynthetic demand required to initiate myogenic differentiation. However, dbcAMP had no effect on the expression of these genes in vitro suggesting that the effects observed in BA-treated pigs might be via other signalling pathways from the activation of the β2-adrenergic receptor, but independent of cAMP, or that there are species differences in the response.
Collapse
Affiliation(s)
- Madelaine C. Brearley
- School of Biosciences, University of Nottingham, Sutton Bonington Campus, Loughborough, Leicestershire, LE12 5RD, UK
| | - Congcong Li
- Key Laboratory of Agricultural Animal Genetics, Breeding, and Reproduction of the Ministry of Education, Huazhong Agricultural University, Wuhan, Hubei, China
| | - Zoe C.T.R. Daniel
- School of Biosciences, University of Nottingham, Sutton Bonington Campus, Loughborough, Leicestershire, LE12 5RD, UK
| | - Paul T. Loughna
- School of Veterinary Medicine & Science, University of Nottingham, Sutton Bonington Campus, Loughborough, Leicestershire, LE12 5RD, UK
| | - Tim Parr
- School of Biosciences, University of Nottingham, Sutton Bonington Campus, Loughborough, Leicestershire, LE12 5RD, UK
| | - John M. Brameld
- School of Biosciences, University of Nottingham, Sutton Bonington Campus, Loughborough, Leicestershire, LE12 5RD, UK
| |
Collapse
|
24
|
Ruparelia AA, Ratnayake D, Currie PD. Stem cells in skeletal muscle growth and regeneration in amniotes and teleosts: Emerging themes. WILEY INTERDISCIPLINARY REVIEWS-DEVELOPMENTAL BIOLOGY 2019; 9:e365. [PMID: 31743958 DOI: 10.1002/wdev.365] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Revised: 09/22/2019] [Accepted: 10/03/2019] [Indexed: 12/19/2022]
Abstract
Skeletal muscle is a contractile, postmitotic tissue that retains the capacity to grow and regenerate throughout life in amniotes and teleost. Both muscle growth and regeneration are regulated by obligate tissue resident muscle stem cells. Given that considerable knowledge exists on the myogenic process, recent studies have focused on examining the molecular markers of muscle stem cells, and on the intrinsic and extrinsic signals regulating their function. From this, two themes emerge: firstly, muscle stem cells display remarkable heterogeneity not only with regards to their gene expression profile, but also with respect to their behavior and function; and secondly, the stem cell niche is a critical regulator of muscle stem cell function during growth and regeneration. Here, we will address the current understanding of these emerging themes with emphasis on the distinct processes used by amniotes and teleost, and discuss the challenges and opportunities in the muscle growth and regeneration fields. This article is characterized under: Adult Stem Cells, Tissue Renewal, and Regeneration > Tissue Stem Cells and Niches Early Embryonic Development > Development to the Basic Body Plan Vertebrate Organogenesis > Musculoskeletal and Vascular.
Collapse
Affiliation(s)
- Avnika A Ruparelia
- Australian Regenerative Medicine Institute, Monash University, Melbourne, Victoria, Australia.,EMBL Australia, Monash University, Melbourne, Victoria, Australia
| | - Dhanushika Ratnayake
- Australian Regenerative Medicine Institute, Monash University, Melbourne, Victoria, Australia.,EMBL Australia, Monash University, Melbourne, Victoria, Australia
| | - Peter D Currie
- Australian Regenerative Medicine Institute, Monash University, Melbourne, Victoria, Australia.,EMBL Australia, Monash University, Melbourne, Victoria, Australia
| |
Collapse
|
25
|
Functions and Regulatory Mechanisms of lncRNAs in Skeletal Myogenesis, Muscle Disease and Meat Production. Cells 2019; 8:cells8091107. [PMID: 31546877 PMCID: PMC6769631 DOI: 10.3390/cells8091107] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Revised: 09/04/2019] [Accepted: 09/17/2019] [Indexed: 12/20/2022] Open
Abstract
Myogenesis is a complex biological process, and understanding the regulatory network of skeletal myogenesis will contribute to the treatment of human muscle related diseases and improvement of agricultural animal meat production. Long noncoding RNAs (lncRNAs) serve as regulators in gene expression networks, and participate in various biological processes. Recent studies have identified functional lncRNAs involved in skeletal muscle development and disease. These lncRNAs regulate the proliferation, differentiation, and fusion of myoblasts through multiple mechanisms, such as chromatin modification, transcription regulation, and microRNA sponge activity. In this review, we presented the latest advances regarding the functions and regulatory activities of lncRNAs involved in muscle development, muscle disease, and meat production. Moreover, challenges and future perspectives related to the identification of functional lncRNAs were also discussed.
Collapse
|
26
|
Blaauw B. Platelet-Derived Growth Factor signaling and the role of cellular crosstalk in functional muscle growth. FEBS Lett 2019; 591:690-692. [PMID: 28297119 DOI: 10.1002/1873-3468.12602] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Affiliation(s)
- Bert Blaauw
- Department of Biomedical Sciences, University of Padova, Italy.,Venetian Institute of Molecular Medicine, Padova, Italy
| |
Collapse
|
27
|
Choi YH, Kim SH, Kim IG, Lee JH, Kwon SK. Injectable basic fibroblast growth factor-loaded alginate/hyaluronic acid hydrogel for rejuvenation of geriatric larynx. Acta Biomater 2019; 89:104-114. [PMID: 30849562 DOI: 10.1016/j.actbio.2019.03.005] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Revised: 02/28/2019] [Accepted: 03/04/2019] [Indexed: 01/01/2023]
Abstract
Increase in the geriatric population has led to an increase in the number of elderly patients with laryngeal atrophy and dysfunction. Symptoms of voice change, dysphagia, and aspiration pneumonia negatively influence patient's health status, quality of life, and life span. Injection laryngoplasty used to treat laryngeal dysfunctions does not recover intrinsic functions of the larynx. Thus, we fabricated an injectable basic fibroblast growth factor (bFGF)-loaded alginate (ALG)/hyaluronic acid (HA) hydrogel for inducing rejuvenation of geriatric laryngeal muscles. Optimal in situ-forming bFGF-loaded ALG/HA hydrogel for injection laryngoplasty was prepared and the release profile of bFGF was analyzed. For in vivo analysis, the bFGF-loaded ALG/HA hydrogel was injected into the laryngeal muscles of 18-month-old Sprague-Dawley rats. The rejuvenation efficacy of bFGF-loaded ALG/HA hydrogel in geriatric laryngeal muscle tissues 4- and 12-weeks post-injection was evaluated by quantitative polymerase chain reaction (qPCR), histology, immune-fluorescence staining and functionality analysis. The bFGF-loaded ALG/HA hydrogel induced an increase in the expression of myogenic regulatory factor-related genes, hypertrophy of muscle fiber, proliferation of muscle satellite cells, and angiogenesis and decreased interstitial fibrosis. Administration of the bFGF-loaded ALG/HA hydrogel caused successful glottal gap closure. Thus, the bFGF-loaded ALG/HA hydrogel could be a promising candidate for laryngoplasty aimed at rejuvenating geriatric larynx. STATEMENT OF SIGNIFICANCE: In this manuscript, optimal in situ-forming bFGF-loaded ALG/HA hydrogel for injection laryngoplasty was prepared and the release profile of bFGF was analyzed. Herein, we introduced the materials and methods of injection laryngoplasty for geriatric rat experiment. In addition, we studied effects of bFGF-loaded ALG/HA hydrogel on the therapeutic rejuvenation of geriatric rat larynx. The bFGF-loaded ALG/HA hydrogel induced an increase in the expression of myogenic regulatory factor-related genes, hypertrophy of muscle fiber, proliferation of muscle satellite cells, and angiogenesis and decreased interstitial fibrosis. Furthermore, our functional analysis through the high-speed camera setup demonstrated that the administration of the bFGF-loaded ALG/HA hydrogel induced successful glottal gap closure. Thus, the bFGF-loaded ALG/HA hydrogel could be a promising candidate for injection laryngoplasty with therapeutic effects.
Collapse
Affiliation(s)
- Young Hwan Choi
- Department of Otorhinolaryngology-Head and Neck, Seoul National University Hospital, Seoul 03080, Republic of Korea; School of Chemical and Biological Engineering, Seoul National University, Seoul 08826, Republic of Korea
| | - Sae Hyun Kim
- Department of Advanced Materials and Chemical Engineering, Hannam University, Daejeon 34054, Republic of Korea
| | - In Gul Kim
- Department of Otorhinolaryngology-Head and Neck, Seoul National University Hospital, Seoul 03080, Republic of Korea
| | - Jin Ho Lee
- Department of Advanced Materials and Chemical Engineering, Hannam University, Daejeon 34054, Republic of Korea.
| | - Seong Keun Kwon
- Department of Otorhinolaryngology-Head and Neck, Seoul National University Hospital, Seoul 03080, Republic of Korea; Department of Otorhinolaryngology-Head and Neck Surgery, Seoul National University College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
28
|
Forcina L, Miano C, Pelosi L, Musarò A. An Overview about the Biology of Skeletal Muscle Satellite Cells. Curr Genomics 2019; 20:24-37. [PMID: 31015789 PMCID: PMC6446479 DOI: 10.2174/1389202920666190116094736] [Citation(s) in RCA: 88] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Revised: 12/19/2018] [Accepted: 01/08/2019] [Indexed: 12/14/2022] Open
Abstract
The peculiar ability of skeletal muscle tissue to operate adaptive changes during post-natal de-velopment and adulthood has been associated with the existence of adult somatic stem cells. Satellite cells, occupying an exclusive niche within the adult muscle tissue, are considered bona fide stem cells with both stem-like properties and myogenic activities. Indeed, satellite cells retain the capability to both maintain the quiescence in uninjured muscles and to be promptly activated in response to growth or re-generative signals, re-engaging the cell cycle. Activated cells can undergo myogenic differentiation or self-renewal moving back to the quiescent state. Satellite cells behavior and their fate decision are finely controlled by mechanisms involving both cell-autonomous and external stimuli. Alterations in these regu-latory networks profoundly affect muscle homeostasis and the dynamic response to tissue damage, con-tributing to the decline of skeletal muscle that occurs under physio-pathologic conditions. Although the clear myogenic activity of satellite cells has been described and their pivotal role in muscle growth and regeneration has been reported, a comprehensive picture of inter-related mechanisms guiding muscle stem cell activity has still to be defined. Here, we reviewed the main regulatory networks determining satellite cell behavior. In particular, we focused on genetic and epigenetic mechanisms underlining satel-lite cell maintenance and commitment. Besides intrinsic regulations, we reported current evidences about the influence of environmental stimuli, derived from other cell populations within muscle tissue, on satel-lite cell biology.
Collapse
Affiliation(s)
- Laura Forcina
- DAHFMO-Unit of Histology and Medical Embryology, Sapienza University of Rome, Laboratory Affiliated to Istituto Pasteur Italia - Fondazione Cenci Bolognetti, Via A. Scarpa, 14 Rome 00161, Italy
| | - Carmen Miano
- DAHFMO-Unit of Histology and Medical Embryology, Sapienza University of Rome, Laboratory Affiliated to Istituto Pasteur Italia - Fondazione Cenci Bolognetti, Via A. Scarpa, 14 Rome 00161, Italy
| | - Laura Pelosi
- DAHFMO-Unit of Histology and Medical Embryology, Sapienza University of Rome, Laboratory Affiliated to Istituto Pasteur Italia - Fondazione Cenci Bolognetti, Via A. Scarpa, 14 Rome 00161, Italy
| | - Antonio Musarò
- DAHFMO-Unit of Histology and Medical Embryology, Sapienza University of Rome, Laboratory Affiliated to Istituto Pasteur Italia - Fondazione Cenci Bolognetti, Via A. Scarpa, 14 Rome 00161, Italy
| |
Collapse
|
29
|
Change of Direction Speed: Toward a Strength Training Approach with Accentuated Eccentric Muscle Actions. Sports Med 2018; 48:1773-1779. [PMID: 29594958 DOI: 10.1007/s40279-018-0907-3] [Citation(s) in RCA: 84] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
There is growing evidence that eccentric strength training appears to have benefits over traditional strength training (i.e., strength training with combined concentric and eccentric muscle actions) from muscular, neuromuscular, tendinous, and metabolic perspectives. Eccentric muscle strength is particularly needed to decelerate and stabilize the body during the braking phase of a jump exercise or during rapid changes of direction (CoD) tasks. However, surprisingly little research has been conducted to elucidate the effects of eccentric strength training or strength training with accentuated eccentric muscle actions on CoD speed performance. In this current opinion article, we present findings from cross-sectional studies on the relationship between measures of eccentric muscle strength and CoD speed performance. In addition, we summarize the few available studies on the effects of strength training with accentuated eccentric muscle actions on CoD speed performance in athletic populations. Finally, we propose strength training with accentuated eccentric muscle actions as a promising element in strength and conditioning programs of sports with high CoD speed demands. Our findings from five cross-sectional studies revealed statistically significant moderate- to large-sized correlations (r = 0.45-0.89) between measures of eccentric muscle strength and CoD speed performance in athletic populations. The identified three intervention studies were of limited methodological quality and reported small- to large-sized effects (d = 0.46-1.31) of strength training with accentuated eccentric muscle actions on CoD speed performance in athletes. With reference to the available but preliminary literature and from a performance-related point of view, we recommend strength and conditioning coaches to include strength training with accentuated eccentric muscle actions in training routines of sports with high CoD speed demands (e.g., soccer, handball, basketball, hockey) to enhance sport-specific performance. Future comparative studies are needed to deepen our knowledge of the effects of strength training with accentuated eccentric muscle actions on CoD speed performance in athletes.
Collapse
|
30
|
Wilkinson D, Piasecki M, Atherton P. The age-related loss of skeletal muscle mass and function: Measurement and physiology of muscle fibre atrophy and muscle fibre loss in humans. Ageing Res Rev 2018; 47:123-132. [PMID: 30048806 PMCID: PMC6202460 DOI: 10.1016/j.arr.2018.07.005] [Citation(s) in RCA: 476] [Impact Index Per Article: 68.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2018] [Revised: 06/20/2018] [Accepted: 07/18/2018] [Indexed: 12/11/2022]
Abstract
Loss of muscle mass with age is due to atrophy and loss of individual muscle fibres. Anabolic resistance is fundamental in age-related fibre atrophy. Fibre loss is associated with denervation and remodelling of motor units. The plasticity of both factors should be considered in future research.
Age-related loss of skeletal muscle mass and function, sarcopenia, is associated with physical frailty and increased risk of morbidity (chronic diseases), in addition to all-cause mortality. The loss of muscle mass occurs incipiently from middle-age (∼1%/year), and in severe instances can lead to a loss of ∼50% by the 8–9th decade of life. This review will focus on muscle deterioration with ageing and highlight the two underpinning mechanisms regulating declines in muscle mass and function: muscle fibre atrophy and muscle fibre loss (hypoplasia) – and their measurement. The mechanisms of muscle fibre atrophy in humans relate to imbalances in muscle protein synthesis (MPS) and breakdown (MPB); however, since there is limited evidence for basal alterations in muscle protein turnover, it would appear that “anabolic resistance” to fundamental environmental cues regulating diurnal muscle homeostasis (namely physical activity and nutrition), underlie age-related catabolic perturbations in muscle proteostasis. While the ‘upstream’ drivers of the desensitization of aged muscle to anabolic stimuli are poorly defined, they most likely relate to impaired efficiency of the conversion of nutritional/exercise stimuli into signalling impacting mRNA translation and proteolysis. Additionally, loss of muscle fibres has been shown in cadaveric studies using anatomical fibre counts, and from iEMG studies demonstrating motor unit loss, albeit with few molecular investigations of this in humans. We suggest that defining countermeasures against sarcopenia requires improved understandings of the co-ordinated regulation of muscle fibre atrophy and fibre loss, which are likely to be inextricably linked.
Collapse
|
31
|
Lim CH, Gil JH, Quan H, Viet DH, Kim CK. Effect of 8-week leucine supplementation and resistance exercise training on muscle hypertrophy and satellite cell activation in rats. Physiol Rep 2018; 6:e13725. [PMID: 29952091 PMCID: PMC6021278 DOI: 10.14814/phy2.13725] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Revised: 05/04/2018] [Accepted: 05/09/2018] [Indexed: 12/30/2022] Open
Abstract
We investigated the effects of regular leucine intake and/or resistance exercise training on skeletal muscle hypertrophy and satellite cell activity after the administration of different doses of leucine. Ten-week-old Sprague-Dawley rats were assigned to six groups (n = 7 per group): a control group (Con), two groups receiving either 10% (0.135 g/kg.wt) (Leu10) or 50% (0.675 g/kg.wt) (Leu50) leucine supplementation, and three exercise groups receiving 0% (Ex), 10% (Leu10Ex), and 50% (Leu50Ex) leucine supplementation. The rats performed ladder climbing exercises thrice per week for 8 weeks, and received leucine supplements at the same time daily. Muscle phenotypes were assessed by immunohistochemistry. MyoD, myogenin, and IGF1 protein levels were determined by western blot. The Leu50Ex group displayed significantly higher numbers of positive embryonic myosin fibers (0.35 ± 0.08, 250%) and myonuclei (3.29 ± 0.3, 118.7%) than all other groups. And exercise training groups increased the cross-sectional area, the number of satellite cells and protein expression of MyoD, myogenin, and IGF1alpha relative to the Control group (P < 0.05). However, Only leucine supplementation group did not increase skeletal muscle hypertrophy and satellite cell activity, regardless of the dose (P > 0.05). Leucine intake accompanied by regular exercise training may increase satellite cell activation in skeletal muscles, and improve muscle quality more effectively than continuous leucine ingestion alone.
Collapse
MESH Headings
- Animals
- Cell Differentiation/drug effects
- Combined Modality Therapy
- Dietary Supplements
- Dose-Response Relationship, Drug
- Hypertrophy/metabolism
- Hypertrophy/pathology
- Hypertrophy/prevention & control
- Leucine/administration & dosage
- Leucine/pharmacology
- Leucine/therapeutic use
- Male
- Muscle Fibers, Skeletal/drug effects
- Muscle Fibers, Skeletal/pathology
- Muscle Proteins/metabolism
- Muscle Strength/physiology
- Muscle, Skeletal/drug effects
- Muscle, Skeletal/metabolism
- Muscle, Skeletal/pathology
- Muscle, Skeletal/physiopathology
- Physical Conditioning, Animal/physiology
- Rats, Sprague-Dawley
- Resistance Training/methods
- Satellite Cells, Skeletal Muscle/drug effects
- Satellite Cells, Skeletal Muscle/pathology
- Weight Gain/drug effects
- Weight Gain/physiology
- Weight-Bearing/physiology
Collapse
Affiliation(s)
- Chang Hyun Lim
- Human physiology, KoreaNational Sport UniversitySeoulKorea
- Exercise and Metabolism Research CenterZhejiang Normal UniversityJinhuaChina
| | - Ju Hyun Gil
- Human physiology, KoreaNational Sport UniversitySeoulKorea
| | - Helong Quan
- Exercise and Metabolism Research CenterZhejiang Normal UniversityJinhuaChina
- College of Physical Education and Health ScienceZhejiang Normal UniversityJinhuaChina
| | - Dang Ha Viet
- Sport Science and Technology InstituteHochiminh City University of SportHochiminh CityVietnam
| | - Chang Keun Kim
- Human physiology, KoreaNational Sport UniversitySeoulKorea
- Exercise and Metabolism Research CenterZhejiang Normal UniversityJinhuaChina
| |
Collapse
|
32
|
Reidy PT, Fry CS, Dickinson JM, Drummond MJ, Rasmussen BB. Postexercise essential amino acid supplementation amplifies skeletal muscle satellite cell proliferation in older men 24 hours postexercise. Physiol Rep 2018; 5:5/11/e13269. [PMID: 28596299 PMCID: PMC5471431 DOI: 10.14814/phy2.13269] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Revised: 03/31/2017] [Accepted: 04/06/2017] [Indexed: 12/29/2022] Open
Abstract
Aged skeletal muscle has an attenuated and delayed ability to proliferate satellite cells in response to resistance exercise. The mechanistic target of rapamycin complex 1 (mTORC1) signaling pathway is a focal point for cell growth, however, the effect of postexercise mTORC1 activation on human skeletal muscle satellite cell (SC) proliferation is unknown. To test the proliferative capacity of skeletal muscle SC in aging muscle to a potent mTORC1 activator (i.e., EAA; essential amino acids) we recruited older (~72y) men to conduct leg resistance exercise (8setsx10reps) without (−EAA; n = 8) and with (+EAA: n = 11) ingestion of 10 g of EAA 1 h postexercise. Muscle biopsies were taken before exercise (Pre) and 24 h postexercise (Post) for assessment of expression and fiber type‐specific Pax7+SC, Ki67+Pax7+SC and MyoD+SC. −EAA did not show an increase in Pax7+ satellite cells at Post(P > 0.82). Although statistical significance for an increase in Pax7 + SC at 24 h post‐RE was not observed in +EAA versus −EAA, we observed trends for a treatment difference (P < 0.1). When examining the change from Pre to Post trends were demonstrated (#/myofiber: P = 0.076; and %/myonuclei: P = 0.065) for a greater increase in +EAA versus −EAA. Notably, we found an increase SC proliferation in +EAA, but not −EAA with increase in Ki67+SC and MyoD+ cells (P < 0.05). Ki67+SC also exhibited a significant group difference Post (P < 0.010). Pax7+SC in fast twitch myofibers did not change and were not different between groups (P > 0.10). CDK2, MEF2C, RB1 mRNA only increased in +EAA (P < 0.05). Acute muscle satellite cell proliferative capacity may be partially rescued with postexercise EAA ingestion in older men.
Collapse
Affiliation(s)
- Paul T Reidy
- Center for Recovery, Physical Activity and Nutrition, University of Texas Medical Branch, Galveston, Texas.,Department of Nutrition and Metabolism, University of Texas Medical Branch, Galveston, Texas
| | - Christopher S Fry
- Department of Nutrition and Metabolism, University of Texas Medical Branch, Galveston, Texas
| | - Jared M Dickinson
- Center for Recovery, Physical Activity and Nutrition, University of Texas Medical Branch, Galveston, Texas.,Department of Nutrition and Metabolism, University of Texas Medical Branch, Galveston, Texas
| | - Micah J Drummond
- Department of Nutrition and Metabolism, University of Texas Medical Branch, Galveston, Texas
| | - Blake B Rasmussen
- Center for Recovery, Physical Activity and Nutrition, University of Texas Medical Branch, Galveston, Texas .,Department of Nutrition and Metabolism, University of Texas Medical Branch, Galveston, Texas
| |
Collapse
|
33
|
Shamim B, Hawley JA, Camera DM. Protein Availability and Satellite Cell Dynamics in Skeletal Muscle. Sports Med 2018; 48:1329-1343. [DOI: 10.1007/s40279-018-0883-7] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
34
|
Piórkowska K, Żukowski K, Ropka-Molik K, Tyra M, Gurgul A. A comprehensive transcriptome analysis of skeletal muscles in two Polish pig breeds differing in fat and meat quality traits. Genet Mol Biol 2018; 41:125-136. [PMID: 29658965 PMCID: PMC5901489 DOI: 10.1590/1678-4685-gmb-2016-0101] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Accepted: 09/11/2017] [Indexed: 12/31/2022] Open
Abstract
Pork is the most popular meat in the world. Unfortunately, the selection pressure
focused on high meat content led to a reduction in pork quality. The present
study used RNA-seq technology to identify metabolic process genes related to
pork quality traits and fat deposition. Differentially expressed genes (DEGs)
were identified between pigs of Pulawska and Polish Landrace breeds for two the
most important muscles (semimembranosus and longissimus
dorsi). A total of 71 significant DEGs were reported: 15 for
longissimus dorsi and 56 for
semimembranosus muscles. The genes overexpressed in
Pulawska pigs were involved in lipid metabolism (APOD,
LXRA, LIPE, AP2B1, ENSSSCG00000028753 and
OAS2) and proteolysis (CST6, CTSD, ISG15
and UCHL1). In Polish Landrace pigs, genes playing a role in
biological adhesion (KIT, VCAN, HES1, SFRP2, CDH11, SSX2IP and
PCDH17), actin cytoskeletal organisation (FRMD6,
LIMK1, KIF23 and CNN1) and calcium ion binding
(PVALB, CIB2, PCDH17, VCAN and CDH11) were
transcriptionally more active. The present study allows for better understanding
of the physiological processes associated with lipid metabolism and muscle fiber
organization. This information could be helpful in further research aiming to
estimate the genetic markers.
Collapse
Affiliation(s)
- Katarzyna Piórkowska
- Department of Animal Molecular Biology, National Research Institute of Animal Production, Balice, Poland
| | - Kacper Żukowski
- Department of Cattle Breeding, National Research Institute of Animal Production, Balice, Poland
| | - Katarzyna Ropka-Molik
- Department of Animal Molecular Biology, National Research Institute of Animal Production, Balice, Poland
| | - Mirosław Tyra
- Department of Pig Breeding, National Research Institute of Animal Production, Balice, Poland
| | - Artur Gurgul
- Department of Animal Molecular Biology, National Research Institute of Animal Production, Balice, Poland
| |
Collapse
|
35
|
Dennis RA, Garner KK, Kortebein PM, Parkes CM, Bopp MM, Li S, Padala KP, Padala PR, Sullivan DH. Single-Arm Resistance Training Study to Determine the Relationship between Training Outcomes and Muscle Growth Factor mRNAs in Older Adults Consuming Numerous Medications and Supplements. J Nutr Health Aging 2018; 22:269-275. [PMID: 29380855 DOI: 10.1007/s12603-017-0913-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
OBJECTIVES Determine if the muscle mRNA levels of three growth factors (insulin-like growth factor-1 [IGF1], ciliary neurotropic factor [CNTF], and vascular endothelial growth factor-D [VEGFD]) are correlated with muscle size and strength gains from resistance exercise while piloting a training program in older adults taking medications and supplements for age-associated problems. DESIGN Single-arm prospective study. SETTING US Veterans Affairs hospital. PARTICIPANTS Older (70±6 yrs) male Veterans (N=14) of US military service. INTERVENTION Thirty-five sessions of high-intensity (80% one-rep max) resistance training including leg press, knee curl, and knee extension to target the thigh muscles. MEASUREMENTS Vastus lateralis biopsies were collected and body composition (DEXA) was determined pre- and post-training. Simple Pearson correlations were used to compare training outcomes to growth factor mRNA levels and other independent variables such as medication and supplement use. RESULTS Average strength increase for the group was ≥ 25% for each exercise. Subjects averaged taking numerous medications (N=5±3) and supplements (N=2±2). Of the growth factors, a significant correlation (R>0.7, P≤0.003) was only found between pre-training VEGFD and gains in lean thigh mass and extension strength. Mass and strength gains were also correlated with use of α-1 antagonists (R=0.55, P=0.04) and pre-training lean mass (R=0.56, P=0.04), respectively. CONCLUSIONS Muscle VEGFD, muscle mass, and use of α-1 antagonists may be predisposing factors that influence the response to training in this population of older adults but additional investigation is required to determine if these relationships are due to muscle angiogenesis and blood supply.
Collapse
Affiliation(s)
- R A Dennis
- Richard A Dennis, Geriatric Research Education and Clinical Center, Central Arkansas Veterans Healthcare System, 2200 Fort Roots Drive, 170/3J, North Little Rock, AR 72114, USA, or 501-257-3503
| | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Randrianarison-Huetz V, Papaefthymiou A, Herledan G, Noviello C, Faradova U, Collard L, Pincini A, Schol E, Decaux JF, Maire P, Vassilopoulos S, Sotiropoulos A. Srf controls satellite cell fusion through the maintenance of actin architecture. J Cell Biol 2017; 217:685-700. [PMID: 29269426 PMCID: PMC5800804 DOI: 10.1083/jcb.201705130] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2017] [Revised: 10/20/2017] [Accepted: 11/21/2017] [Indexed: 01/17/2023] Open
Abstract
This work describes a crucial role for the transcription factor Srf and F-actin scaffold to drive muscle stem cell fusion in vitro and in vivo and provides evidence of how actin cytoskeleton architecture affects myoblast fusion in vertebrates. Satellite cells (SCs) are adult muscle stem cells that are mobilized when muscle homeostasis is perturbed. Here, we show that serum response factor (Srf) is needed for optimal SC-mediated hypertrophic growth. We identified Srf as a master regulator of SC fusion required in both fusion partners, whereas it was dispensable for SC proliferation and differentiation. We show that SC-specific Srf deletion leads to impaired actin cytoskeleton and report the existence of finger-like actin–based protrusions at fusion sites in vertebrates that were notoriously absent in fusion-defective myoblasts lacking Srf. Restoration of a polymerized actin network by overexpression of an α-actin isoform in Srf mutant SCs rescued their fusion with a control cell in vitro and in vivo and reestablished overload-induced muscle growth. These findings demonstrate the importance of Srf in controlling the organization of actin cytoskeleton and actin-based protrusions for myoblast fusion in mammals and its requirement to achieve efficient hypertrophic myofiber growth.
Collapse
Affiliation(s)
- Voahangy Randrianarison-Huetz
- Institut National de la Santé et de la Recherche Médicale U1016, Institut Cochin, Paris, France.,Centre National de la Recherche Scientifique UMR8104, Paris, France.,Université Paris Descartes, Paris, France
| | - Aikaterini Papaefthymiou
- Institut National de la Santé et de la Recherche Médicale U1016, Institut Cochin, Paris, France.,Centre National de la Recherche Scientifique UMR8104, Paris, France.,Université Paris Descartes, Paris, France
| | - Gaëlle Herledan
- Institut National de la Santé et de la Recherche Médicale U1016, Institut Cochin, Paris, France.,Centre National de la Recherche Scientifique UMR8104, Paris, France.,Université Paris Descartes, Paris, France
| | - Chiara Noviello
- Institut National de la Santé et de la Recherche Médicale U1016, Institut Cochin, Paris, France.,Centre National de la Recherche Scientifique UMR8104, Paris, France.,Université Paris Descartes, Paris, France
| | - Ulduz Faradova
- Institut National de la Santé et de la Recherche Médicale U1016, Institut Cochin, Paris, France.,Centre National de la Recherche Scientifique UMR8104, Paris, France.,Université Paris Descartes, Paris, France
| | | | - Alessandra Pincini
- Institut National de la Santé et de la Recherche Médicale U1016, Institut Cochin, Paris, France.,Centre National de la Recherche Scientifique UMR8104, Paris, France.,Université Paris Descartes, Paris, France
| | - Emilie Schol
- Institut National de la Santé et de la Recherche Médicale U1016, Institut Cochin, Paris, France.,Centre National de la Recherche Scientifique UMR8104, Paris, France.,Université Paris Descartes, Paris, France
| | - Jean François Decaux
- Université Pierre et Marie Curie Paris 6, Centre National de la Recherche Scientifique UMR8256, Institut National de la Santé et de la Recherche Médicale U1164, Institute of Biology Paris-Seine, Paris, France
| | - Pascal Maire
- Institut National de la Santé et de la Recherche Médicale U1016, Institut Cochin, Paris, France.,Centre National de la Recherche Scientifique UMR8104, Paris, France.,Université Paris Descartes, Paris, France
| | - Stéphane Vassilopoulos
- Institut National de la Santé et de la Recherche Médicale/University Pierre and Marie Curie UMR-S974, Institut de Myologie, Paris, France
| | - Athanassia Sotiropoulos
- Institut National de la Santé et de la Recherche Médicale U1016, Institut Cochin, Paris, France .,Centre National de la Recherche Scientifique UMR8104, Paris, France.,Université Paris Descartes, Paris, France
| |
Collapse
|
37
|
Egner IM, Bruusgaard JC, Gundersen K. Satellite cell depletion prevents fiber hypertrophy in skeletal muscle. Development 2017; 143:2898-906. [PMID: 27531949 DOI: 10.1242/dev.134411] [Citation(s) in RCA: 154] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2015] [Accepted: 06/28/2016] [Indexed: 12/18/2022]
Abstract
The largest mammalian cells are the muscle fibers, and they have multiple nuclei to support their large cytoplasmic volumes. During hypertrophic growth, new myonuclei are recruited from satellite stem cells into the fiber syncytia, but it was recently suggested that such recruitment is not obligatory: overload hypertrophy after synergist ablation of the plantaris muscle appeared normal in transgenic mice in which most of the satellite cells were abolished. When we essentially repeated these experiments analyzing the muscles by immunohistochemistry and in vivo and ex vivo imaging, we found that overload hypertrophy was prevented in the satellite cell-deficient mice, in both the plantaris and the extensor digitorum longus muscles. We attribute the previous findings to a reliance on muscle mass as a proxy for fiber hypertrophy, and to the inclusion of a significant number of regenerating fibers in the analysis. We discuss that there is currently no model in which functional, sustainable hypertrophy has been unequivocally demonstrated in the absence of satellite cells; an exception is re-growth, which can occur using previously recruited myonuclei without addition of new myonuclei.
Collapse
Affiliation(s)
- Ingrid M Egner
- Department of Biosciences, University of Oslo, Blindern, Oslo N-0316, Norway
| | - Jo C Bruusgaard
- Department of Biosciences, University of Oslo, Blindern, Oslo N-0316, Norway Department of Health Sciences, Kristiania University College, P.O. Box 1190, Sentrum, Oslo N-0107, Norway
| | - Kristian Gundersen
- Department of Biosciences, University of Oslo, Blindern, Oslo N-0316, Norway
| |
Collapse
|
38
|
Nielsen JL, Aagaard P, Prokhorova TA, Nygaard T, Bech RD, Suetta C, Frandsen U. Blood flow restricted training leads to myocellular macrophage infiltration and upregulation of heat shock proteins, but no apparent muscle damage. J Physiol 2017; 595:4857-4873. [PMID: 28481416 DOI: 10.1113/jp273907] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2016] [Accepted: 05/02/2017] [Indexed: 12/27/2022] Open
Abstract
KEY POINTS Muscular contractions performed using a combination of low external loads and partial restriction of limb blood flow appear to induce substantial gains in muscle strength and muscle mass. This exercise regime may initially induce muscular stress and damage; however, the effects of a period of blood flow restricted training on these parameters remain largely unknown. The present study shows that short-term, high-frequency, low-load muscle training performed with partial blood flow restriction does not induce significant muscular damage. However, signs of myocellular stress and inflammation that were observed in the early phase of training and after the training intervention, respectively, may be facilitating the previously reported gains in myogenic satellite cell content and muscle hypertrophy. The present results improve our current knowledge about the physiological effects of low-load muscular contractions performed under blood flow restriction and may provide important information of relevance for future therapeutic treatment of muscular atrophy. ABSTRACT Previous studies indicate that low-load muscle contractions performed under local blood flow restriction (BFR) may initially induce muscle damage and stress. However, whether these factors are evoked with longitudinal BFR training remains unexplored at the myocellular level. Two distinct study protocols were conducted, covering 3 weeks (3 wk) or one week (1 wk). Subjects performed BFR exercise (100 mmHg, 20% 1RM) to concentric failure (BFRE) (3 wk/1 wk), while controls performed work-matched (LLE) (3 wk) or high-load (HLE; 70% 1RM) (1 wk) free-flow exercise. Muscle biopsies (3 wk) were obtained at baseline (Pre), 8 days into the intervention (Mid8), and 3 and 10 days after training cessation (Post3, Post10) to examine macrophage (M1/M2) content as well as heat shock protein (HSP27/70) and tenascin-C expression. Blood samples (1 wk) were collected before and after (0.1-24 h) the first and last training session to examine markers of muscle damage (creatine kinase), oxidative stress (total antibody capacity, glutathione) and inflammation (monocyte chemotactic protein-1, interleukin-6, tumour necrosis factor α). M1-macrophage content increased 108-165% with BFRE and LLE at Post3 (P < 0.05), while M2-macrophages increased (163%) with BFRE only (P < 0.01). Membrane and intracellular HSP27 expression increased 60-132% at Mid8 with BFRE (P < 0.05-0.01). No or only minor changes were observed in circulating markers of muscle damage, oxidative stress and inflammation. The amplitude, timing and localization of the above changes indicate that only limited muscle damage was evoked with BFRE. This study is the first to show that a period of high-frequency, low-load BFR training does not appear to induce general myocellular damage. However, signs of tissue inflammation and focal myocellular membrane stress and/or reorganization were observed that may be involved in the adaptation processes evoked by BFR muscle exercise.
Collapse
Affiliation(s)
- Jakob L Nielsen
- Department of Sports Science and Clinical Biomechanics, and SDU Muscle Research Cluster (SMRC), University of Southern Denmark, Odense, Denmark
| | - Per Aagaard
- Department of Sports Science and Clinical Biomechanics, and SDU Muscle Research Cluster (SMRC), University of Southern Denmark, Odense, Denmark
| | - Tatyana A Prokhorova
- Department of Sports Science and Clinical Biomechanics, and SDU Muscle Research Cluster (SMRC), University of Southern Denmark, Odense, Denmark
| | - Tobias Nygaard
- Department of Orthopaedic Surgery, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Rune D Bech
- Department of Orthopaedic Surgery, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Charlotte Suetta
- Department of Clinical Physiology, Nuclear Medicine and PET, Rigshospitalet Glostrup, University of Copenhagen, Copenhagen, Denmark
| | - Ulrik Frandsen
- Department of Sports Science and Clinical Biomechanics, and SDU Muscle Research Cluster (SMRC), University of Southern Denmark, Odense, Denmark
| |
Collapse
|
39
|
Gundersen K. Muscle memory and a new cellular model for muscle atrophy and hypertrophy. ACTA ACUST UNITED AC 2016; 219:235-42. [PMID: 26792335 DOI: 10.1242/jeb.124495] [Citation(s) in RCA: 96] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Memory is a process in which information is encoded, stored, and retrieved. For vertebrates, the modern view has been that it occurs only in the brain. This review describes a cellular memory in skeletal muscle in which hypertrophy is 'remembered' such that a fibre that has previously been large, but subsequently lost its mass, can regain mass faster than naive fibres. A new cell biological model based on the literature, with the most reliable methods for identifying myonuclei, can explain this phenomenon. According to this model, previously untrained fibres recruit myonuclei from activated satellite cells before hypertrophic growth. Even if subsequently subjected to grave atrophy, the higher number of myonuclei is retained, and the myonuclei seem to be protected against the elevated apoptotic activity observed in atrophying muscle tissue. Fibres that have acquired a higher number of myonuclei grow faster when subjected to overload exercise, thus the nuclei represent a functionally important 'memory' of previous strength. This memory might be very long lasting in humans, as myonuclei are stable for at least 15 years and might even be permanent. However, myonuclei are harder to recruit in the elderly, and if the long-lasting muscle memory also exists in humans, one should consider early strength training as a public health advice. In addition, myonuclei are recruited during steroid use and encode a muscle memory, at least in rodents. Thus, extending the exclusion time for doping offenders should be considered.
Collapse
Affiliation(s)
- Kristian Gundersen
- Department of Biosciences, University of Oslo, Blindernveien 31, Oslo N0316, Norway
| |
Collapse
|
40
|
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
|
41
|
|
42
|
Farup J, Dalgas U, Keytsman C, Eijnde BO, Wens I. High Intensity Training May Reverse the Fiber Type Specific Decline in Myogenic Stem Cells in Multiple Sclerosis Patients. Front Physiol 2016; 7:193. [PMID: 27303309 PMCID: PMC4885877 DOI: 10.3389/fphys.2016.00193] [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: 12/04/2015] [Accepted: 05/13/2016] [Indexed: 12/05/2022] Open
Abstract
Multiple sclerosis (MS) is associated with loss of skeletal muscle mass and function. The myogenic stem cells (satellite cells—SCs) are instrumental to accretion of myonuclei, but remain to be investigated in MS. The present study aimed to compare the SC and myonuclei content between MS patients (n = 23) and age matched healthy controls (HC, n = 18). Furthermore, the effects of 12 weeks of high intensity training on SC and myonuclei content were explored in MS. Muscle biopsies were obtained from m. Vastus Lateralis at baseline (MS and HC) and following 12 weeks of training (MS only). Frozen biopsies were sectioned followed by immunohistochemical analysis for fiber type specific SCs (Pax7+), myonuclei (MN) and central nuclei content and fiber cross-sectional area (fCSA) was quantified using ATPase histochemistry. At baseline the SCs per fiber was lower in type II compared to type I fibers in both MS (119%, p < 0.01) and HC (69%, p < 0.05), whereas the SCs per fCSA was lower in type II fibers compared to type I only in MS (72%, p < 0.05). No differences were observed in MN or central nuclei between MS and HC. Following training the type II fiber SCs per fiber and per fCSA in MS patients increased by 165% (p < 0.05) and 135% (p < 0.05), respectively. Furthermore, the type II fiber MN content tended (p = 0.06) to be increased by 35% following training. In conclusion, the SC content is lower in type II compared to type I fibers in both MS and HC. Furthermore, high intensity training was observed to selectively increase the SC and myonuclei content in type II fibers in MS patients.
Collapse
Affiliation(s)
- Jean Farup
- Section of Sport Science, Department of Public Health, Aarhus UniversityAarhus, Denmark; Research Laboratory for Biochemical Pathology, Department of Clinical Medicine, Aarhus UniversityAarhus, Denmark
| | - Ulrik Dalgas
- Section of Sport Science, Department of Public Health, Aarhus University Aarhus, Denmark
| | - Charly Keytsman
- Faculty of Medicine and Life Sciences, REVAL Rehabilitation Research Center, BIOMED Biomedical Research Institute, Hasselt University Diepenbeek, Belgium
| | - Bert O Eijnde
- Faculty of Medicine and Life Sciences, REVAL Rehabilitation Research Center, BIOMED Biomedical Research Institute, Hasselt University Diepenbeek, Belgium
| | - Inez Wens
- Faculty of Medicine and Life Sciences, REVAL Rehabilitation Research Center, BIOMED Biomedical Research Institute, Hasselt University Diepenbeek, Belgium
| |
Collapse
|
43
|
Guo AY, Leung KS, Qin JH, Chow SKH, Cheung WH. Effect of Low-Magnitude, High-Frequency Vibration Treatment on Retardation of Sarcopenia: Senescence-Accelerated Mouse-P8 Model. Rejuvenation Res 2016; 19:293-302. [PMID: 26608404 DOI: 10.1089/rej.2015.1759] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Sarcopenia-related falls and fall-related injuries in community-dwelling elderly people garnered more and more interest in recent years. Low-magnitude high-frequency vibration (LMHFV) was proven beneficial to musculoskeletal system and recommended for sarcopenia treatment. This study aimed to evaluate the effects of LMHFV on the sarcopenic animals and explore the mechanism of the stimulatory effects. Senescence-accelerated mouse P8 (SAMP8) mice at month 6 were randomized into control (Ctrl) and vibration (Vib) groups and the mice in the Vib group were given LMHFV (0.3 g, 20 min/day, 5 days/week) treatment. At months 0, 1, 2, 3, and 4 post-treatment, muscle mass, structure, and function were assessed. The potential proliferation capacity of the muscle was also evaluated by investigating satellite cells (SCs) pool and serum myostatin expression. At late stage, the mice in the Vib group showed higher muscle strength (month 4, p = 0.028). Generally, contractibility was significantly improved by LMHFV (contraction time [CT], p = 0.000; half-relaxation time [RT50], p = 0.000). Enlarged cross-sectional area of fiber type IIA was observed in the Vib group when compared with Ctrl group (p = 0.000). No significant difference of muscle mass was observed. The promotive effect of LMHFV on myoregeneration was reflected by suppressed SC pool reduction (month 3, p = 0.000; month 4, p = 0.000) and low myostatin expression (p = 0.052). LMHFV significantly improved the structural and functional outcomes of the skeletal muscle, hence retarding the progress of sarcopenia in SAMP8. It would be a good recommendation for prevention of the diseases related to skeletal muscle atrophy.
Collapse
Affiliation(s)
- An-Yun Guo
- 1 Department of Orthopaedics and Traumatology, Prince of Wales Hospital, The Chinese University of Hong Kong , Hong Kong SAR, China
| | - Kwok-Sui Leung
- 1 Department of Orthopaedics and Traumatology, Prince of Wales Hospital, The Chinese University of Hong Kong , Hong Kong SAR, China .,2 Translational Medicine Research & Development Center, Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology , Chinese Academy of Sciences, Beijing, China
| | - Jiang-Hui Qin
- 1 Department of Orthopaedics and Traumatology, Prince of Wales Hospital, The Chinese University of Hong Kong , Hong Kong SAR, China
| | - Simon Kwoon-Ho Chow
- 1 Department of Orthopaedics and Traumatology, Prince of Wales Hospital, The Chinese University of Hong Kong , Hong Kong SAR, China
| | - Wing-Hoi Cheung
- 1 Department of Orthopaedics and Traumatology, Prince of Wales Hospital, The Chinese University of Hong Kong , Hong Kong SAR, China .,2 Translational Medicine Research & Development Center, Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology , Chinese Academy of Sciences, Beijing, China .,3 The CUHK-ACC Space Medicine Centre on Health Maintenance of Musculoskeletal System, The Chinese University of Hong Kong Shenzhen Research Institute , Shenzhen, China
| |
Collapse
|
44
|
Goodman CA, Hornberger TA, Robling AG. Bone and skeletal muscle: Key players in mechanotransduction and potential overlapping mechanisms. Bone 2015; 80:24-36. [PMID: 26453495 PMCID: PMC4600534 DOI: 10.1016/j.bone.2015.04.014] [Citation(s) in RCA: 107] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2014] [Revised: 03/18/2015] [Accepted: 04/07/2015] [Indexed: 12/16/2022]
Abstract
The development and maintenance of skeletal muscle and bone mass is critical for movement, health and issues associated with the quality of life. Skeletal muscle and bone mass are regulated by a variety of factors that include changes in mechanical loading. Moreover, bone mass is, in large part, regulated by muscle-derived mechanical forces and thus by changes in muscle mass/strength. A thorough understanding of the cellular mechanism(s) responsible for mechanotransduction in bone and skeletal muscle is essential for the development of effective exercise and pharmaceutical strategies aimed at increasing, and/or preventing the loss of, mass in these tissues. Thus, in this review we will attempt to summarize the current evidence for the major molecular mechanisms involved in mechanotransduction in skeletal muscle and bone. By examining the differences and similarities in mechanotransduction between these two tissues, it is hoped that this review will stimulate new insights and ideas for future research and promote collaboration between bone and muscle biologists.(1).
Collapse
Affiliation(s)
- Craig A Goodman
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI 53706, USA; Centre for Chronic Disease Prevention and Management, College of Health and Biomedicine, Victoria University, Melbourne, Australia; Institute of Sport, Exercise and Active Living (ISEAL), Victoria University, Melbourne, VIC, Australia.
| | - Troy A Hornberger
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Alexander G Robling
- Department of Anatomy and Cell Biology, Indiana University School of Medicine, Indianapolis, IN 46202, USA; Roudebush Veterans Affairs Medical Center, Indianapolis, IN 46202, USA; Department of Biomedical Engineering, Indiana University-Purdue University at Indianapolis, Indianapolis, IN 46202, USA
| |
Collapse
|
45
|
Morton RW, McGlory C, Phillips SM. Nutritional interventions to augment resistance training-induced skeletal muscle hypertrophy. Front Physiol 2015; 6:245. [PMID: 26388782 PMCID: PMC4558471 DOI: 10.3389/fphys.2015.00245] [Citation(s) in RCA: 158] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2015] [Accepted: 08/17/2015] [Indexed: 11/16/2022] Open
Abstract
Skeletal muscle mass is regulated by a balance between muscle protein synthesis (MPS) and muscle protein breakdown (MPB). In healthy humans, MPS is more sensitive (varying 4–5 times more than MPB) to changes in protein feeding and loading rendering it the primary locus determining gains in muscle mass. Performing resistance exercise (RE) followed by the consumption of protein results in an augmentation of MPS and, over time, can lead to muscle hypertrophy. The magnitude of the RE-induced increase in MPS is dictated by a variety of factors including: the dose of protein, source of protein, and possibly the distribution and timing of post-exercise protein ingestion. In addition, RE variables such as frequency of sessions, time under tension, volume, and training status play roles in regulating MPS. This review provides a brief overview of our current understanding of how RE and protein ingestion can influence gains in skeletal muscle mass in young, healthy individuals. It is the goal of this review to provide nutritional recommendations for optimal skeletal muscle adaptation. Specifically, we will focus on how the manipulation of protein intake during the recovery period following RE augments the adaptive response.
Collapse
Affiliation(s)
- Robert W Morton
- Exercise Metabolism Research Group, Department of Kinesiology, McMaster University Hamilton, ON, Canada
| | - Chris McGlory
- Exercise Metabolism Research Group, Department of Kinesiology, McMaster University Hamilton, ON, Canada
| | - Stuart M Phillips
- Exercise Metabolism Research Group, Department of Kinesiology, McMaster University Hamilton, ON, Canada
| |
Collapse
|
46
|
Hjorth M, Norheim F, Meen AJ, Pourteymour S, Lee S, Holen T, Jensen J, Birkeland KI, Martinov VN, Langleite TM, Eckardt K, Drevon CA, Kolset SO. The effect of acute and long-term physical activity on extracellular matrix and serglycin in human skeletal muscle. Physiol Rep 2015; 3:e12473. [PMID: 26290530 PMCID: PMC4562559 DOI: 10.14814/phy2.12473] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2015] [Revised: 07/01/2015] [Accepted: 07/02/2015] [Indexed: 12/20/2022] Open
Abstract
Remodeling of extracellular matrix (ECM), including regulation of proteoglycans in skeletal muscle can be important for physiological adaptation to exercise. To investigate the effects of acute and long-term exercise on the expression of ECM-related genes and proteoglycans in particular, 26 middle-aged, sedentary men underwent a 12 weeks supervised endurance and strength training intervention and two acute, 45 min bicycle tests (70% VO2max), one at baseline and one after 12 weeks of training. Total gene expression in biopsies from m. vastus lateralis was measured with deep mRNA sequencing. After 45 min of bicycling approximately 550 gene transcripts were >50% upregulated. Of these, 28 genes (5%) were directly related to ECM. In response to long-term exercise of 12 weeks 289 genes exhibited enhanced expression (>50%) and 20% of them were ECM related. Further analyses of proteoglycan mRNA expression revealed that more than half of the proteoglycans expressed in muscle were significantly enhanced after 12 weeks intervention. The proteoglycan serglycin (SRGN) has not been studied in skeletal muscle and was one of few proteoglycans that showed increased expression after acute (2.2-fold, P < 0.001) as well as long-term exercise (1.4-fold, P < 0.001). Cultured, primary human skeletal muscle cells expressed and secreted SRGN. When the expression of SRGN was knocked down, the expression and secretion of serpin E1 (SERPINE1) increased. In conclusion, acute and especially long-term exercise promotes enhanced expression of several ECM components and proteoglycans. SRGN is a novel exercise-regulated proteoglycan in skeletal muscle with a potential role in exercise adaptation.
Collapse
Affiliation(s)
- Marit Hjorth
- Department of Nutrition, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Frode Norheim
- Department of Nutrition, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Astri J Meen
- Department of Nutrition, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Shirin Pourteymour
- Department of Nutrition, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Sindre Lee
- Department of Nutrition, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Torgeir Holen
- Department of Nutrition, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Jørgen Jensen
- Department of Physical Performance, Norwegian School of Sport Sciences, Oslo, Norway
| | - Kåre I Birkeland
- Department of Endocrinology, Morbid Obesity and Preventive Medicine, Oslo University Hospital and Institute of Clinical Medicine University of Oslo, Oslo, Norway
| | - Vladimir N Martinov
- Department of Nutrition, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Torgrim M Langleite
- Department of Nutrition, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, Oslo, Norway Department of Endocrinology, Morbid Obesity and Preventive Medicine, Oslo University Hospital and Institute of Clinical Medicine University of Oslo, Oslo, Norway
| | - Kristin Eckardt
- Department of Nutrition, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Christian A Drevon
- Department of Nutrition, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Svein O Kolset
- Department of Nutrition, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, Oslo, Norway
| |
Collapse
|
47
|
Farup J, Madaro L, Puri PL, Mikkelsen UR. Interactions between muscle stem cells, mesenchymal-derived cells and immune cells in muscle homeostasis, regeneration and disease. Cell Death Dis 2015. [PMID: 26203859 PMCID: PMC4650743 DOI: 10.1038/cddis.2015.198] [Citation(s) in RCA: 115] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Recent evidence has revealed the importance of reciprocal functional interactions between different types of mononuclear cells in coordinating the repair of injured muscles. In particular, signals released from the inflammatory infiltrate and from mesenchymal interstitial cells (also known as fibro-adipogenic progenitors (FAPs)) appear to instruct muscle stem cells (satellite cells) to break quiescence, proliferate and differentiate. Interestingly, conditions that compromise the functional integrity of this network can bias muscle repair toward pathological outcomes that are typically observed in chronic muscular disorders, that is, fibrotic and fatty muscle degeneration as well as myofiber atrophy. In this review, we will summarize the current knowledge on the regulation of this network in physiological and pathological conditions, and anticipate the potential contribution of its cellular components to relatively unexplored conditions, such as aging and physical exercise.
Collapse
Affiliation(s)
- J Farup
- Section for Sports Science, Institute of Public Health, Aarhus University, Aarhus, Denmark
| | - L Madaro
- 1] Sanford-Burnham Medical Research Institute, Sanford Children's Health Research Center, La Jolla, CA, USA [2] IRCCS Fondazione Santa Lucia, Rome, Italy
| | - P L Puri
- 1] Sanford-Burnham Medical Research Institute, Sanford Children's Health Research Center, La Jolla, CA, USA [2] IRCCS Fondazione Santa Lucia, Rome, Italy
| | - U R Mikkelsen
- 1] Section for Sports Science, Institute of Public Health, Aarhus University, Aarhus, Denmark [2] Institute of Sports Medicine, Department of Orthopaedic Surgery M, Bispebjerg Hospital and Center for Healthy Aging, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
48
|
Biressi S, Gopinath SD. The quasi-parallel lives of satellite cells and atrophying muscle. Front Aging Neurosci 2015; 7:140. [PMID: 26257645 PMCID: PMC4510774 DOI: 10.3389/fnagi.2015.00140] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2015] [Accepted: 07/06/2015] [Indexed: 12/25/2022] Open
Abstract
Skeletal muscle atrophy or wasting accompanies various chronic illnesses and the aging process, thereby reducing muscle function. One of the most important components contributing to effective muscle repair in postnatal organisms, the satellite cells (SCs), have recently become the focus of several studies examining factors participating in the atrophic process. We critically examine here the experimental evidence linking SC function with muscle loss in connection with various diseases as well as aging, and in the subsequent recovery process. Several recent reports have investigated the changes in SCs in terms of their differentiation and proliferative capacity in response to various atrophic stimuli. In this regard, we review the molecular changes within SCs that contribute to their dysfunctional status in atrophy, with the intention of shedding light on novel potential pharmacological targets to counteract the loss of muscle mass.
Collapse
Affiliation(s)
- Stefano Biressi
- Dulbecco Telethon Institute and Centre for Integrative Biology (CIBIO), University of TrentoTrento, Italy
| | | |
Collapse
|
49
|
Frontera WR, Ochala J. Skeletal muscle: a brief review of structure and function. Calcif Tissue Int 2015; 96:183-95. [PMID: 25294644 DOI: 10.1007/s00223-014-9915-y] [Citation(s) in RCA: 908] [Impact Index Per Article: 90.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2014] [Accepted: 09/16/2014] [Indexed: 01/23/2023]
Abstract
Skeletal muscle is one of the most dynamic and plastic tissues of the human body. In humans, skeletal muscle comprises approximately 40% of total body weight and contains 50-75% of all body proteins. In general, muscle mass depends on the balance between protein synthesis and degradation and both processes are sensitive to factors such as nutritional status, hormonal balance, physical activity/exercise, and injury or disease, among others. In this review, we discuss the various domains of muscle structure and function including its cytoskeletal architecture, excitation-contraction coupling, energy metabolism, and force and power generation. We will limit the discussion to human skeletal muscle and emphasize recent scientific literature on single muscle fibers.
Collapse
Affiliation(s)
- Walter R Frontera
- Department of Physical Medicine and Rehabilitation, Vanderbilt University School of Medicine, Suite 1318, 2201 Children's Way, Nashville, TN, 37212, USA,
| | | |
Collapse
|
50
|
Yates DT, Clarke DS, Macko AR, Anderson MJ, Shelton LA, Nearing M, Allen RE, Rhoads RP, Limesand SW. Myoblasts from intrauterine growth-restricted sheep fetuses exhibit intrinsic deficiencies in proliferation that contribute to smaller semitendinosus myofibres. J Physiol 2014; 592:3113-25. [PMID: 24860171 PMCID: PMC4214663 DOI: 10.1113/jphysiol.2014.272591] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2014] [Accepted: 05/09/2014] [Indexed: 12/21/2022] Open
Abstract
Intrauterine growth restriction (IUGR) reduces skeletal muscle mass in fetuses and offspring. Our objective was to determine whether myoblast dysfunction due to intrinsic cellular deficiencies or serum factors reduces myofibre hypertrophy in IUGR fetal sheep. At 134 days, IUGR fetuses weighed 67% less (P < 0.05) than controls and had smaller (P < 0.05) carcasses and semitendinosus myofibre areas. IUGR semitendinosus muscles had similar percentages of pax7-positive nuclei and pax7 mRNA but lower (P < 0.05) percentages of myogenin-positive nuclei (7 ± 2% and 13 ± 2%), less myoD and myogenin mRNA, and fewer (P < 0.05) proliferating myoblasts (PNCA-positive-pax7-positive) than controls (44 ± 2% vs. 52 ± 1%). Primary myoblasts were isolated from hindlimb muscles, and after 3 days in growth media (20% fetal bovine serum, FBS), myoblasts from IUGR fetuses had 34% fewer (P < 0.05) myoD-positive cells than controls and replicated 20% less (P < 0.05) during a 2 h BrdU pulse. IUGR myoblasts also replicated less (P < 0.05) than controls during a BrdU pulse after 3 days in media containing 10% control or IUGR fetal sheep serum (FSS). Both myoblast types replicated less (P < 0.05) with IUGR FSS-supplemented media compared to control FSS-supplemented media. In differentiation-promoting media (2% FBS), IUGR and control myoblasts had similar percentages of myogenin-positive nuclei after 5 days and formed similar-sized myotubes after 7 days. We conclude that intrinsic cellular deficiencies in IUGR myoblasts and factors in IUGR serum diminish myoblast proliferation and myofibre size in IUGR fetuses, but intrinsic myoblast deficiencies do not affect differentiation. Furthermore, the persistent reduction in IUGR myoblast replication shows adaptive deficiencies that explain poor muscle growth in IUGR newborn offspring.
Collapse
Affiliation(s)
- Dustin T Yates
- School of Animal and Comparative Biomedical Sciences, The University of Arizona, Tucson, AZ, USA
| | - Derek S Clarke
- School of Animal and Comparative Biomedical Sciences, The University of Arizona, Tucson, AZ, USA
| | - Antoni R Macko
- School of Animal and Comparative Biomedical Sciences, The University of Arizona, Tucson, AZ, USA
| | - Miranda J Anderson
- School of Animal and Comparative Biomedical Sciences, The University of Arizona, Tucson, AZ, USA
| | - Leslie A Shelton
- School of Animal and Comparative Biomedical Sciences, The University of Arizona, Tucson, AZ, USA
| | - Marie Nearing
- School of Animal and Comparative Biomedical Sciences, The University of Arizona, Tucson, AZ, USA
| | - Ronald E Allen
- School of Animal and Comparative Biomedical Sciences, The University of Arizona, Tucson, AZ, USA
| | - Robert P Rhoads
- Department of Animal and Poultry Sciences, Virginia Tech, Blacksburg, VA, USA
| | - Sean W Limesand
- School of Animal and Comparative Biomedical Sciences, The University of Arizona, Tucson, AZ, USA
| |
Collapse
|