1
|
Smith MJ, Hoffman NJ, Jose AJS, Burke LM, Opar DA. Nutritional Interventions to Attenuate Quadriceps Muscle Deficits following Anterior Cruciate Ligament Injury and Reconstruction. Sports Med 2025:10.1007/s40279-025-02174-w. [PMID: 39853659 DOI: 10.1007/s40279-025-02174-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/08/2025] [Indexed: 01/26/2025]
Abstract
Following anterior cruciate ligament (ACL) injury, quadriceps muscle atrophy persists despite rehabilitation, leading to loss of lower limb strength, osteoarthritis, poor knee joint health and reduced quality of life. However, the molecular mechanisms responsible for these deficits in hypertrophic adaptations within the quadriceps muscle following ACL injury and reconstruction are poorly understood. While resistance exercise training stimulates skeletal muscle hypertrophy, attenuation of these hypertrophic pathways can hinder rehabilitation following ACL injury and reconstruction, and ultimately lead to skeletal muscle atrophy that persists beyond ACL reconstruction, similar to disuse atrophy. Numerous studies have documented beneficial roles of nutritional support, including nutritional supplementation, in maintaining and/or increasing muscle mass. There are three main mechanisms by which nutritional supplementation may attenuate muscle atrophy and promote hypertrophy: (1) by directly affecting muscle protein synthetic machinery; (2) indirectly increasing an individual's ability to work harder; and/or (3) directly affecting satellite cell proliferation and differentiation. We propose that nutritional support may enhance rehabilitative responses to exercise training and positively impact molecular machinery underlying muscle hypertrophy. As one of the fastest growing knee injuries worldwide, a better understanding of the potential mechanisms involved in quadriceps muscle deficits following ACL injury and reconstruction, and potential benefits of nutritional support, are required to help restore quadriceps muscle mass and/or strength. This review discusses our current understanding of the molecular mechanisms involved in muscle hypertrophy and disuse atrophy, and how nutritional supplements may leverage these pathways to maximise recovery from ACL injury and reconstruction.
Collapse
Affiliation(s)
- Miriam J Smith
- School of Behavioural and Health Sciences, Australian Catholic University, Melbourne, VIC, Australia
- Sports Performance, Recovery, Injury and New Technologies (SPRINT) Research Centre, Australian Catholic University, Melbourne, VIC, Australia
- Exercise and Nutrition Research Program, Mary MacKillop Institute for Health Research, Australian Catholic University, Melbourne, VIC, Australia
| | - Nolan J Hoffman
- Exercise and Nutrition Research Program, Mary MacKillop Institute for Health Research, Australian Catholic University, Melbourne, VIC, Australia
| | - Argell J San Jose
- School of Behavioural and Health Sciences, Australian Catholic University, Melbourne, VIC, Australia
- OrthoSport Victoria Institute (OSVi), Richmond, VIC, Australia
| | - Louise M Burke
- Exercise and Nutrition Research Program, Mary MacKillop Institute for Health Research, Australian Catholic University, Melbourne, VIC, Australia
| | - David A Opar
- School of Behavioural and Health Sciences, Australian Catholic University, Melbourne, VIC, Australia.
- Sports Performance, Recovery, Injury and New Technologies (SPRINT) Research Centre, Australian Catholic University, Melbourne, VIC, Australia.
- , Level 1, Daniel Mannix Building, 17 Young Street, Fitzroy, VIC, 3065, Australia.
| |
Collapse
|
2
|
Liu X, Ye L, Ding Y, Gong W, Qian H, Jin K, Niu Y, Zuo Q, Song J, Han W, Chen G, Li B. Role of PI3K/AKT signaling pathway involved in self-renewing and maintaining biological properties of chicken primordial germ cells. Poult Sci 2024; 103:104140. [PMID: 39173217 PMCID: PMC11379996 DOI: 10.1016/j.psj.2024.104140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 06/29/2024] [Accepted: 07/25/2024] [Indexed: 08/24/2024] Open
Abstract
Avian primordial germ cells (PGCs) are important culture cells for the production of transgenic chickens and preservation of the genetic resources of endangered species; however, culturing these cells in vitro proves challenging. Although the proliferation of chicken PGCs is dependent on insulin, the underlying molecular mechanisms remain unclear. In the present study, we explored the expression of the PI3K/AKT signaling pathway in PGCs, investigated its effects on PGC self-renewal and biological properties, and identified the underlying mechanisms. Our findings indicated that although supplementation with the PI3K/AKT activator IGF-1 failed to promote proliferation under the assessed culture conditions, the PI3K/AKT inhibitor LY294002 resulted in retarded cell proliferation and reduced expression of germ cell-related markers. We further demonstrated that inhibition of PI3K/AKT regulates the cell cycle and promotes apoptosis in PGCs by activating the expression of BAX and inhibiting that of Bcl-2. These findings indicated that the PI3K/AKT pathway is required for cell renewal, apoptosis, and maintenance of the reproductive potential in chicken PGCs. This study aimed to provide a theoretical basis for the optimization and improvement of a culture system for chicken PGCs and provide insights into the self-renewal of vertebrate PGCs as well as potential evolutionary changes in this unique cell population.
Collapse
Affiliation(s)
- Xin Liu
- Key Laboratory of Animal Breeding Reproduction and Molecular Design for Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; Institutes of Agricultural Science and Technology Development, Yangzhou University, Yangzhou 225009, China; Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou University, Yangzhou 225009, China
| | - Liu Ye
- Key Laboratory of Animal Breeding Reproduction and Molecular Design for Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; Institutes of Agricultural Science and Technology Development, Yangzhou University, Yangzhou 225009, China; Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou University, Yangzhou 225009, China
| | - Ying Ding
- Key Laboratory of Animal Breeding Reproduction and Molecular Design for Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; Institutes of Agricultural Science and Technology Development, Yangzhou University, Yangzhou 225009, China; Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou University, Yangzhou 225009, China
| | - Wei Gong
- Key Laboratory of Animal Breeding Reproduction and Molecular Design for Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; Institutes of Agricultural Science and Technology Development, Yangzhou University, Yangzhou 225009, China; Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou University, Yangzhou 225009, China
| | - Hongwu Qian
- Key Laboratory of Animal Breeding Reproduction and Molecular Design for Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; Institutes of Agricultural Science and Technology Development, Yangzhou University, Yangzhou 225009, China; Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou University, Yangzhou 225009, China
| | - Kai Jin
- Key Laboratory of Animal Breeding Reproduction and Molecular Design for Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; Institutes of Agricultural Science and Technology Development, Yangzhou University, Yangzhou 225009, China; Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou University, Yangzhou 225009, China
| | - Yingjie Niu
- Key Laboratory of Animal Breeding Reproduction and Molecular Design for Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; Institutes of Agricultural Science and Technology Development, Yangzhou University, Yangzhou 225009, China; Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou University, Yangzhou 225009, China
| | - Qisheng Zuo
- Key Laboratory of Animal Breeding Reproduction and Molecular Design for Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; Institutes of Agricultural Science and Technology Development, Yangzhou University, Yangzhou 225009, China; Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou University, Yangzhou 225009, China
| | - Jiuzhou Song
- Animal & Avian Sciences, University of Maryland, College Park, MA 20742, USA
| | - Wei Han
- Poultry Institute, Chinese Academy of Agricultural Sciences Poultry Institute of Jiangsu, Yangzhou 225003, China
| | - Guohong Chen
- Key Laboratory of Animal Breeding Reproduction and Molecular Design for Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; Institutes of Agricultural Science and Technology Development, Yangzhou University, Yangzhou 225009, China; Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou University, Yangzhou 225009, China
| | - Bichun Li
- Key Laboratory of Animal Breeding Reproduction and Molecular Design for Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; Institutes of Agricultural Science and Technology Development, Yangzhou University, Yangzhou 225009, China; Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou University, Yangzhou 225009, China; College of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang 212100, China.
| |
Collapse
|
3
|
Thomas ACQ, Stead CA, Burniston JG, Phillips SM. Exercise-specific adaptations in human skeletal muscle: Molecular mechanisms of making muscles fit and mighty. Free Radic Biol Med 2024; 223:341-356. [PMID: 39147070 DOI: 10.1016/j.freeradbiomed.2024.08.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 07/30/2024] [Accepted: 08/09/2024] [Indexed: 08/17/2024]
Abstract
The mechanisms leading to a predominantly hypertrophied phenotype versus a predominantly oxidative phenotype, the hallmarks of resistance training (RT) or aerobic training (AT), respectively, are being unraveled. In humans, exposure of naïve persons to either AT or RT results in their skeletal muscle exhibiting generic 'exercise stress-related' signaling, transcription, and translation responses. However, with increasing engagement in AT or RT, the responses become refined, and the phenotype typically associated with each form of exercise emerges. Here, we review some of the mechanisms underpinning the adaptations of how muscles become, through AT, 'fit' and RT, 'mighty.' Much of our understanding of molecular exercise physiology has arisen from targeted analysis of post-translational modifications and measures of protein synthesis. Phosphorylation of specific residue sites has been a dominant focus, with canonical signaling pathways (AMPK and mTOR) studied extensively in the context of AT and RT, respectively. These alone, along with protein synthesis, have only begun to elucidate key differences in AT and RT signaling. Still, key yet uncharacterized differences exist in signaling and regulation of protein synthesis that drive unique adaptation to AT and RT. Omic studies are required to better understand the divergent relationship between exercise and phenotypic outcomes of training.
Collapse
Affiliation(s)
- Aaron C Q Thomas
- Protein Metabolism Research Lab, Department of Kinesiology, McMaster University, Hamilton, ON, Canada; Research Institute for Sport & Exercise Sciences, Liverpool John Moores University, Liverpool, United Kingdom
| | - Connor A Stead
- Research Institute for Sport & Exercise Sciences, Liverpool John Moores University, Liverpool, United Kingdom
| | - Jatin G Burniston
- Research Institute for Sport & Exercise Sciences, Liverpool John Moores University, Liverpool, United Kingdom
| | - Stuart M Phillips
- Protein Metabolism Research Lab, Department of Kinesiology, McMaster University, Hamilton, ON, Canada.
| |
Collapse
|
4
|
Roberts MD, McCarthy JJ, Hornberger TA, Phillips SM, Mackey AL, Nader GA, Boppart MD, Kavazis AN, Reidy PT, Ogasawara R, Libardi CA, Ugrinowitsch C, Booth FW, Esser KA. Mechanisms of mechanical overload-induced skeletal muscle hypertrophy: current understanding and future directions. Physiol Rev 2023; 103:2679-2757. [PMID: 37382939 PMCID: PMC10625844 DOI: 10.1152/physrev.00039.2022] [Citation(s) in RCA: 48] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 06/12/2023] [Accepted: 06/21/2023] [Indexed: 06/30/2023] Open
Abstract
Mechanisms underlying mechanical overload-induced skeletal muscle hypertrophy have been extensively researched since the landmark report by Morpurgo (1897) of "work-induced hypertrophy" in dogs that were treadmill trained. Much of the preclinical rodent and human resistance training research to date supports that involved mechanisms include enhanced mammalian/mechanistic target of rapamycin complex 1 (mTORC1) signaling, an expansion in translational capacity through ribosome biogenesis, increased satellite cell abundance and myonuclear accretion, and postexercise elevations in muscle protein synthesis rates. However, several lines of past and emerging evidence suggest that additional mechanisms that feed into or are independent of these processes are also involved. This review first provides a historical account of how mechanistic research into skeletal muscle hypertrophy has progressed. A comprehensive list of mechanisms associated with skeletal muscle hypertrophy is then outlined, and areas of disagreement involving these mechanisms are presented. Finally, future research directions involving many of the discussed mechanisms are proposed.
Collapse
Affiliation(s)
- Michael D Roberts
- School of Kinesiology, Auburn University, Auburn, Alabama, United States
| | - John J McCarthy
- Department of Physiology, College of Medicine, University of Kentucky, Lexington, Kentucky, United States
| | - Troy A Hornberger
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, Wisconsin, United States
| | - Stuart M Phillips
- Department of Kinesiology, McMaster University, Hamilton, Ontario, Canada
| | - Abigail L Mackey
- Institute of Sports Medicine Copenhagen, Department of Orthopedic Surgery, Copenhagen University Hospital-Bispebjerg and Frederiksberg, and Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Gustavo A Nader
- Department of Kinesiology and Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, Pennsylvania, United States
| | - Marni D Boppart
- Department of Kinesiology and Community Health, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States
| | - Andreas N Kavazis
- School of Kinesiology, Auburn University, Auburn, Alabama, United States
| | - Paul T Reidy
- Department of Kinesiology, Nutrition and Health, Miami University, Oxford, Ohio, United States
| | - Riki Ogasawara
- Healthy Food Science Research Group, Cellular and Molecular Biotechnology Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba, Japan
| | - Cleiton A Libardi
- MUSCULAB-Laboratory of Neuromuscular Adaptations to Resistance Training, Department of Physical Education, Federal University of São Carlos, São Carlos, Brazil
| | - Carlos Ugrinowitsch
- School of Physical Education and Sport, University of São Paulo, São Paulo, Brazil
| | - Frank W Booth
- Department of Biomedical Sciences, University of Missouri, Columbia, Missouri, United States
| | - Karyn A Esser
- Department of Physiology and Aging, College of Medicine, University of Florida, Gainesville, Florida, United States
| |
Collapse
|
5
|
Zhang Y, Wang L, Kang H, Lin CY, Fan Y. Unlocking the Therapeutic Potential of Irisin: Harnessing Its Function in Degenerative Disorders and Tissue Regeneration. Int J Mol Sci 2023; 24:ijms24076551. [PMID: 37047523 PMCID: PMC10095399 DOI: 10.3390/ijms24076551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 03/26/2023] [Accepted: 03/28/2023] [Indexed: 04/03/2023] Open
Abstract
Physical activity is well-established as an important protective factor against degenerative conditions and a promoter of tissue growth and renewal. The discovery of Fibronectin domain-containing protein 5 (FNDC5) as the precursor of Irisin in 2012 sparked significant interest in its potential as a diagnostic biomarker and a therapeutic agent for various diseases. Clinical studies have examined the correlation between plasma Irisin levels and pathological conditions using a range of assays, but the lack of reliable measurements for endogenous Irisin has led to uncertainty about its prognostic/diagnostic potential as an exercise surrogate. Animal and tissue-engineering models have shown the protective effects of Irisin treatment in reversing functional impairment and potentially permanent damage, but dosage ambiguities remain unresolved. This review provides a comprehensive examination of the clinical and basic studies of Irisin in the context of degenerative conditions and explores its potential as a therapeutic approach in the physiological processes involved in tissue repair/regeneration.
Collapse
Affiliation(s)
- Yuwei Zhang
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Centre for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100083, China
| | - Lizhen Wang
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Centre for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100083, China
- Correspondence:
| | - Hongyan Kang
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Centre for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100083, China
| | - Chia-Ying Lin
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Centre for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100083, China
- Department of Biomedical, Chemical & Environmental Engineering, University of Cincinnati, Cincinnati, OH 45267, USA
- Department of Orthopaedic Surgery, University of Cincinnati, Cincinnati, OH 45267, USA
- Department of Neurosurgery, University of Cincinnati, Cincinnati, OH 45267, USA
| | - Yubo Fan
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Centre for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100083, China
- School of Engineering Medicine, Beihang University, Beijing 100083, China
| |
Collapse
|
6
|
Lang CH. IMPORTANCE OF THE INNATE IMMUNE RESPONSE IN SKELETAL MUSCLE TO SEPSIS-INDUCED ALTERATIONS IN PROTEIN BALANCE. Shock 2023; 59:214-223. [PMID: 36730901 PMCID: PMC9957944 DOI: 10.1097/shk.0000000000002029] [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] [Indexed: 02/04/2023]
Abstract
ABSTRACT There is growing appreciation that skeletal muscle is a fully functional component of the body's innate immune system with the potential to actively participate in the host response to invading bacteria as opposed to being a passive target. In this regard, skeletal muscle in general and myocytes specifically possess an afferent limb that recognizes a wide variety of host pathogens via their interaction with multiple classes of cell membrane-bound and intracellular receptors, including toll-like receptors, cytokine receptors, NOD-like receptors, and the NLRP inflammasome. The efferent limb of the innate immune system in muscle is equally robust and with an increased synthesis and secretion of a variety of myocyte-derived cytokines (i.e., myokines), including TNF-α, IL-1, IL-6, and NO as well as multiple chemokines in response to appropriate stimulation. Herein, the current narrative review focuses primarily on the immune response of myocytes per se as opposed to other cell types within whole muscle. Moreover, because there are important differences, this review focuses specifically on systemic infection and inflammation as opposed to the response of muscle to direct injury and various types of muscular dystrophies. To date, however, there are few definitive muscle-specific studies that are necessary to directly address the relative importance of muscle-derived immune activation as a contributor to either the systemic immune response or the local immune microenvironment within muscle during sepsis and the resultant downstream metabolic disturbances.
Collapse
Affiliation(s)
- Charles H Lang
- Cellular and Molecular Physiology, Penn State College of Medicine, Hershey, Pennsylvania
| |
Collapse
|
7
|
Correia IM, da Silva Rodrigues G, Noronha NY, Watanabe LM, Luciano de Almeida M, Sobrinho ACDS, Nonino CB, Bueno Júnior CR. Older postmenopausal women with lower lean mass have hypermethylated sites in the PI3K-Akt pathway. Front Physiol 2023; 14:1150821. [PMID: 37123284 PMCID: PMC10143498 DOI: 10.3389/fphys.2023.1150821] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Accepted: 03/28/2023] [Indexed: 05/02/2023] Open
Abstract
Introduction: The decrease in lean mass is directly related to the loss of independence, muscle strength, and worse quality of life over the years. Although the genetic determinants of muscle mass were well recognized, recent literature has been uncovering new epigenetic factors affecting the state of muscular tissue. This study aimed to verify differences in the DNA methylation profile among Brazilian postmenopausal women aged 50-70 years according to the lean mass evaluation. Methods: A cross-sectional study comprised 40 women aged 50-70 years. After K-means cluster analysis the 40 participants were divided into two groups, the Lower Lean Mass group with 20 participants (61.1 ± 4.6 years) and the Higher Lean Mass group with 20 participants (60.7 ± 3.2 years). Lean mass was measured by dual-energy X-ray emission densitometry (DEXA). The participants' DNA was extracted using the Salting Out technique and subsequently, the Illumina 850k EPIC Infinium Methylation BeadChip was performed to obtain methylation data. Results: We obtained 1,913 differentially methylated sites (p ≤ 0.005 of β > 5% and β < -5%) in a total of 979 genes between groups (p ≤ 0.005; -5% > β > 5%). In addition, the PI3K-Akt pathway had the greatest power of significance with an FDR of 4.6 × 10-3. Conclusion: Our results demonstrate a differentiation between specific sites of different genes, which have essential functions in body composition and energy metabolism, supporting future studies that aim to relate lean mass with epigenetics.
Collapse
Affiliation(s)
- Igor Massari Correia
- School of Physical Education and Sport of Ribeirão Preto, University of São Paulo, São Paulo, Brazil
| | - Guilherme da Silva Rodrigues
- Department of Internal Medicine, Ribeirão Preto Medical School, University of São Paulo, São Paulo, Brazil
- *Correspondence: Guilherme da Silva Rodrigues,
| | - Natália Yumi Noronha
- Department of Internal Medicine, Ribeirão Preto Medical School, University of São Paulo, São Paulo, Brazil
| | - Lígia Moriguchi Watanabe
- Department of Health Sciences, Ribeirão Preto Medical School, University of São Paulo, São Paulo, Brazil
| | | | | | - Carla Barbosa Nonino
- College of Nursing of Ribeirão Preto, University of São Paulo, São Paulo, Brazil
| | - Carlos Roberto Bueno Júnior
- School of Physical Education and Sport of Ribeirão Preto, University of São Paulo, São Paulo, Brazil
- Department of Internal Medicine, Ribeirão Preto Medical School, University of São Paulo, São Paulo, Brazil
- College of Nursing of Ribeirão Preto, University of São Paulo, São Paulo, Brazil
| |
Collapse
|
8
|
Molecular mechanisms of exercise contributing to tissue regeneration. Signal Transduct Target Ther 2022; 7:383. [PMID: 36446784 PMCID: PMC9709153 DOI: 10.1038/s41392-022-01233-2] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 10/03/2022] [Accepted: 10/17/2022] [Indexed: 12/03/2022] Open
Abstract
Physical activity has been known as an essential element to promote human health for centuries. Thus, exercise intervention is encouraged to battle against sedentary lifestyle. Recent rapid advances in molecular biotechnology have demonstrated that both endurance and resistance exercise training, two traditional types of exercise, trigger a series of physiological responses, unraveling the mechanisms of exercise regulating on the human body. Therefore, exercise has been expected as a candidate approach of alleviating a wide range of diseases, such as metabolic diseases, neurodegenerative disorders, tumors, and cardiovascular diseases. In particular, the capacity of exercise to promote tissue regeneration has attracted the attention of many researchers in recent decades. Since most adult human organs have a weak regenerative capacity, it is currently a key challenge in regenerative medicine to improve the efficiency of tissue regeneration. As research progresses, exercise-induced tissue regeneration seems to provide a novel approach for fighting against injury or senescence, establishing strong theoretical basis for more and more "exercise mimetics." These drugs are acting as the pharmaceutical alternatives of those individuals who cannot experience the benefits of exercise. Here, we comprehensively provide a description of the benefits of exercise on tissue regeneration in diverse organs, mainly focusing on musculoskeletal system, cardiovascular system, and nervous system. We also discuss the underlying molecular mechanisms associated with the regenerative effects of exercise and emerging therapeutic exercise mimetics for regeneration, as well as the associated opportunities and challenges. We aim to describe an integrated perspective on the current advances of distinct physiological mechanisms associated with exercise-induced tissue regeneration on various organs and facilitate the development of drugs that mimics the benefits of exercise.
Collapse
|
9
|
Schoenfeld BJ, Wackerhage H, De Souza E. Inter-set stretch: A potential time-efficient strategy for enhancing skeletal muscle adaptations. Front Sports Act Living 2022; 4:1035190. [PMID: 36457663 PMCID: PMC9706104 DOI: 10.3389/fspor.2022.1035190] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Accepted: 11/02/2022] [Indexed: 08/10/2023] Open
Abstract
Time is considered a primary barrier to exercise adherence. Therefore, developing time-efficient resistance training (RT) strategies that optimize muscular adaptations is of primary interest to practitioners. A novel approach to the problem involves combining intensive stretch protocols with RT. Conceivably, integrating stretch into the inter-set period may provide an added stimulus for muscle growth without increasing session duration. Mechanistically, stretch can regulate anabolic signaling via both active and passive force sensors. Emerging evidence indicates that both lengthening contractions against a high load as well as passive stretch can acutely activate anabolic intracellular signaling pathways involved in muscle hypertrophy. Although longitudinal research investigating the effects of stretching between RT sets is limited, some evidence suggests it may in fact enhance hypertrophic adaptations. Accordingly, the purpose of this paper is threefold: (1) to review how the active force of a muscle contraction and the force of a passive stretched are sensed; (2) to present evidence for the effectiveness of RT with inter-set stretch for muscle hypertrophy (3) to provide practical recommendations for application of inter-set stretch in program design as well as directions for future research.
Collapse
Affiliation(s)
- Brad J. Schoenfeld
- Department of Exercise Science and Recreation, Lehman College, Bronx, NY, United States
| | - Henning Wackerhage
- Department of Sport and Exercise Sciences, Technical University of Munich, Munich, Germany
| | - Eduardo De Souza
- Department of Health Sciences and Human Performance, The University of Tampa, Tampa, FL, United States
| |
Collapse
|
10
|
Sato Y, Kawashima K, Fukui E, Matsumoto H, Yoshizawa F, Sato Y. Functional analysis reveals that Tinagl1 is required for normal muscle development in mice through the activation of ERK signaling. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2022; 1869:119294. [PMID: 35597451 DOI: 10.1016/j.bbamcr.2022.119294] [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: 02/18/2022] [Revised: 04/21/2022] [Accepted: 05/07/2022] [Indexed: 06/15/2023]
Abstract
Tinagl1 (tubulointerstitial nephritis antigen-like 1) is a matricellular protein involved in female infertility and breast cancer tumorigenesis. In this study, we analyzed the function of Tinagl1 in skeletal muscle using knockout mice and cell experiments. Although primary myoblasts isolated from Tinagl1-decifient (Tinagl1-/-) mice differentiated into normal myotubes, and treatment with recombinant Tinagl1 did not affect the proliferation or differentiation of C2C12 myoblasts, Tinagl1-/- mice exhibited reduced body mass and calf muscle weights compared to the control group (Tinagl1flox/flox). Furthermore, Tinagl1-/- mice showed myofibers with centrally located nuclei, which is a morphological marker of regenerating muscle or myopathy. In addition, the capillary density in the soleus muscle of Tinagl1-/- mice showed a decreasing trend compared to that of the control group. Importantly, si-RNA-mediated knockdown of TINAGL1 resulted in reduced tube formation in human umbilical vein endothelial cells (HUVECs), whereas treatment with Tinagl1 promoted tube formation. Immunoblot analysis revealed that Tinagl1 activates ERK signaling in both HUVECs and C2C12 myoblasts and myotubes, which are involved in the regulation of myogenic differentiation, proliferation, metabolism, and angiogenesis. Our results demonstrate that Tinagl1 may be required for normal muscle and capillary development through the activation of ERK signaling.
Collapse
Affiliation(s)
- Yoriko Sato
- Department of Animal Science, School of Agriculture, Tokai University, Kumamoto 8628652, Japan
| | - Keisuke Kawashima
- Department of Agrobiology and Bioresources, School of Agriculture, Utsunomiya University, Tochigi, 3218505, Japan
| | - Emiko Fukui
- Department of Agrobiology and Bioresources, School of Agriculture, Utsunomiya University, Tochigi, 3218505, Japan
| | - Hiromichi Matsumoto
- Department of Agrobiology and Bioresources, School of Agriculture, Utsunomiya University, Tochigi, 3218505, Japan
| | - Fumiaki Yoshizawa
- Department of Agrobiology and Bioresources, School of Agriculture, Utsunomiya University, Tochigi, 3218505, Japan
| | - Yusuke Sato
- Department of Animal Science, School of Agriculture, Tokai University, Kumamoto 8628652, Japan.
| |
Collapse
|
11
|
Attwaters M, Hughes SM. Cellular and molecular pathways controlling muscle size in response to exercise. FEBS J 2022; 289:1428-1456. [PMID: 33755332 DOI: 10.1111/febs.15820] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 02/27/2021] [Accepted: 03/12/2021] [Indexed: 12/14/2022]
Abstract
From the discovery of ATP and motor proteins to synaptic neurotransmitters and growth factor control of cell differentiation, skeletal muscle has provided an extreme model system in which to understand aspects of tissue function. Muscle is one of the few tissues that can undergo both increase and decrease in size during everyday life. Muscle size depends on its contractile activity, but the precise cellular and molecular pathway(s) by which the activity stimulus influences muscle size and strength remain unclear. Four correlates of muscle contraction could, in theory, regulate muscle growth: nerve-derived signals, cytoplasmic calcium dynamics, the rate of ATP consumption and physical force. Here, we summarise the evidence for and against each stimulus and what is known or remains unclear concerning their molecular signal transduction pathways and cellular effects. Skeletal muscle can grow in three ways, by generation of new syncytial fibres, addition of nuclei from muscle stem cells to existing fibres or increase in cytoplasmic volume/nucleus. Evidence suggests the latter two processes contribute to exercise-induced growth. Fibre growth requires increase in sarcolemmal surface area and cytoplasmic volume at different rates. It has long been known that high-force exercise is a particularly effective growth stimulus, but how this stimulus is sensed and drives coordinated growth that is appropriately scaled across organelles remains a mystery.
Collapse
Affiliation(s)
- Michael Attwaters
- Randall Centre for Cell and Molecular Biophysics, School of Basic and Medical Biosciences, King's College London, UK
| | - Simon M Hughes
- Randall Centre for Cell and Molecular Biophysics, School of Basic and Medical Biosciences, King's College London, UK
| |
Collapse
|
12
|
Cannataro R, Carbone L, Petro JL, Cione E, Vargas S, Angulo H, Forero DA, Odriozola-Martínez A, Kreider RB, Bonilla DA. Sarcopenia: Etiology, Nutritional Approaches, and miRNAs. Int J Mol Sci 2021; 22:9724. [PMID: 34575884 PMCID: PMC8466275 DOI: 10.3390/ijms22189724] [Citation(s) in RCA: 82] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Revised: 09/03/2021] [Accepted: 09/06/2021] [Indexed: 02/06/2023] Open
Abstract
Sarcopenia, an age-related decline in skeletal muscle mass and function, dramatically affects the quality of life. Although there is a consensus that sarcopenia is a multifactorial syndrome, the etiology and underlying mechanisms are not yet delineated. Moreover, research about nutritional interventions to prevent the development of sarcopenia is mainly focused on the amount and quality of protein intake. The impact of several nutrition strategies that consider timing of food intake, anti-inflammatory nutrients, metabolic control, and the role of mitochondrial function on the progression of sarcopenia is not fully understood. This narrative review summarizes the metabolic background of this phenomenon and proposes an integral nutritional approach (including dietary supplements such as creatine monohydrate) to target potential molecular pathways that may affect reduce or ameliorate the adverse effects of sarcopenia. Lastly, miRNAs, in particular those produced by skeletal muscle (MyomiR), might represent a valid tool to evaluate sarcopenia progression as a potential rapid and early biomarker for diagnosis and characterization.
Collapse
Affiliation(s)
- Roberto Cannataro
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende, Italy;
- Galascreen Laboratories, University of Calabria, 87036 Rende, Italy
- Research Division, Dynamical Business & Science Society, DBSS International SAS, Bogotá 110311, Colombia; (J.L.P.); (S.V.); (D.A.B.)
| | - Leandro Carbone
- Research Division, Dynamical Business & Science Society, DBSS International SAS, Bogotá 110311, Colombia; (J.L.P.); (S.V.); (D.A.B.)
- Faculty of Medicine, University of Salvador, Buenos Aires 1020, Argentina
| | - Jorge L. Petro
- Research Division, Dynamical Business & Science Society, DBSS International SAS, Bogotá 110311, Colombia; (J.L.P.); (S.V.); (D.A.B.)
- Research Group in Physical Activity, Sports and Health Sciences (GICAFS), Universidad de Córdoba, Montería 230002, Colombia
| | - Erika Cione
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende, Italy;
- Galascreen Laboratories, University of Calabria, 87036 Rende, Italy
| | - Salvador Vargas
- Research Division, Dynamical Business & Science Society, DBSS International SAS, Bogotá 110311, Colombia; (J.L.P.); (S.V.); (D.A.B.)
- Faculty of Sport Sciences, EADE-University of Wales Trinity Saint David, 29018 Málaga, Spain
| | - Heidy Angulo
- Grupo de Investigación Programa de Medicina (GINUMED), Corporación Universitaria Rafael Núñez, Cartagena 130001, Colombia;
| | - Diego A. Forero
- Health and Sport Sciences Research Group, School of Health and Sport Sciences, Fundación Universitaria del Área Andina, Bogotá 111221, Colombia;
| | - Adrián Odriozola-Martínez
- Sport Genomics Research Group, Department of Genetics, Physical Anthropology and Animal Physiology, University of the Basque Country UPV/EHU, 48940 Leioa, Spain;
- kDNA Genomics, Joxe Mari Korta Research Center, University of the Basque Country UPV/EHU, 20018 Donostia-San Sebastián, Spain
| | - Richard B. Kreider
- Exercise & Sport Nutrition Lab, Human Clinical Research Facility, Texas A&M University, College Station, TX 77843, USA;
| | - Diego A. Bonilla
- Research Division, Dynamical Business & Science Society, DBSS International SAS, Bogotá 110311, Colombia; (J.L.P.); (S.V.); (D.A.B.)
- Research Group in Physical Activity, Sports and Health Sciences (GICAFS), Universidad de Córdoba, Montería 230002, Colombia
- kDNA Genomics, Joxe Mari Korta Research Center, University of the Basque Country UPV/EHU, 20018 Donostia-San Sebastián, Spain
- Research Group in Biochemistry and Molecular Biology, Universidad Distrital Francisco José de Caldas, Bogotá 110311, Colombia
| |
Collapse
|
13
|
Eftestøl E, Franchi MV, Kasper S, Flück M. JNK activation in TA and EDL muscle is load-dependent in rats receiving identical excitation patterns. Sci Rep 2021; 11:16405. [PMID: 34385505 PMCID: PMC8361015 DOI: 10.1038/s41598-021-94930-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Accepted: 07/12/2021] [Indexed: 11/09/2022] Open
Abstract
As the excitation-contraction coupling is inseparable during voluntary exercise, the relative contribution of the mechanical and neural input on hypertrophy-related molecular signalling is still poorly understood. Herein, we use a rat in-vivo strength exercise model with an electrically-induced standardized excitation pattern, previously shown to induce a load-dependent increase in myonuclear number and hypertrophy, to study acute effects of load on molecular signalling. We assessed protein abundance and specific phosphorylation of the four protein kinases FAK, mTOR, p70S6K and JNK after 2, 10 and 28 min of a low- or high-load contraction, in order to assess the effects of load, exercise duration and muscle-type on their response to exercise. Specific phosphorylation of mTOR, p70S6K and JNK was increased after 28 min of exercise under the low- and high-load protocol. Elevated phosphorylation of mTOR and JNK was detectable already after 2 and 10 min of exercise, respectively, but greatest after 28 min of exercise, and JNK phosphorylation was highly load-dependent. The abundance of all four kinases was higher in TA compared to EDL muscle, p70S6K abundance was increased after exercise in a load-independent manner, and FAK and JNK abundance was reduced after 28 min of exercise in both the exercised and control muscles. In conclusion, the current study shows that JNK activation after a single resistance exercise is load-specific, resembling the previously reported degree of myonuclear accrual and muscle hypertrophy with repetition of the exercise stimulus.
Collapse
Affiliation(s)
- Einar Eftestøl
- Department of Biosciences, University of Oslo, Kristine Bonnevies hus, Blindernveien 31, 0371, Oslo, Norway.
| | - Martino V Franchi
- Laboratory for Muscle Plasticity, Department of Orthopaedics, University of Zürich, Zurich, Switzerland.,Department of Biomedical Sciences, University of Padova, Padua, Italy
| | - Stephanie Kasper
- Laboratory for Muscle Plasticity, Department of Orthopaedics, University of Zürich, Zurich, Switzerland
| | - Martin Flück
- Laboratory for Muscle Plasticity, Department of Orthopaedics, University of Zürich, Zurich, Switzerland
| |
Collapse
|
14
|
Gene Expression and Carcass Traits Are Different between Different Quality Grade Groups in Red-Faced Hereford Steers. Animals (Basel) 2021; 11:ani11071910. [PMID: 34198984 PMCID: PMC8300355 DOI: 10.3390/ani11071910] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 06/19/2021] [Accepted: 06/23/2021] [Indexed: 12/19/2022] Open
Abstract
Simple Summary Producing a consistent and positive experience for beef consumers is challenging. The gene expression in muscle at harvest may provide insight into better prediction of United States Department of Agriculture (USDA) quality grade. In this pilot study muscle samples were collected at harvest on sixteen steers with a similar background and identical management. Muscle transcripts were sequenced to determine gene expression. Transcripts related to the extracellular matrix, stem cell differentiation, and focal cell adhesions were differentially expressed in muscle tissue from carcasses with differing USDA quality grades. This confirmed the application of this technique to provide insight into muscle development and fat deposition necessary for better prediction and selection to improve consistency and consumer experience. Abstract Fat deposition is important to carcass value and some palatability characteristics. Carcasses with higher USDA quality grades produce more value for producers and processors in the US system and are more likely to have greater eating satisfaction. Using genomics to identify genes impacting marbling deposition provides insight into muscle biochemistry that may lead to ways to better predict fat deposition, especially marbling and thus quality grade. Hereford steers (16) were managed the same from birth through harvest after 270 days on feed. Samples were obtained for tenderness and transcriptome profiling. As expected, steaks from Choice carcasses had a lower shear force value than steaks from Select carcasses; however, steaks from Standard carcasses were not different from steaks from Choice carcasses. A significant number of differentially expressed (DE) genes was observed in the longissimus lumborum between Choice and Standard carcass RNA pools (1257 genes, p < 0.05), but not many DE genes were observed between Choice and Select RNA pools. Exploratory analysis of global muscle tissue transcriptome from Standard and Choice carcasses provided insight into muscle biochemistry, specifically the upregulation of extracellular matrix development and focal adhesion pathways and the downregulation of RNA processing and metabolism in Choice versus Standard. Additional research is needed to explore the function and timing of gene expression changes.
Collapse
|
15
|
Deane CS, Willis CRG, Phillips BE, Atherton PJ, Harries LW, Ames RM, Szewczyk NJ, Etheridge T. Transcriptomic meta-analysis of disuse muscle atrophy vs. resistance exercise-induced hypertrophy in young and older humans. J Cachexia Sarcopenia Muscle 2021; 12:629-645. [PMID: 33951310 PMCID: PMC8200445 DOI: 10.1002/jcsm.12706] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Revised: 02/26/2021] [Accepted: 03/29/2021] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Skeletal muscle atrophy manifests across numerous diseases; however, the extent of similarities/differences in causal mechanisms between atrophying conditions in unclear. Ageing and disuse represent two of the most prevalent and costly atrophic conditions, with resistance exercise training (RET) being the most effective lifestyle countermeasure. We employed gene-level and network-level meta-analyses to contrast transcriptomic signatures of disuse and RET, plus young and older RET to establish a consensus on the molecular features of, and therapeutic targets against, muscle atrophy in conditions of high socio-economic relevance. METHODS Integrated gene-level and network-level meta-analysis was performed on publicly available microarray data sets generated from young (18-35 years) m. vastus lateralis muscle subjected to disuse (unilateral limb immobilization or bed rest) lasting ≥7 days or RET lasting ≥3 weeks, and resistance-trained older (≥60 years) muscle. RESULTS Disuse and RET displayed predominantly separate transcriptional responses, and transcripts altered across conditions were mostly unidirectional. However, disuse and RET induced directly inverted expression profiles for mitochondrial function and translation regulation genes, with COX4I1, ENDOG, GOT2, MRPL12, and NDUFV2, the central hub components of altered mitochondrial networks, and ZMYND11, a hub gene of altered translation regulation. A substantial number of genes (n = 140) up-regulated post-RET in younger muscle were not similarly up-regulated in older muscle, with young muscle displaying a more pronounced extracellular matrix (ECM) and immune/inflammatory gene expression response. Both young and older muscle exhibited similar RET-induced ubiquitination/RNA processing gene signatures with associated PWP1, PSMB1, and RAF1 hub genes. CONCLUSIONS Despite limited opposing gene profiles, transcriptional signatures of disuse are not simply the converse of RET. Thus, the mechanisms of unloading cannot be derived from studying muscle loading alone and provides a molecular basis for understanding why RET fails to target all transcriptional features of disuse. Loss of RET-induced ECM mechanotransduction and inflammatory profiles might also contribute to suboptimal ageing muscle adaptations to RET. Disuse and age-dependent molecular candidates further establish a framework for understanding and treating disuse/ageing atrophy.
Collapse
Affiliation(s)
- Colleen S Deane
- Department of Sport and Health Sciences, College of Life and Environmental Sciences, University of Exeter, St. Luke's Campus, Exeter, UK.,Living Systems Institute, University of Exeter, Exeter, UK
| | - Craig R G Willis
- Department of Sport and Health Sciences, College of Life and Environmental Sciences, University of Exeter, St. Luke's Campus, Exeter, UK
| | - Bethan E Phillips
- MRC-ARUK Centre for Musculoskeletal Ageing Research and National Institute of Health Research, Biomedical Research Centre, Division of Medical Sciences and Graduate Entry Medicine, Royal Derby Hospital Centre, School of Medicine, University of Nottingham, Derby, UK
| | - Philip J Atherton
- MRC-ARUK Centre for Musculoskeletal Ageing Research and National Institute of Health Research, Biomedical Research Centre, Division of Medical Sciences and Graduate Entry Medicine, Royal Derby Hospital Centre, School of Medicine, University of Nottingham, Derby, UK
| | - Lorna W Harries
- RNA-Mediated Mechanisms of Disease Group, Institute of Biomedical and Clinical Sciences, University of Exeter Medical School, University of Exeter, Exeter, UK
| | - Ryan M Ames
- Living Systems Institute, University of Exeter, Exeter, UK
| | - Nathaniel J Szewczyk
- MRC-ARUK Centre for Musculoskeletal Ageing Research and National Institute of Health Research, Biomedical Research Centre, Division of Medical Sciences and Graduate Entry Medicine, Royal Derby Hospital Centre, School of Medicine, University of Nottingham, Derby, UK.,Ohio Musculoskeletal and Neurological Institute & Department of Biomedical Sciences, Ohio University, Athens, OH, USA
| | - Timothy Etheridge
- Department of Sport and Health Sciences, College of Life and Environmental Sciences, University of Exeter, St. Luke's Campus, Exeter, UK
| |
Collapse
|
16
|
Bouviere J, Fortunato RS, Dupuy C, Werneck-de-Castro JP, Carvalho DP, Louzada RA. Exercise-Stimulated ROS Sensitive Signaling Pathways in Skeletal Muscle. Antioxidants (Basel) 2021; 10:antiox10040537. [PMID: 33808211 PMCID: PMC8066165 DOI: 10.3390/antiox10040537] [Citation(s) in RCA: 90] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2021] [Revised: 03/16/2021] [Accepted: 03/26/2021] [Indexed: 12/11/2022] Open
Abstract
Physical exercise represents a major challenge to whole-body homeostasis, provoking acute and adaptative responses at the cellular and systemic levels. Different sources of reactive oxygen species (ROS) have been described in skeletal muscle (e.g., NADPH oxidases, xanthine oxidase, and mitochondria) and are closely related to the physiological changes induced by physical exercise through the modulation of several signaling pathways. Many signaling pathways that are regulated by exercise-induced ROS generation, such as adenosine monophosphate-activated protein kinase (AMPK), mitogen activated protein kinase (MAPK), nuclear respiratory factor2 (NRF2), and PGC-1α are involved in skeletal muscle responses to physical exercise, such as increased glucose uptake, mitochondriogenesis, and hypertrophy, among others. Most of these adaptations are blunted by antioxidants, revealing the crucial role played by ROS during and after physical exercise. When ROS generation is either insufficient or exacerbated, ROS-mediated signaling is disrupted, as well as physical exercise adaptations. Thus, an understanding the limit between "ROS that can promote beneficial effects" and "ROS that can promote harmful effects" is a challenging question in exercise biology. The identification of new mediators that cause reductive stress and thereby disrupt exercise-stimulated ROS signaling is a trending on this topic and are covered in this current review.
Collapse
Affiliation(s)
- Jessica Bouviere
- Institut of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, Brazil; (J.B.); (R.S.F.); (D.P.C.)
| | - Rodrigo S. Fortunato
- Institut of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, Brazil; (J.B.); (R.S.F.); (D.P.C.)
| | - Corinne Dupuy
- Université Paris-Saclay, UMR 9019CNRS, Gustave Roussy, 94800 Villejuif, France;
| | - Joao Pedro Werneck-de-Castro
- Division of Endocrinology, Diabetes and Metabolism, Miller School of Medicine, University of Miami, Miami, FL 33136, USA;
| | - Denise P. Carvalho
- Institut of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, Brazil; (J.B.); (R.S.F.); (D.P.C.)
| | - Ruy A. Louzada
- Institut of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, Brazil; (J.B.); (R.S.F.); (D.P.C.)
- Université Paris-Saclay, UMR 9019CNRS, Gustave Roussy, 94800 Villejuif, France;
- Division of Endocrinology, Diabetes and Metabolism, Miller School of Medicine, University of Miami, Miami, FL 33136, USA;
- Correspondence:
| |
Collapse
|
17
|
Bass JJ, Kazi AA, Deane CS, Nakhuda A, Ashcroft SP, 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. The mechanisms of skeletal muscle atrophy in response to transient knockdown of the vitamin D receptor in vivo. J Physiol 2021; 599:963-979. [PMID: 33258480 PMCID: PMC7986223 DOI: 10.1113/jp280652] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Accepted: 11/25/2020] [Indexed: 12/12/2022] Open
Abstract
KEY POINTS Reduced vitamin D receptor (VDR) expression prompts skeletal muscle atrophy. Atrophy occurs through catabolic processes, namely the induction of autophagy, while anabolism remains unchanged. In response to VDR-knockdown mitochondrial function and related gene-set expression is impaired. In vitro VDR knockdown induces myogenic dysregulation occurring through impaired differentiation. These results highlight the autonomous role the VDR has within skeletal muscle mass regulation. ABSTRACT Vitamin D deficiency is estimated to affect ∼40% of the world's population and has been associated with impaired muscle maintenance. Vitamin D exerts its actions through the vitamin D receptor (VDR), the expression of which was recently confirmed in skeletal muscle, and its down-regulation is linked to reduced muscle mass and functional decline. To identify potential mechanisms underlying muscle atrophy, we studied the impact of VDR knockdown (KD) on mature skeletal muscle in vivo, and myogenic regulation in vitro in C2C12 cells. Male Wistar rats underwent in vivo electrotransfer (IVE) to knock down the VDR in hind-limb tibialis anterior (TA) muscle for 10 days. Comprehensive metabolic and physiological analysis was undertaken to define the influence loss of the VDR on muscle fibre composition, protein synthesis, anabolic and catabolic signalling, mitochondrial phenotype and gene expression. Finally, in vitro lentiviral transfection was used to induce sustained VDR-KD in C2C12 cells to analyse myogenic regulation. Muscle VDR-KD elicited atrophy through a reduction in total protein content, resulting in lower myofibre area. Activation of autophagic processes was observed, with no effect upon muscle protein synthesis or anabolic signalling. Furthermore, RNA-sequencing analysis identified systematic down-regulation of multiple mitochondrial respiration-related protein and genesets. Finally, in vitro VDR-knockdown impaired myogenesis (cell cycling, differentiation and myotube formation). Together, these data indicate a fundamental regulatory role of the VDR in the regulation of myogenesis and muscle mass, whereby it acts to maintain muscle mitochondrial function and limit autophagy.
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 MedicineUniversity of NottinghamNottinghamUK
| | - Abid A. Kazi
- Department of Cellular and Molecular PhysiologyPennsylvania State University College of MedicineHersheyPAUSA
| | - Colleen S. Deane
- Department of Sport and Health SciencesUniversity of ExeterExeterUK
- Living Systems InstituteUniversity of ExeterExeterUK
| | - Asif Nakhuda
- MRC/ARUK Centre for Musculoskeletal Ageing Research and National Institute for Health Research (NIHR)Nottingham Biomedical Research Centre (BRC)School of MedicineUniversity of NottinghamNottinghamUK
| | - Stephen P. Ashcroft
- School of Sport, Exercise and Rehabilitation SciencesUniversity of BirminghamBirminghamUK
| | - Matthew S. Brook
- MRC/ARUK Centre for Musculoskeletal Ageing Research and National Institute for Health Research (NIHR)Nottingham Biomedical Research Centre (BRC)School of MedicineUniversity of NottinghamNottinghamUK
| | - Daniel J. Wilkinson
- MRC/ARUK Centre for Musculoskeletal Ageing Research and National Institute for Health Research (NIHR)Nottingham Biomedical Research Centre (BRC)School of MedicineUniversity of NottinghamNottinghamUK
| | - Bethan E. Phillips
- MRC/ARUK Centre for Musculoskeletal Ageing Research and National Institute for Health Research (NIHR)Nottingham Biomedical Research Centre (BRC)School of MedicineUniversity of NottinghamNottinghamUK
| | - Andrew Philp
- School of Sport, Exercise and Rehabilitation SciencesUniversity of BirminghamBirminghamUK
- Mitochondrial Metabolism & Ageing Laboratory, Diabetes and Metabolism DivisionGarvan Institute of Medical ResearchNew South WalesAustralia
- St Vincent's Medical School, UNSW Medicine, UNSWSydneyAustralia
| | - Janelle Tarum
- School of Health SciencesÖrebro UniversityÖrebroSweden
| | - Fawzi Kadi
- School of Health SciencesÖrebro UniversityÖrebroSweden
| | - Ditte Andersen
- Molecular Physiology of Diabetes LaboratoryDepartment of Comparative Biomedical SciencesRoyal Veterinary CollegeLondonUK
| | - Amadeo Muñoz Garcia
- Institute of Metabolism and Systems ResearchThe University of BirminghamBirminghamUK
- Department of Bioinformatics – BiGCaTNUTRIM School of Nutrition and Metabolism in Translational ResearchMaastricht UniversityMaastrichtThe Netherlands
| | - Ken Smith
- MRC/ARUK Centre for Musculoskeletal Ageing Research and National Institute for Health Research (NIHR)Nottingham Biomedical Research Centre (BRC)School of MedicineUniversity of NottinghamNottinghamUK
| | - Iain J. Gallagher
- Physiology, Exercise and Nutrition Research GroupFaculty of Health Sciences and SportUniversity of StirlingStirlingUK
| | - Nathaniel J. Szewczyk
- MRC/ARUK Centre for Musculoskeletal Ageing Research and National Institute for Health Research (NIHR)Nottingham Biomedical Research Centre (BRC)School of MedicineUniversity of NottinghamNottinghamUK
| | - Mark E. Cleasby
- Molecular Physiology of Diabetes LaboratoryDepartment of Comparative Biomedical SciencesRoyal Veterinary CollegeLondonUK
| | - Philip J Atherton
- MRC/ARUK Centre for Musculoskeletal Ageing Research and National Institute for Health Research (NIHR)Nottingham Biomedical Research Centre (BRC)School of MedicineUniversity of NottinghamNottinghamUK
| |
Collapse
|
18
|
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: 53] [Impact Index Per Article: 10.6] [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
|
19
|
FAK Signaling in Rhabdomyosarcoma. Int J Mol Sci 2020; 21:ijms21228422. [PMID: 33182556 PMCID: PMC7697003 DOI: 10.3390/ijms21228422] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 11/07/2020] [Accepted: 11/08/2020] [Indexed: 01/01/2023] Open
Abstract
Rhabdomyosarcoma (RMS) is the most common soft tissue sarcoma of children and adolescents. The fusion-positive (FP)-RMS variant expressing chimeric oncoproteins such as PAX3-FOXO1 and PAX7-FOXO1 is at high risk. The fusion negative subgroup, FN-RMS, has a good prognosis when non-metastatic. Despite a multimodal therapeutic approach, FP-RMS and metastatic FN-RMS often show a dismal prognosis with 5-year survival of less than 30%. Therefore, novel targets need to be discovered to develop therapies that halt tumor progression, reducing long-term side effects in young patients. Focal adhesion kinase (FAK) is a non-receptor tyrosine kinase that regulates focal contacts at the cellular edges. It plays a role in cell motility, survival, and proliferation in response to integrin and growth factor receptors’ activation. FAK is often dysregulated in cancer, being upregulated and/or overactivated in several adult and pediatric tumor types. In RMS, both in vitro and preclinical studies point to a role of FAK in tumor cell motility/invasion and proliferation, which is inhibited by FAK inhibitors. In this review, we summarize the data on FAK expression and modulation in RMS. Moreover, we give an overview of the approaches to inhibit FAK in both preclinical and clinical cancer settings.
Collapse
|
20
|
Mirzoev TM. Skeletal Muscle Recovery from Disuse Atrophy: Protein Turnover Signaling and Strategies for Accelerating Muscle Regrowth. Int J Mol Sci 2020; 21:ijms21217940. [PMID: 33114683 PMCID: PMC7663166 DOI: 10.3390/ijms21217940] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Revised: 10/08/2020] [Accepted: 10/23/2020] [Indexed: 12/15/2022] Open
Abstract
Skeletal muscle fibers have a unique capacity to adjust their metabolism and phenotype in response to alternations in mechanical loading. Indeed, chronic mechanical loading leads to an increase in skeletal muscle mass, while prolonged mechanical unloading results in a significant decrease in muscle mass (muscle atrophy). The maintenance of skeletal muscle mass is dependent on the balance between rates of muscle protein synthesis and breakdown. While molecular mechanisms regulating protein synthesis during mechanical unloading have been relatively well studied, signaling events implicated in protein turnover during skeletal muscle recovery from unloading are poorly defined. A better understanding of the molecular events that underpin muscle mass recovery following disuse-induced atrophy is of significant importance for both clinical and space medicine. This review focuses on the molecular mechanisms that may be involved in the activation of protein synthesis and subsequent restoration of muscle mass after a period of mechanical unloading. In addition, the efficiency of strategies proposed to improve muscle protein gain during recovery is also discussed.
Collapse
Affiliation(s)
- Timur M Mirzoev
- Myology Laboratory, Institute of Biomedical Problems RAS, Moscow 123007, Russia
| |
Collapse
|
21
|
Wilburn DT, Machek SB, Cardaci TD, Willoughby DS. Carbohydrate-Induced Insulin Signaling Activates Focal Adhesion Kinase: A Nutrient and Mechanotransduction Crossroads. Nutrients 2020; 12:nu12103145. [PMID: 33076263 PMCID: PMC7602406 DOI: 10.3390/nu12103145] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Revised: 10/03/2020] [Accepted: 10/13/2020] [Indexed: 12/17/2022] Open
Abstract
Research has suggested that nutrient, exercise, and metabolism-related proteins interact to regulate mammalian target of rapamycin complex one (mTOR) post-exercise and their interactions needs clarification. In a double-blind, cross-over, repeated measures design, ten participants completed four sets to failure at 70% of 1-repitition maximum (1-RM) with 45 s rest on angled leg press with or without pre-exercise maltodextrin (2 g/kg) after a 3 h fast. Vastus lateralis biopsies were collected at baseline before supplementation and 1 h post-exercise to analyze Focal Adhesion Kinase (FAK), ribosomal protein S6 kinase beta-1 (p70S6K), insulin receptor substrate 1 (IRS-1), phosphatidylinositol 3-kinase (PI3K), and 5' AMP-activated protein kinase (AMPK) activation. FAK and IRS-1 activity were only elevated 1 h post-exercise with carbohydrate ingestion (p < 0.05). PI3K and p70S6K activation were both elevated after exercise in both conditions (p < 0.05). However, AMPK activity did not change from baseline in both conditions (p > 0.05). We conclude that FAK does not induce mTOR activation through PI3K crosstalk in response to exercise alone. In addition, FAK may not be regulated by AMPK catalytic activity, but this needs further research. Interestingly, carbohydrate-induced insulin signaling appears to activate FAK at the level of IRS-1 but did not enhance mTOR activity 1 h post-exercise greater than the placebo condition. Future research should investigate these interactions under different conditions and within different time frames to clearly understand the interactions between these signaling molecules.
Collapse
Affiliation(s)
- Dylan T. Wilburn
- Exercise and Biochemical Nutrition Laboratory, Department of Health, Human Performance, and Recreation, Baylor University, Waco, TX 76706, USA; (D.T.W.); (S.B.M.); (T.D.C.)
| | - Steven B. Machek
- Exercise and Biochemical Nutrition Laboratory, Department of Health, Human Performance, and Recreation, Baylor University, Waco, TX 76706, USA; (D.T.W.); (S.B.M.); (T.D.C.)
| | - Thomas D. Cardaci
- Exercise and Biochemical Nutrition Laboratory, Department of Health, Human Performance, and Recreation, Baylor University, Waco, TX 76706, USA; (D.T.W.); (S.B.M.); (T.D.C.)
- Department of Exercise Science, Arnold School of Public Health, University of South Carolina, Columbia, SC 29208, USA
| | - Darryn S. Willoughby
- Exercise and Biochemical Nutrition Laboratory, Department of Health, Human Performance, and Recreation, Baylor University, Waco, TX 76706, USA; (D.T.W.); (S.B.M.); (T.D.C.)
- School of Exercise and Sport Science, University of Mary Hardin-Baylor, Belton, TX 76513, USA
- Correspondence:
| |
Collapse
|
22
|
Ma B, Zhang L, Li J, Xing T, Jiang Y, Gao F. Heat stress alters muscle protein and amino acid metabolism and accelerates liver gluconeogenesis for energy supply in broilers. Poult Sci 2020; 100:215-223. [PMID: 33357684 PMCID: PMC7772709 DOI: 10.1016/j.psj.2020.09.090] [Citation(s) in RCA: 72] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2020] [Revised: 09/05/2020] [Accepted: 09/15/2020] [Indexed: 12/30/2022] Open
Abstract
Heat stress impairs growth performance and alters body protein and amino acid metabolism. This study was investigated to explore how body protein and amino acid metabolism changed under heat stress (HS) and the stress adaptation mechanism. A total of 144 broilers (28 d old) were divided into 3 treatment groups for 1 wk: HS group (32°C), normal control group (22°C), and pair-feeding group (22°C). We found that HS elevated the feed-to-gain ratio, reduced the ADFI and ADG, decreased breast muscle mass and plasma levels of several amino acids (glycine, lysine, threonine, and tyrosine), and increased serum glutamic oxaloacetic transaminase (GOT) activity and corticosterone (CORT) level and liver GOT and glutamic pyruvic transaminase activities. Heat stress elevated muscle atrophy F-box mRNA expression and reduced mRNA expression of the 70-kD ribosomal protein S6 kinase in the breast muscle of broilers. Broilers in the HS group exhibited striking increases of mRNA expressions of solute carrier family 1 member 1, family 3 member 1, family 7 member 1, and family 7 member-like in the liver and liver gluconeogenesis genes (PCKc, PCKm, PC, and FBP1) in comparison with the other 2 groups. In conclusion, HS increased the circulating CORT level and subsequently caused muscle protein breakdown to provide amino acid substrates to liver gluconeogenesis responsible for energy supply.
Collapse
Affiliation(s)
- Bingbing Ma
- College of Animal Science and Technology, Key Laboratory of Animal Origin Food Production and Safety Guarantee of Jiangsu Province, Jiangsu Collaborative Innovation Center of Meat Production and Processing, Quality and Safety Control, Joint International Research Laboratory of Animal Health and Food Safety, National Experimental Teaching Demonstration Center of Animal Science, Nanjing Agricultural University, Nanjing 210095, PR China
| | - Lin Zhang
- College of Animal Science and Technology, Key Laboratory of Animal Origin Food Production and Safety Guarantee of Jiangsu Province, Jiangsu Collaborative Innovation Center of Meat Production and Processing, Quality and Safety Control, Joint International Research Laboratory of Animal Health and Food Safety, National Experimental Teaching Demonstration Center of Animal Science, Nanjing Agricultural University, Nanjing 210095, PR China
| | - Jiaolong Li
- College of Animal Science and Technology, Key Laboratory of Animal Origin Food Production and Safety Guarantee of Jiangsu Province, Jiangsu Collaborative Innovation Center of Meat Production and Processing, Quality and Safety Control, Joint International Research Laboratory of Animal Health and Food Safety, National Experimental Teaching Demonstration Center of Animal Science, Nanjing Agricultural University, Nanjing 210095, PR China
| | - Tong Xing
- College of Animal Science and Technology, Key Laboratory of Animal Origin Food Production and Safety Guarantee of Jiangsu Province, Jiangsu Collaborative Innovation Center of Meat Production and Processing, Quality and Safety Control, Joint International Research Laboratory of Animal Health and Food Safety, National Experimental Teaching Demonstration Center of Animal Science, Nanjing Agricultural University, Nanjing 210095, PR China
| | - Yun Jiang
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing 210023, PR China
| | - Feng Gao
- College of Animal Science and Technology, Key Laboratory of Animal Origin Food Production and Safety Guarantee of Jiangsu Province, Jiangsu Collaborative Innovation Center of Meat Production and Processing, Quality and Safety Control, Joint International Research Laboratory of Animal Health and Food Safety, National Experimental Teaching Demonstration Center of Animal Science, Nanjing Agricultural University, Nanjing 210095, PR China.
| |
Collapse
|
23
|
Howard EE, Pasiakos SM, Fussell MA, Rodriguez NR. Skeletal Muscle Disuse Atrophy and the Rehabilitative Role of Protein in Recovery from Musculoskeletal Injury. Adv Nutr 2020; 11:989-1001. [PMID: 32167129 PMCID: PMC7360452 DOI: 10.1093/advances/nmaa015] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Revised: 12/29/2019] [Accepted: 02/04/2020] [Indexed: 01/05/2023] Open
Abstract
Muscle atrophy and weakness occur as a consequence of disuse after musculoskeletal injury (MSI). The slow recovery and persistence of these deficits even after physical rehabilitation efforts indicate that interventions designed to attenuate muscle atrophy and protect muscle function are necessary to accelerate and optimize recovery from MSI. Evidence suggests that manipulating protein intake via dietary protein or free amino acid-based supplementation diminishes muscle atrophy and/or preserves muscle function in experimental models of disuse (i.e., immobilization and bed rest in healthy populations). However, this concept has rarely been considered in the context of disuse following MSI, which often occurs with some muscle activation during postinjury physical rehabilitation. Given that exercise sensitizes skeletal muscle to the anabolic effect of protein ingestion, early rehabilitation may act synergistically with dietary protein to protect muscle mass and function during postinjury disuse conditions. This narrative review explores mechanisms of skeletal muscle disuse atrophy and recent advances delineating the role of protein intake as a potential countermeasure. The possible synergistic effect of protein-based interventions and postinjury rehabilitation in attenuating muscle atrophy and weakness following MSI is also considered.
Collapse
Affiliation(s)
- Emily E Howard
- Department of Nutritional Sciences, University of Connecticut, Storrs, CT, USA,Military Nutrition Division, U.S. Army Research Institute of Environmental Medicine, Natick, MA, USA,Oak Ridge Institute for Science and Education, Oak Ridge, TN, USA
| | - Stefan M Pasiakos
- Military Nutrition Division, U.S. Army Research Institute of Environmental Medicine, Natick, MA, USA
| | - Maya A Fussell
- Department of Nutritional Sciences, University of Connecticut, Storrs, CT, USA
| | | |
Collapse
|
24
|
Jeong YJ, Hwang SK, Magae J, Chang YC. Ascofuranone suppresses invasion and F-actin cytoskeleton organization in cancer cells by inhibiting the mTOR complex 1 signaling pathway. Cell Oncol (Dordr) 2020; 43:793-805. [PMID: 32488849 DOI: 10.1007/s13402-020-00520-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Revised: 03/29/2020] [Accepted: 04/15/2020] [Indexed: 11/28/2022] Open
Abstract
PURPOSE Ascofuranone is an antiviral antibiotic that is known to exert multiple anti-tumor effects, including cell cycle arrest, inhibition of mitochondrial respiration, and inhibition of angiogenesis. In this study, we investigated the molecular mechanisms underlying the anti-metastatic effects of ascofuranone in insulin-like growth factor-I (IGF-1)-responsive cancer cells. METHODS The inhibitory effect of ascofuranone on cancer cell migration and invasion was assessed using scratch wound healing and Matrigel invasion assays, respectively. F-actin cytoskeleton organization was assessed using FITC conjugated phalloidin staining. Target gene expression was evaluated using Western blotting and gene silencing was performed using siRNA transfections. Finally, the anti-metastatic effect of ascofuranone was investigated in vivo. RESULTS We found that ascofuranone suppressed IGF-1-induced cell migration, invasion and motility in multiple cancer cell lines. The effects of ascofuranone on actin cytoskeleton organization were found to be mediated by suppression of the mTOR/p70S6K/4EBP1 pathway. Ascofuranone inhibited IGF-1-induced mTOR phosphorylation and actin cytoskeleton organization via upregulation of AMPK and downregulation of Akt phosphorylation. It also selectively suppressed the IGF-1-induced mTOR complex (mTORC)1 by phosphorylation of Raptor, but did not affect mTORC2. Furthermore, we found that focal adhesion kinase (FAK) activation decreased in response to ascofuranone, rapamycin, compound C and wortmannin treatment. Finally, we found that ascofuranone suppressed phosphorylation of FAK and mTOR and dephosphorylation of Raptor in cancerous metastatic lung tissues in vivo. CONCLUSIONS Our data indicate that ascofuranone suppresses IGF-1-induced cancer cell migration and invasion by blocking actin cytoskeleton organization and FAK activation through inhibition of the mTORC1 pathway, and reveal a novel anti-metastatic function of this compound.
Collapse
Affiliation(s)
- Yun-Jeong Jeong
- Research Institute of Biomedical Engineering, Department of Medicine, Catholic University of Daegu School of Medicine, 42472, Deagu, Korea
| | - Soon-Kyung Hwang
- Research Institute of Biomedical Engineering, Department of Medicine, Catholic University of Daegu School of Medicine, 42472, Deagu, Korea
| | - Junji Magae
- Magae Bioscience Institute, 49-4 Fujimidai, 300-1263, Tsukuba, Japan
| | - Young-Chae Chang
- Research Institute of Biomedical Engineering, Department of Medicine, Catholic University of Daegu School of Medicine, 42472, Deagu, Korea. .,Department of Cell Biology, Catholic University of Daegu School of Medicine, 3056-6, Daemyung-4-Dong, Nam-gu, 42472, Daegu, Korea.
| |
Collapse
|
25
|
Crossland H, Smith K, Atherton PJ, Wilkinson DJ. A novel stable isotope tracer method to simultaneously quantify skeletal muscle protein synthesis and breakdown. Metabol Open 2020; 5:100022. [PMID: 32494771 PMCID: PMC7259457 DOI: 10.1016/j.metop.2020.100022] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 12/11/2019] [Accepted: 01/01/2020] [Indexed: 01/08/2023] Open
Abstract
Background/aims Methodological challenges have been associated with the dynamic measurement of muscle protein breakdown (MPB), as have the measurement of both muscle protein synthesis (MPS) and MPB within the same experiment. Our aim was to use the transmethylation properties of methionine as proof-of-concept to measure rates of MPB via its methylation of histidine within skeletal muscle myofibrillar proteins, whilst simultaneously utilising methionine incorporation into bound protein to measure MPS. Results During the synthesis measurement period, incorporation of methyl[D3]-13C-methionine into cellular protein in C2C12 myotubes was observed (representative of MPS), alongside an increase in the appearance of methyl[D3]-methylhistidine into the media following methylation of histidine (representative of MPB). For further validation of this approach, fractional synthetic rates (FSR) of muscle protein were increased following treatment of the cells with the anabolic factors insulin-like growth factor-1 (IGF-1) and insulin, while dexamethasone expectedly reduced MPS. Conversely, rates of MPB were reduced with IGF-1 and insulin treatments, whereas dexamethasone accelerated MPB. Conclusions This is a novel stable isotope tracer approach that permits the dual assessment of muscle cellular protein synthesis and breakdown rates, through the provision of a single methionine amino acid tracer that could be utilised in a wide range of biological settings.
Collapse
Affiliation(s)
- Hannah Crossland
- MRC-ARUK Centre for Musculoskeletal Ageing Research, National Institute for Health Research (NIHR) Biomedical Research Centre (BRC), Clinical, Metabolic and Molecular Physiology, University of Nottingham, Royal Derby Hospital, Derby, UK
| | - Kenneth Smith
- MRC-ARUK Centre for Musculoskeletal Ageing Research, National Institute for Health Research (NIHR) Biomedical Research Centre (BRC), Clinical, Metabolic and Molecular Physiology, University of Nottingham, Royal Derby Hospital, Derby, UK
| | - Philip J Atherton
- MRC-ARUK Centre for Musculoskeletal Ageing Research, National Institute for Health Research (NIHR) Biomedical Research Centre (BRC), Clinical, Metabolic and Molecular Physiology, University of Nottingham, Royal Derby Hospital, Derby, UK
| | - Daniel J Wilkinson
- MRC-ARUK Centre for Musculoskeletal Ageing Research, National Institute for Health Research (NIHR) Biomedical Research Centre (BRC), Clinical, Metabolic and Molecular Physiology, University of Nottingham, Royal Derby Hospital, Derby, UK
| |
Collapse
|
26
|
Hadj-Moussa H, Zhang J, Pifferi F, Perret M, Storey KB. Profiling torpor-responsive microRNAs in muscles of the hibernating primate Microcebus murinus. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2020; 1863:194473. [DOI: 10.1016/j.bbagrm.2019.194473] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Revised: 12/09/2019] [Accepted: 12/09/2019] [Indexed: 12/25/2022]
|
27
|
Kraemer WJ, Ratamess NA, Hymer WC, Nindl BC, Fragala MS. Growth Hormone(s), Testosterone, Insulin-Like Growth Factors, and Cortisol: Roles and Integration for Cellular Development and Growth With Exercise. Front Endocrinol (Lausanne) 2020; 11:33. [PMID: 32158429 PMCID: PMC7052063 DOI: 10.3389/fendo.2020.00033] [Citation(s) in RCA: 144] [Impact Index Per Article: 28.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Accepted: 01/16/2020] [Indexed: 12/16/2022] Open
Abstract
Hormones are largely responsible for the integrated communication of several physiological systems responsible for modulating cellular growth and development. Although the specific hormonal influence must be considered within the context of the entire endocrine system and its relationship with other physiological systems, three key hormones are considered the "anabolic giants" in cellular growth and repair: testosterone, the growth hormone superfamily, and the insulin-like growth factor (IGF) superfamily. In addition to these anabolic hormones, glucocorticoids, mainly cortisol must also be considered because of their profound opposing influence on human skeletal muscle anabolism in many instances. This review presents emerging research on: (1) Testosterone signaling pathways, responses, and adaptations to resistance training; (2) Growth hormone: presents new complexity with exercise stress; (3) Current perspectives on IGF-I and physiological adaptations and complexity these hormones as related to training; and (4) Glucocorticoid roles in integrated communication for anabolic/catabolic signaling. Specifically, the review describes (1) Testosterone as the primary anabolic hormone, with an anabolic influence largely dictated primarily by genomic and possible non-genomic signaling, satellite cell activation, interaction with other anabolic signaling pathways, upregulation or downregulation of the androgen receptor, and potential roles in co-activators and transcriptional activity; (2) Differential influences of growth hormones depending on the "type" of the hormone being assayed and the magnitude of the physiological stress; (3) The exquisite regulation of IGF-1 by a family of binding proteins (IGFBPs 1-6), which can either stimulate or inhibit biological action depending on binding; and (4) Circadian patterning and newly discovered variants of glucocorticoid isoforms largely dictating glucocorticoid sensitivity and catabolic, muscle sparing, or pathological influence. The downstream integrated anabolic and catabolic mechanisms of these hormones not only affect the ability of skeletal muscle to generate force; they also have implications for pharmaceutical treatments, aging, and prevalent chronic conditions such as metabolic syndrome, insulin resistance, and hypertension. Thus, advances in our understanding of hormones that impact anabolic: catabolic processes have relevance for athletes and the general population, alike.
Collapse
Affiliation(s)
- William J. Kraemer
- Department of Human Sciences, The Ohio State University, Columbus, OH, United States
- *Correspondence: William J. Kraemer
| | - Nicholas A. Ratamess
- Department of Health and Exercise Science, The College of New Jersey, Ewing, NJ, United States
| | - Wesley C. Hymer
- Biochemistry and Molecular Biology, The Pennsylvania State University, University Park, PA, United States
| | - Bradley C. Nindl
- Department of Sports Medicine, School of Health and Rehabilitation Sciences, University of Pittsburgh, Pittsburgh, PA, United States
| | | |
Collapse
|
28
|
Godoy‐Parejo C, Deng C, Liu W, Chen G. Insulin Stimulates PI3K/AKT and Cell Adhesion to Promote the Survival of Individualized Human Embryonic Stem Cells. Stem Cells 2019; 37:1030-1041. [PMID: 31021484 PMCID: PMC6852186 DOI: 10.1002/stem.3026] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2018] [Revised: 03/30/2019] [Accepted: 04/15/2019] [Indexed: 12/14/2022]
Abstract
Insulin is present in most maintenance media for human embryonic stem cells (hESCs), but little is known about its essential role in the cell survival of individualized cells during passage. In this article, we show that insulin suppresses caspase cleavage and apoptosis after dissociation. Insulin activates insulin-like growth factor (IGF) receptor and PI3K/AKT cascade to promote cell survival and its function is independent of rho-associated protein kinase regulation. During niche reformation after passaging, insulin activates integrin that is essential for cell survival. IGF receptor colocalizes with focal adhesion complex and stimulates protein phosphorylation involved in focal adhesion formation. Insulin promotes cell spreading on matrigel-coated surfaces and suppresses myosin light chain phosphorylation. Further study showed that insulin is also required for the cell survival on E-cadherin coated surface and in suspension, indicating its essential role in cell-cell adhesion. This work highlights insulin's complex roles in signal transduction and niche re-establishment in hESCs. Stem Cells 2019;37:1030-1041.
Collapse
Affiliation(s)
- Carlos Godoy‐Parejo
- Centre of Reproduction, Development, and Aging, Faculty of Health SciencesUniversity of MacauMacau SARPeople's Republic of China
| | - Chunhao Deng
- Centre of Reproduction, Development, and Aging, Faculty of Health SciencesUniversity of MacauMacau SARPeople's Republic of China
| | - Weiwei Liu
- Centre of Reproduction, Development, and Aging, Faculty of Health SciencesUniversity of MacauMacau SARPeople's Republic of China
- Bioimaging and Stem Cell Core Facility, Faculty of Health SciencesUniversity of MacauMacau SARPeople's Republic of China
| | - Guokai Chen
- Centre of Reproduction, Development, and Aging, Faculty of Health SciencesUniversity of MacauMacau SARPeople's Republic of China
- Bioimaging and Stem Cell Core Facility, Faculty of Health SciencesUniversity of MacauMacau SARPeople's Republic of China
- Institute of Translational Medicine, Faculty of Health SciencesUniversity of MacauMacau SARPeople's Republic of China
| |
Collapse
|
29
|
Sun J, Zhou Y, Ye Z, Tan WS. Transforming growth factor-β1 stimulates mesenchymal stem cell proliferation by altering cell cycle through FAK-Akt-mTOR pathway. Connect Tissue Res 2019; 60:406-417. [PMID: 30642198 DOI: 10.1080/03008207.2019.1570171] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Background: Mesenchymal stem cells (MSCs) are promising for cell therapy and regenerative medicine. An increased need for expanding of MSCs under serum-free condition to achieve a sufficient quantity for therapeutic applications is inevitable. Transforming growth factor-β1 (TGF-β1) is widely used for expanding clinical-grade MSCs in vitro. This work focuses on the influence of TGF-β1 on proliferation in rat bone marrow-derived MSCs (BMSCs) and the underlying mechanism. Materials and Methods: BMSCs were isolated and cultured with or without TGF-β1 in a serum-free medium and Cell Counting Kit-8 assay was used to detect BMSCs proliferation. Cell cycle transition was also analyzed. Further, the expression levels of cyclin D1, phosphorylated focal adhesion kinase, and downstream effectors in Akt-mTOR-S6K1 signaling pathway were examined by western blotting. Results and Conclusion: TGF-β1 triggered proliferation via accelerating G1/S cell cycle transition in BMSCs. The addition of TGF-β1 can activate Akt-mTOR-S6K1 pathway. Additionally, FAK was found to be involved in the process. Upon adding the FAK inhibitor, both the activation of Akt-mTOR-S6K1 and TGF-β1-induced cell proliferation were abrogated. Together, an insight understanding of how TGF-β1 influences BMSCs proliferation is achieved. This study provides a possible strategy of supplementing TGF-β1 in serum-free medium for in vitro expansion, which eventually would advance the production of clinical-grade MSCs for regenerative medicine.
Collapse
Affiliation(s)
- Jie Sun
- a State Key Laboratory of Bioreactor Engineering , East China University of Science and Technology , Shanghai , P. R. China
| | - Yan Zhou
- a State Key Laboratory of Bioreactor Engineering , East China University of Science and Technology , Shanghai , P. R. China
| | - Zhaoyang Ye
- a State Key Laboratory of Bioreactor Engineering , East China University of Science and Technology , Shanghai , P. R. China
| | - Wen-Song Tan
- a State Key Laboratory of Bioreactor Engineering , East China University of Science and Technology , Shanghai , P. R. China
| |
Collapse
|
30
|
Gorgey AS, Witt O, O’Brien L, Cardozo C, Chen Q, Lesnefsky EJ, Graham ZA. Mitochondrial health and muscle plasticity after spinal cord injury. Eur J Appl Physiol 2018; 119:315-331. [DOI: 10.1007/s00421-018-4039-0] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2018] [Accepted: 11/22/2018] [Indexed: 01/15/2023]
|
31
|
Wackerhage H, Schoenfeld BJ, Hamilton DL, Lehti M, Hulmi JJ. Stimuli and sensors that initiate skeletal muscle hypertrophy following resistance exercise. J Appl Physiol (1985) 2018; 126:30-43. [PMID: 30335577 DOI: 10.1152/japplphysiol.00685.2018] [Citation(s) in RCA: 173] [Impact Index Per Article: 24.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
One of the most striking adaptations to exercise is the skeletal muscle hypertrophy that occurs in response to resistance exercise. A large body of work shows that a mammalian target of rapamycin complex 1 (mTORC1)-mediated increase of muscle protein synthesis is the key, but not sole, mechanism by which resistance exercise causes muscle hypertrophy. While much of the hypertrophy signaling cascade has been identified, the initiating, resistance exercise-induced and hypertrophy-stimulating stimuli have remained elusive. For the purpose of this review, we define an initiating, resistance exercise-induced and hypertrophy-stimulating signal as "hypertrophy stimulus," and the sensor of such a signal as "hypertrophy sensor." In this review we discuss our current knowledge of specific mechanical stimuli, damage/injury-associated and metabolic stress-associated triggers, as potential hypertrophy stimuli. Mechanical signals are the prime hypertrophy stimuli candidates, and a filamin-C-BAG3-dependent regulation of mTORC1, Hippo, and autophagy signaling is a plausible albeit still incompletely characterized hypertrophy sensor. Other candidate mechanosensing mechanisms are nuclear deformation-initiated signaling or several mechanisms related to costameres, which are the functional equivalents of focal adhesions in other cells. While exercise-induced muscle damage is probably not essential for hypertrophy, it is still unclear whether and how such muscle damage could augment a hypertrophic response. Interventions that combine blood flow restriction and especially low load resistance exercise suggest that resistance exercise-regulated metabolites could be hypertrophy stimuli, but this is based on indirect evidence and metabolite candidates are poorly characterized.
Collapse
Affiliation(s)
- Henning Wackerhage
- Department of Sport and Exercise Sciences, Technical University of Munich , Munich , Germany
| | | | - D Lee Hamilton
- Faculty of Health, School of Exercise and Nutrition Sciences, Deakin University , Victoria , Australia
| | - Maarit Lehti
- LIKES Research Centre for Physical Activity and Health , Jyväskylä , Finland
| | - Juha J Hulmi
- Neuromuscular Research Center, Biology of Physical Activity, Faculty of Sport and Health Sciences, University of Jyväskylä , Jyväskylä , Finland
| |
Collapse
|
32
|
Jackman SR, Brook MS, Pulsford RM, Cockcroft EJ, Campbell MI, Rankin D, Atherton P, Smith K, Bowtell JL. Tart cherry concentrate does not enhance muscle protein synthesis response to exercise and protein in healthy older men. Exp Gerontol 2018; 110:202-208. [PMID: 29890270 DOI: 10.1016/j.exger.2018.06.007] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Revised: 05/30/2018] [Accepted: 06/07/2018] [Indexed: 01/08/2023]
Abstract
BACKGROUND Oxidative stress and inflammation may contribute to anabolic resistance in response to protein and exercise in older adults. We investigated whether consumption of montmorency cherry concentrate (MCC) increased anabolic sensitivity to protein ingestion and resistance exercise in healthy older men. METHODS Sixteen healthy older men were randomized to receive MCC (60 mL·d-1) or placebo (PLA) for two weeks, after baseline measures in week 1. During week 3, participants consumed 10 g whey protein·d-1 and completed three bouts of unilateral leg resistance exercise (4 × 8-10 repetitions at 80% 1RM). Participants consumed a bolus (150 mL) and weekly (50 mL) doses of deuterated water. Body water 2H enrichment was measured in saliva and vastus lateralis biopsies were taken from the non-exercised leg after weeks 1, 2 and 3, and the exercised leg after week 3, to measure tracer incorporation at rest, in response to protein and protein + exercise. RESULTS Myofibrillar protein synthesis increased in response to exercise + protein compared to rest (p < 0.05) in both groups, but there was no added effect of supplement (MCC: 1.79 ± 0.75 EX vs 1.15 ± 0.40 rest; PLA: 2.22 ± 0.54 vs 1.21 ± 0.18; all %·d-1). Muscle total NFĸB protein was decreased with exercise and protein in MCC (NFĸB: -20.7 ± 17.5%) but increased in PLA (NFĸB: 17.8 ± 31.3%, p = 0.073). CONCLUSION Short-term MCC ingestion does not affect the anabolic response to protein and exercise in healthy, relatively active, older men, despite MCC ingestion attenuating expression of proteins involved in the muscle inflammatory response to exercise, which may influence the chronic training response.
Collapse
Affiliation(s)
- Sarah R Jackman
- Sport and Health Sciences, College of Life and Environmental Sciences, University of Exeter, Exeter, United Kingdom
| | - Matthew S Brook
- Medical Research Council-Arthritis Research United Kingdom Centre of Excellence for Musculoskeletal Ageing Research, University of Nottingham, Nottingham, United Kingdom
| | - Richard M Pulsford
- Sport and Health Sciences, College of Life and Environmental Sciences, University of Exeter, Exeter, United Kingdom
| | - Emma J Cockcroft
- Sport and Health Sciences, College of Life and Environmental Sciences, University of Exeter, Exeter, United Kingdom
| | - Matthew I Campbell
- Sport and Health Sciences, College of Life and Environmental Sciences, University of Exeter, Exeter, United Kingdom
| | - Debbie Rankin
- Medical Research Council-Arthritis Research United Kingdom Centre of Excellence for Musculoskeletal Ageing Research, University of Nottingham, Nottingham, United Kingdom
| | - Philip Atherton
- Medical Research Council-Arthritis Research United Kingdom Centre of Excellence for Musculoskeletal Ageing Research, University of Nottingham, Nottingham, United Kingdom
| | - Kenneth Smith
- Medical Research Council-Arthritis Research United Kingdom Centre of Excellence for Musculoskeletal Ageing Research, University of Nottingham, Nottingham, United Kingdom
| | - Joanna L Bowtell
- Sport and Health Sciences, College of Life and Environmental Sciences, University of Exeter, Exeter, United Kingdom.
| |
Collapse
|
33
|
Franchi MV, Ruoss S, Valdivieso P, Mitchell KW, Smith K, Atherton PJ, Narici MV, Flück M. Regional regulation of focal adhesion kinase after concentric and eccentric loading is related to remodelling of human skeletal muscle. Acta Physiol (Oxf) 2018; 223:e13056. [PMID: 29438584 DOI: 10.1111/apha.13056] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Revised: 01/31/2018] [Accepted: 02/07/2018] [Indexed: 12/31/2022]
Abstract
AIMS We assessed focal adhesion kinase (FAK) response to concentric (CON) vs eccentric (ECC) resistance training (RT) at two vastus lateralis (VL) sites, and the relationships between FAK, muscle protein synthesis (MPS) and morphological remodelling. METHODS Six young males trained both legs unilaterally 3 times/week for 8 weeks; one leg performed CON RT, the contralateral performed ECC RT. Muscle biopsies were collected after training from VL mid-belly (MID) and distal (distal) sites at 0, 4, 8 weeks. Focal adhesion kinase content and activation were evaluated by immunoblotting. MPS was assessed by deuterium oxide tracer; morphological adaptations were evaluated by ultrasound and DXA. RESULTS pY397-FAK 8 weeks levels were ~4-fold greater after ECC at the distal site compared to CON (P < .05); pY397FAK to total FAK ratio was greater in ECC vs CON at 4 (~2.2-fold, P < .05) and 8 weeks (~9-fold, P < .001) at the distal site. Meta-vinculin was found transiently increased at 4 weeks at the distal site only after ECC RT. ECC presented greater fascicle length (Lf) increases (10.5% vs 4%), whereas CON showed greater in pennation angle (PA) changes (12.3% vs 2.1%). MPS did not differ between exercise types or muscle sites at all time points. distal pY397-FAK and pY397-FAK/FAK values correlated to changes in Lf at 8 weeks (r = .76, P < .01 and r = .66, P < .05 respectively). CONCLUSION Focal adhesion kinase phosphorylation was greater at 8 weeks after ECC RT and was muscle region-specific. FAK activity correlated to contraction-dependent architectural remodelling, suggesting a potential role of FAK in orienting muscle structural changes in response to distinct mechanical stimuli.
Collapse
Affiliation(s)
- M V Franchi
- Laboratory for Muscle Plasticity, Balgrist University Hospital, University of Zurich, Zurich, Switzerland
- MRC-ARUK Centre for Musculoskeletal Ageing, Royal Derby Hospital, University of Nottingham, Derby, UK
| | - S Ruoss
- Laboratory for Muscle Plasticity, Balgrist University Hospital, University of Zurich, Zurich, Switzerland
| | - P Valdivieso
- Laboratory for Muscle Plasticity, Balgrist University Hospital, University of Zurich, Zurich, Switzerland
| | - K W Mitchell
- MRC-ARUK Centre for Musculoskeletal Ageing, Royal Derby Hospital, University of Nottingham, Derby, UK
| | - K Smith
- MRC-ARUK Centre for Musculoskeletal Ageing, Royal Derby Hospital, University of Nottingham, Derby, UK
| | - P J Atherton
- MRC-ARUK Centre for Musculoskeletal Ageing, Royal Derby Hospital, University of Nottingham, Derby, UK
| | - M V Narici
- MRC-ARUK Centre for Musculoskeletal Ageing, Royal Derby Hospital, University of Nottingham, Derby, UK
- Department of Biomedical Sciences, Institute of Physiology, University of Padua, Padua, Italy
| | - M Flück
- Laboratory for Muscle Plasticity, Balgrist University Hospital, University of Zurich, Zurich, Switzerland
| |
Collapse
|
34
|
Lassiter DG, Nylén C, Sjögren RJO, Chibalin AV, Wallberg-Henriksson H, Näslund E, Krook A, Zierath JR. FAK tyrosine phosphorylation is regulated by AMPK and controls metabolism in human skeletal muscle. Diabetologia 2018; 61:424-432. [PMID: 29022062 PMCID: PMC6449061 DOI: 10.1007/s00125-017-4451-8] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Accepted: 08/11/2017] [Indexed: 01/28/2023]
Abstract
AIMS/HYPOTHESIS Insulin-mediated signals and AMP-activated protein kinase (AMPK)-mediated signals are activated in response to physiological conditions that represent energy abundance and shortage, respectively. Focal adhesion kinase (FAK) is implicated in insulin signalling and cancer progression in various non-muscle cell types and plays a regulatory role during skeletal muscle differentiation. The role of FAK in skeletal muscle in relation to insulin stimulation or AMPK activation is unknown. We examined the effects of insulin or AMPK activation on FAK phosphorylation in human skeletal muscle and the direct role of FAK on glucose and lipid metabolism. We hypothesised that insulin treatment and AMPK activation would have opposing effects on FAK phosphorylation and that gene silencing of FAK would alter metabolism. METHODS Human muscle was treated with insulin or the AMPK-activating compound 5-aminoimadazole-4-carboxamide ribonucleotide (AICAR) to determine FAK phosphorylation and glucose transport. Primary human skeletal muscle cells were used to study the effects of insulin or AICAR treatment on FAK signalling during serum starvation, as well as to determine the metabolic consequences of silencing the FAK gene, PTK2. RESULTS AMPK activation reduced tyrosine phosphorylation of FAK in skeletal muscle. AICAR reduced p-FAKY397 in isolated human skeletal muscle and cultured myotubes. Insulin stimulation did not alter FAK phosphorylation. Serum starvation increased AMPK activation, as demonstrated by increased p-ACCS222, concomitant with reduced p-FAKY397. FAK signalling was reduced owing to serum starvation and AICAR treatment as demonstrated by reduced p-paxillinY118. Silencing PTK2 in primary human skeletal muscle cells increased palmitate oxidation and reduced glycogen synthesis. CONCLUSIONS/INTERPRETATION AMPK regulates FAK signalling in skeletal muscle. Moreover, siRNA-mediated FAK knockdown enhances lipid oxidation while impairing glycogen synthesis in skeletal muscle. Further exploration of the interaction between AMPK and FAK may lead to novel therapeutic strategies for diabetes and other chronic conditions associated with an altered metabolic homeostasis.
Collapse
Affiliation(s)
- David G Lassiter
- Department of Molecular Medicine and Surgery, Section for Integrative Physiology, Karolinska Institutet, von Eulers väg 4a, IV, SE-171 65, Stockholm, Sweden
| | - Carolina Nylén
- Department of Molecular Medicine and Surgery, Section for Integrative Physiology, Karolinska Institutet, von Eulers väg 4a, IV, SE-171 65, Stockholm, Sweden
| | - Rasmus J O Sjögren
- Department of Molecular Medicine and Surgery, Section for Integrative Physiology, Karolinska Institutet, von Eulers väg 4a, IV, SE-171 65, Stockholm, Sweden
| | - Alexander V Chibalin
- Department of Molecular Medicine and Surgery, Section for Integrative Physiology, Karolinska Institutet, von Eulers väg 4a, IV, SE-171 65, Stockholm, Sweden
| | | | - Erik Näslund
- Division of Surgery, Department of Clinical Sciences, Danderyd Hospital, Karolinska Institutet, Stockholm, Sweden
| | - Anna Krook
- Department of Physiology and Pharmacology, Integrative Physiology, Karolinska Institutet, Stockholm, Sweden
| | - Juleen R Zierath
- Department of Molecular Medicine and Surgery, Section for Integrative Physiology, Karolinska Institutet, von Eulers väg 4a, IV, SE-171 65, Stockholm, Sweden.
- Department of Physiology and Pharmacology, Integrative Physiology, Karolinska Institutet, Stockholm, Sweden.
- Section of Integrative Physiology, The Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Science, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
35
|
Miller BF, Hamilton KL, Majeed ZR, Abshire SM, Confides AL, Hayek AM, Hunt ER, Shipman P, Peelor FF, Butterfield TA, Dupont‐Versteegden EE. Enhanced skeletal muscle regrowth and remodelling in massaged and contralateral non-massaged hindlimb. J Physiol 2018; 596:83-103. [PMID: 29090454 PMCID: PMC5746529 DOI: 10.1113/jp275089] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Accepted: 10/16/2017] [Indexed: 12/21/2022] Open
Abstract
KEY POINTS Muscle fibre cross sectional area is enhanced with massage in the form of cyclic compressive loading during regrowth after atrophy. Massage enhances protein synthesis of the myofibrillar and cytosolic, but not the mitochondrial fraction, in muscle during regrowth. Focal adhesion kinase activation and satellite cell number are elevated in muscles undergoing massage during regrowth. Muscle fibre cross sectional area and protein synthesis of the myofibrillar fraction, but not DNA synthesis, are elevated in muscle of the contralateral non-massaged limb. Massage in the form of cyclic compressive loading is a potential anabolic intervention during muscle regrowth after atrophy. ABSTRACT Massage, in the form of cyclic compressive loading (CCL), is associated with multiple health benefits, but its potential anabolic effect on atrophied muscle has not been investigated. We hypothesized that the mechanical activity associated with CCL induces an anabolic effect in skeletal muscle undergoing regrowth after a period of atrophy. Fischer-Brown Norway rats at 10 months of age were hindlimb unloaded for a period of 2 weeks. The rats were then allowed reambulation with CCL applied at a 4.5 N load at 0.5 Hz frequency for 30 min every other day for four bouts during a regrowth period of 8 days. Muscle fibre cross sectional area was enhanced by 18% with massage during regrowth compared to reloading alone, and this was accompanied by elevated myofibrillar and cytosolic protein as well as DNA synthesis. Focal adhesion kinase phosphorylation indicated that CCL increased mechanical stimulation, while a higher number of Pax7+ cells likely explains the elevated DNA synthesis. Surprisingly, the contralateral non-massaged limb exhibited a comparable 17% higher muscle fibre size compared to reloading alone, and myofibrillar protein synthesis, but not DNA synthesis, was also elevated. We conclude that massage in the form of CCL induces an anabolic response in muscles regrowing after an atrophy-inducing event. We suggest that massage can be used as an intervention to aid in the regrowth of muscle lost during immobilization.
Collapse
Affiliation(s)
- Benjamin F. Miller
- Health and Exercise ScienceColorado State UniversityFort CollinsCO80523‐1582USA
| | - Karyn L. Hamilton
- Health and Exercise ScienceColorado State UniversityFort CollinsCO80523‐1582USA
| | - Zana R. Majeed
- Department of Rehabilitation Sciences, College of Health SciencesUniversity of KentuckyLexingtonKY40536‐0200USA
| | - Sarah M. Abshire
- Department of Rehabilitation Sciences, College of Health SciencesUniversity of KentuckyLexingtonKY40536‐0200USA
- Center for Muscle BiologyUniversity of KentuckyLexingtonKY40536‐0200USA
| | - Amy L. Confides
- Department of Rehabilitation Sciences, College of Health SciencesUniversity of KentuckyLexingtonKY40536‐0200USA
- Center for Muscle BiologyUniversity of KentuckyLexingtonKY40536‐0200USA
| | - Amanda M. Hayek
- Department of Rehabilitation Sciences, College of Health SciencesUniversity of KentuckyLexingtonKY40536‐0200USA
| | - Emily R. Hunt
- Department of Rehabilitation Sciences, College of Health SciencesUniversity of KentuckyLexingtonKY40536‐0200USA
| | - Patrick Shipman
- Department of MathematicsColorado State UniversityFort CollinsCO80523‐1582USA
| | - Frederick F. Peelor
- Health and Exercise ScienceColorado State UniversityFort CollinsCO80523‐1582USA
| | - Timothy A. Butterfield
- Department of Rehabilitation Sciences, College of Health SciencesUniversity of KentuckyLexingtonKY40536‐0200USA
- Center for Muscle BiologyUniversity of KentuckyLexingtonKY40536‐0200USA
| | - Esther E. Dupont‐Versteegden
- Department of Rehabilitation Sciences, College of Health SciencesUniversity of KentuckyLexingtonKY40536‐0200USA
- Center for Muscle BiologyUniversity of KentuckyLexingtonKY40536‐0200USA
| |
Collapse
|
36
|
Crossland H, Timmons JA, Atherton PJ. A dynamic ribosomal biogenesis response is not required for IGF-1-mediated hypertrophy of human primary myotubes. FASEB J 2017; 31:5196-5207. [PMID: 28774889 PMCID: PMC5690393 DOI: 10.1096/fj.201700329r] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2017] [Accepted: 07/17/2017] [Indexed: 12/11/2022]
Abstract
Increased ribosomal DNA transcription has been proposed to limit muscle protein synthesis, making ribosome biogenesis central to skeletal muscle hypertrophy. We examined the relationship between ribosomal RNA (rRNA) production and IGF-1-mediated myotube hypertrophy in vitro Primary skeletal myotubes were treated with IGF-1 (50 ng/ml) with or without 0.5 µM CX-5461 (CX), an inhibitor of RNA polymerase I. Myotube diameter, total protein, and RNA and DNA levels were measured along with markers of RNA polymerase I regulatory factors and regulators of protein synthesis. CX treatment reduced 45S pre-rRNA expression (-64 ± 5% vs. IGF-1; P < 0.001) and total RNA content (-16 ± 2% vs. IGF-1; P < 0.001) in IGF-1-treated myotubes. IGF-1-mediated increases in myotube diameter (1.27 ± 0.09-fold, P < 0.05 vs. control) and total protein (+20 ± 2%; P < 0.001 vs. control) were not prevented by CX treatment. Suppression of rRNA synthesis during IGF-1 treatment did not prevent early increases in AKT (+203 ± 39% vs. CX; P < 0.001) and p70 S6K1 (269 ± 41% vs. CX; P < 0.001) phosphorylation. Despite robust inhibition of the dynamic ribosomal biogenesis response to IGF-1, myotube diameter and protein accretion were sustained. Thus, while ribosome biogenesis represents a potential site for the regulation of skeletal muscle protein synthesis and muscle mass, it does not appear to be a prerequisite for IGF-1-induced myotube hypertrophy in vitro.-Crossland, H., Timmons, J. A., Atherton, P. J. A dynamic ribosomal biogenesis response is not required for IGF-1-mediated hypertrophy of human primary myotubes.
Collapse
Affiliation(s)
- Hannah Crossland
- Division of Genetics and Molecular Medicine, Guy's Hospital, King's College London, London, United Kingdom; and
- School of Medicine, Royal Derby Hospital, University of Nottingham, Derby, United Kingdom
| | - James A Timmons
- Division of Genetics and Molecular Medicine, Guy's Hospital, King's College London, London, United Kingdom; and
| | - Philip J Atherton
- School of Medicine, Royal Derby Hospital, University of Nottingham, Derby, United Kingdom
| |
Collapse
|
37
|
Brook MS, Wilkinson DJ, Mitchell WK, Lund JL, Phillips BE, Szewczyk NJ, Kainulainen H, Lensu S, Koch LG, Britton SL, Greenhaff PL, Smith K, Atherton PJ. A novel D 2O tracer method to quantify RNA turnover as a biomarker of de novo ribosomal biogenesis, in vitro, in animal models, and in human skeletal muscle. Am J Physiol Endocrinol Metab 2017; 313:E681-E689. [PMID: 28811296 PMCID: PMC5814597 DOI: 10.1152/ajpendo.00157.2017] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2017] [Revised: 08/10/2017] [Accepted: 08/14/2017] [Indexed: 11/22/2022]
Abstract
Current methods to quantify in vivo RNA dynamics are limited. Here, we developed a novel stable isotope (D2O) methodology to quantify RNA synthesis (i.e., ribosomal biogenesis) in cells, animal models, and humans. First, proliferating C2C12 cells were incubated in D2O-enriched media and myotubes ±50 ng/ml IGF-I. Second, rat quadriceps (untrained, n = 9; 7-wk interval-"like" training, n = 13) were collected after ~3-wk D2O (70 atom %) administration, with body-water enrichment monitored via blood sampling. Finally, 10 (23 ± 1 yr) men consumed 150-ml D2O followed by 50 ml/wk and undertook 6-wk resistance exercise (6 × 8 repetitions, 75% 1-repetition maximum 3/wk) with body-water enrichment monitored by saliva sampling and muscle biopsies (for determination of RNA synthesis) at 0, 3, and 6 wk. Ribose mole percent excess (r-MPE) from purine nucleotides was analyzed via GC-MS/MS. Proliferating C2C12 cell r-MPE exhibited a rise to plateau, whereas IGF-I increased myotube RNA from 76 ± 3 to 123 ± 3 ng/μl and r-MPE by 0.39 ± 0.1% (both P < 0.01). After 3 wk, rat quadriceps r-MPE had increased to 0.25 ± 0.01% (P < 0.01) and was greater with running exercise (0.36 ± 0.02%; P < 0.01). Human muscle r-MPE increased to 0.06 ± 0.01 and 0.13 ± 0.02% at 3/6 wk, respectively, equating to synthesis rates of ~0.8%/day, increasing with resistance exercise to 1.7 ± 0.3%/day (P < 0.01) and 1.2 ± 0.1%/day (P < 0.05) at 3/6 wk, respectively. Therefore, we have developed and physiologically validated a novel technique to explore ribosomal biogenesis in a multimodal fashion.
Collapse
Affiliation(s)
- M S Brook
- Medical Research Council-Arthritis Research UK Centre for Musculoskeletal Ageing Research, Clinical, Metabolic and Molecular Physiology, University of Nottingham, Royal Derby Hospital Centre, Derby, United Kingdom
| | - D J Wilkinson
- Medical Research Council-Arthritis Research UK Centre for Musculoskeletal Ageing Research, Clinical, Metabolic and Molecular Physiology, University of Nottingham, Royal Derby Hospital Centre, Derby, United Kingdom
| | - W K Mitchell
- Medical Research Council-Arthritis Research UK Centre for Musculoskeletal Ageing Research, Clinical, Metabolic and Molecular Physiology, University of Nottingham, Royal Derby Hospital Centre, Derby, United Kingdom
| | - J L Lund
- Medical Research Council-Arthritis Research UK Centre for Musculoskeletal Ageing Research, Clinical, Metabolic and Molecular Physiology, University of Nottingham, Royal Derby Hospital Centre, Derby, United Kingdom
| | - B E Phillips
- Medical Research Council-Arthritis Research UK Centre for Musculoskeletal Ageing Research, Clinical, Metabolic and Molecular Physiology, University of Nottingham, Royal Derby Hospital Centre, Derby, United Kingdom
| | - N J Szewczyk
- Medical Research Council-Arthritis Research UK Centre for Musculoskeletal Ageing Research, Clinical, Metabolic and Molecular Physiology, University of Nottingham, Royal Derby Hospital Centre, Derby, United Kingdom
| | - H Kainulainen
- Department of Biology of Physical Activity, University of Jyväskylä, Jyväskylä, Finland; and
| | - S Lensu
- Department of Biology of Physical Activity, University of Jyväskylä, Jyväskylä, Finland; and
| | - L G Koch
- Department of Anesthesiology, University of Michigan, Ann Arbor, Michigan
| | - S L Britton
- Department of Anesthesiology, University of Michigan, Ann Arbor, Michigan
| | - P L Greenhaff
- Medical Research Council-Arthritis Research UK Centre for Musculoskeletal Ageing Research, Clinical, Metabolic and Molecular Physiology, University of Nottingham, Royal Derby Hospital Centre, Derby, United Kingdom
| | - K Smith
- Medical Research Council-Arthritis Research UK Centre for Musculoskeletal Ageing Research, Clinical, Metabolic and Molecular Physiology, University of Nottingham, Royal Derby Hospital Centre, Derby, United Kingdom
| | - P J Atherton
- Medical Research Council-Arthritis Research UK Centre for Musculoskeletal Ageing Research, Clinical, Metabolic and Molecular Physiology, University of Nottingham, Royal Derby Hospital Centre, Derby, United Kingdom;
| |
Collapse
|
38
|
Deane CS, Wilkinson DJ, Phillips BE, Smith K, Etheridge T, Atherton PJ. "Nutraceuticals" in relation to human skeletal muscle and exercise. Am J Physiol Endocrinol Metab 2017; 312:E282-E299. [PMID: 28143855 PMCID: PMC5406990 DOI: 10.1152/ajpendo.00230.2016] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Revised: 01/25/2017] [Accepted: 01/25/2017] [Indexed: 12/16/2022]
Abstract
Skeletal muscles have a fundamental role in locomotion and whole body metabolism, with muscle mass and quality being linked to improved health and even lifespan. Optimizing nutrition in combination with exercise is considered an established, effective ergogenic practice for athletic performance. Importantly, exercise and nutritional approaches also remain arguably the most effective countermeasure for muscle dysfunction associated with aging and numerous clinical conditions, e.g., cancer cachexia, COPD, and organ failure, via engendering favorable adaptations such as increased muscle mass and oxidative capacity. Therefore, it is important to consider the effects of established and novel effectors of muscle mass, function, and metabolism in relation to nutrition and exercise. To address this gap, in this review, we detail existing evidence surrounding the efficacy of a nonexhaustive list of macronutrient, micronutrient, and "nutraceutical" compounds alone and in combination with exercise in relation to skeletal muscle mass, metabolism (protein and fuel), and exercise performance (i.e., strength and endurance capacity). It has long been established that macronutrients have specific roles and impact upon protein metabolism and exercise performance, (i.e., protein positively influences muscle mass and protein metabolism), whereas carbohydrate and fat intakes can influence fuel metabolism and exercise performance. Regarding novel nutraceuticals, we show that the following ones in particular may have effects in relation to 1) muscle mass/protein metabolism: leucine, hydroxyl β-methylbutyrate, creatine, vitamin-D, ursolic acid, and phosphatidic acid; and 2) exercise performance: (i.e., strength or endurance capacity): hydroxyl β-methylbutyrate, carnitine, creatine, nitrates, and β-alanine.
Collapse
Affiliation(s)
- Colleen S Deane
- Medical Research Council-Arthritis Research UK Centre of Excellence for Musculoskeletal Ageing Research and Clinical, Metabolic, and Molecular Physiology, University of Nottingham, Royal Derby Hospital, Derby, United Kingdom
- Faculty of Health and Social Science, Bournemouth University, Bournemouth, United Kingdom; and
- Department of Sport and Health Science, College of Life and Environmental Sciences, University of Exeter, Exeter, United Kingdom
| | - Daniel J Wilkinson
- Medical Research Council-Arthritis Research UK Centre of Excellence for Musculoskeletal Ageing Research and Clinical, Metabolic, and Molecular Physiology, University of Nottingham, Royal Derby Hospital, Derby, United Kingdom
| | - Bethan E Phillips
- Medical Research Council-Arthritis Research UK Centre of Excellence for Musculoskeletal Ageing Research and Clinical, Metabolic, and Molecular Physiology, University of Nottingham, Royal Derby Hospital, Derby, United Kingdom
| | - Kenneth Smith
- Medical Research Council-Arthritis Research UK Centre of Excellence for Musculoskeletal Ageing Research and Clinical, Metabolic, and Molecular Physiology, University of Nottingham, Royal Derby Hospital, Derby, United Kingdom
| | - Timothy Etheridge
- Department of Sport and Health Science, College of Life and Environmental Sciences, University of Exeter, Exeter, United Kingdom
| | - Philip J Atherton
- Medical Research Council-Arthritis Research UK Centre of Excellence for Musculoskeletal Ageing Research and Clinical, Metabolic, and Molecular Physiology, University of Nottingham, Royal Derby Hospital, Derby, United Kingdom;
| |
Collapse
|
39
|
Kasper AM, Turner DC, Martin NRW, Sharples AP. Mimicking exercise in three-dimensional bioengineered skeletal muscle to investigate cellular and molecular mechanisms of physiological adaptation. J Cell Physiol 2017; 233:1985-1998. [DOI: 10.1002/jcp.25840] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Accepted: 02/02/2017] [Indexed: 12/22/2022]
Affiliation(s)
- Andreas M. Kasper
- Stem Cells, Ageing, and Molecular Physiology (SCAMP) Unit, Exercise Metabolism and Adaptation Research group, Research Institute for Sport and Exercise Sciences (RISES), School of Sport and Exercise Sciences; Liverpool John Moores University; Liverpool UK
| | - Daniel C. Turner
- Stem Cells, Ageing, and Molecular Physiology (SCAMP) Unit, Exercise Metabolism and Adaptation Research group, Research Institute for Sport and Exercise Sciences (RISES), School of Sport and Exercise Sciences; Liverpool John Moores University; Liverpool UK
| | - Neil R. W. Martin
- Musculoskeletal Biology Research Group, School of Sport, Exercise, and Health Sciences; Loughborough University; Loughborough UK
| | - Adam P. Sharples
- Stem Cells, Ageing, and Molecular Physiology (SCAMP) Unit, Exercise Metabolism and Adaptation Research group, Research Institute for Sport and Exercise Sciences (RISES), School of Sport and Exercise Sciences; Liverpool John Moores University; Liverpool UK
| |
Collapse
|
40
|
Bass JJ, Wilkinson DJ, Rankin D, Phillips BE, Szewczyk NJ, Smith K, Atherton PJ. An overview of technical considerations for Western blotting applications to physiological research. Scand J Med Sci Sports 2017; 27:4-25. [PMID: 27263489 PMCID: PMC5138151 DOI: 10.1111/sms.12702] [Citation(s) in RCA: 240] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/25/2016] [Indexed: 12/11/2022]
Abstract
The applications of Western/immunoblotting (WB) techniques have reached multiple layers of the scientific community and are now considered routine procedures in the field of physiology. This is none more so than in relation to skeletal muscle physiology (i.e., resolving the mechanisms underpinning adaptations to exercise). Indeed, the inclusion of WB data is now considered an essential aspect of many such physiological publications to provide mechanistic insight into regulatory processes. Despite this popularity, and due to the ubiquitous and relatively inexpensive availability of WB equipment, the quality of WB in publications and subsequent analysis and interpretation of the data can be variable, perhaps resulting in spurious conclusions. This may be due to poor laboratory technique and/or lack of comprehension of the critical steps involved in WB and what quality control procedures should be in place to ensure robust data generation. The present review aims to provide a detailed description and critique of WB procedures and technicalities, from sample collection through preparation, blotting and detection, to analysis of the data collected. We aim to provide the reader with improved expertise to critically conduct, evaluate, and troubleshoot the WB process, to produce reproducible and reliable blots.
Collapse
Affiliation(s)
- J J Bass
- MRC/ARUK Centre of Excellence for Musculoskeletal Ageing Research, School of Medicine, University of Nottingham, Derby, UK
| | - D J Wilkinson
- MRC/ARUK Centre of Excellence for Musculoskeletal Ageing Research, School of Medicine, University of Nottingham, Derby, UK
| | - D Rankin
- MRC/ARUK Centre of Excellence for Musculoskeletal Ageing Research, School of Medicine, University of Nottingham, Derby, UK
| | - B E Phillips
- MRC/ARUK Centre of Excellence for Musculoskeletal Ageing Research, School of Medicine, University of Nottingham, Derby, UK
| | - N J Szewczyk
- MRC/ARUK Centre of Excellence for Musculoskeletal Ageing Research, School of Medicine, University of Nottingham, Derby, UK
| | - K Smith
- MRC/ARUK Centre of Excellence for Musculoskeletal Ageing Research, School of Medicine, University of Nottingham, Derby, UK
| | - P J Atherton
- MRC/ARUK Centre of Excellence for Musculoskeletal Ageing Research, School of Medicine, University of Nottingham, Derby, UK
| |
Collapse
|
41
|
Brook MS, Wilkinson DJ, Mitchell WK, Lund JN, Phillips BE, Szewczyk NJ, Greenhaff PL, Smith K, Atherton PJ. Synchronous deficits in cumulative muscle protein synthesis and ribosomal biogenesis underlie age-related anabolic resistance to exercise in humans. J Physiol 2016; 594:7399-7417. [PMID: 27654940 PMCID: PMC5157077 DOI: 10.1113/jp272857] [Citation(s) in RCA: 141] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2016] [Accepted: 09/19/2016] [Indexed: 12/25/2022] Open
Abstract
KEY POINTS Resistance exercise training (RET) is one of the most effective strategies for preventing declines in skeletal muscle mass and strength with age. Hypertrophic responses to RET with age are diminished compared to younger individuals. In response to 6 weeks RET, we found blunted hypertrophic responses with age are underpinned by chronic deficits in long-term muscle protein synthesis. We show this is likely to be the result of multifactorial deficits in anabolic hormones and blunted translational efficiency and capacity. These results provide great insight into age-related exercise adaptations and provide a platform on which to devise appropriate nutritional and exercise interventions on a longer term basis. ABSTRACT Ageing is associated with impaired hypertrophic responses to resistance exercise training (RET). Here we investigated the aetiology of 'anabolic resistance' in older humans. Twenty healthy male individuals, 10 younger (Y; 23 ± 1 years) and 10 older (O; 69 ± 3 years), performed 6 weeks unilateral RET (6 × 8 repetitions, 75% of one repetition maximum (1-RM), 3 times per week). After baseline bilateral vastus lateralis (VL) muscle biopsies, subjects consumed 150 ml D2 O (70 atom%; thereafter 50 ml week-1 ), further bilateral VL muscle biopsies were taken at 3 and 6 weeks to quantify muscle protein synthesis (MPS) via gas chromatography-pyrolysis-isotope ratio mass spectrometry. After RET, 1-RM increased in Y (+35 ± 4%) and O (+25 ± 3%; P < 0.01), while MVC increased in Y (+21 ± 5%; P < 0.01) but not O (+6 ± 3%; not significant (NS)). In comparison to Y, O displayed blunted RET-induced increases in muscle thickness (at 3 and 6 weeks, respectively, Y: +8 ± 1% and +11 ± 2%, P < 0.01; O: +2.6 ± 1% and +3.5 ± 2%, NS). While 'basal' longer term MPS was identical between Y and O (∼1.35 ± 0.1% day-1 ), MPS increased in response to RET only in Y (3 weeks, Y: 1.61 ± 0.1% day-1 ; O: 1.49 ± 0.1% day-1 ). Consistent with this, O exhibited inferior ribosomal biogenesis (RNA:DNA ratio and c-MYC induction: Y: +4 ± 2 fold change; O: +1.9 ± 1 fold change), translational efficiency (S6K1 phosphorylation, Y: +10 ± 4 fold change; O: +4 ± 2 fold change) and anabolic hormone milieu (testosterone, Y: 367 ± 19; O: 274 ± 19 ng dl-1 (all P < 0.05). Anabolic resistance is thus multifactorial.
Collapse
Affiliation(s)
- Matthew S. Brook
- MRC‐ARUK Centre of Excellence for Musculoskeletal Ageing ResearchClinical, Metabolic and Molecular PhysiologyUniversity of NottinghamDerbyUK
| | - Daniel J. Wilkinson
- MRC‐ARUK Centre of Excellence for Musculoskeletal Ageing ResearchClinical, Metabolic and Molecular PhysiologyUniversity of NottinghamDerbyUK
| | - William K. Mitchell
- MRC‐ARUK Centre of Excellence for Musculoskeletal Ageing ResearchClinical, Metabolic and Molecular PhysiologyUniversity of NottinghamDerbyUK
- Departments of SurgeryRoyal Derby HospitalDerbyUK
| | - Jonathan N. Lund
- MRC‐ARUK Centre of Excellence for Musculoskeletal Ageing ResearchClinical, Metabolic and Molecular PhysiologyUniversity of NottinghamDerbyUK
- Departments of SurgeryRoyal Derby HospitalDerbyUK
| | - Bethan E. Phillips
- MRC‐ARUK Centre of Excellence for Musculoskeletal Ageing ResearchClinical, Metabolic and Molecular PhysiologyUniversity of NottinghamDerbyUK
| | - Nathaniel J. Szewczyk
- MRC‐ARUK Centre of Excellence for Musculoskeletal Ageing ResearchClinical, Metabolic and Molecular PhysiologyUniversity of NottinghamDerbyUK
| | - Paul L. Greenhaff
- MRC‐ARUK Centre of Excellence for Musculoskeletal Ageing ResearchClinical, Metabolic and Molecular PhysiologyUniversity of NottinghamDerbyUK
| | - Kenneth Smith
- MRC‐ARUK Centre of Excellence for Musculoskeletal Ageing ResearchClinical, Metabolic and Molecular PhysiologyUniversity of NottinghamDerbyUK
| | - Philip J. Atherton
- MRC‐ARUK Centre of Excellence for Musculoskeletal Ageing ResearchClinical, Metabolic and Molecular PhysiologyUniversity of NottinghamDerbyUK
| |
Collapse
|
42
|
Rindom E, Vissing K. Mechanosensitive Molecular Networks Involved in Transducing Resistance Exercise-Signals into Muscle Protein Accretion. Front Physiol 2016; 7:547. [PMID: 27909410 PMCID: PMC5112233 DOI: 10.3389/fphys.2016.00547] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2016] [Accepted: 10/31/2016] [Indexed: 02/05/2023] Open
Abstract
Loss of skeletal muscle myofibrillar protein with disease and/or inactivity can severely deteriorate muscle strength and function. Strategies to counteract wasting of muscle myofibrillar protein are therefore desirable and invite for considerations on the potential superiority of specific modes of resistance exercise and/or the adequacy of low load resistance exercise regimens as well as underlying mechanisms. In this regard, delineation of the potentially mechanosensitive molecular mechanisms underlying muscle protein synthesis (MPS), may contribute to an understanding on how differentiated resistance exercise can transduce a mechanical signal into stimulation of muscle accretion. Recent findings suggest specific upstream exercise-induced mechano-sensitive myocellular signaling pathways to converge on mammalian target of rapamycin complex 1 (mTORC1), to influence MPS. This may e.g. implicate mechanical activation of signaling through a diacylglycerol kinase (DGKζ)-phosphatidic acid (PA) axis or implicate integrin deformation to signal through a Focal adhesion kinase (FAK)-Tuberous Sclerosis Complex 2 (TSC2)-Ras homolog enriched in brain (Rheb) axis. Moreover, since initiation of translation is reliant on mRNA, it is also relevant to consider potentially mechanosensitive signaling pathways involved in muscle myofibrillar gene transcription and whether some of these pathways converge with those affecting mTORC1 activation for MPS. In this regard, recent findings suggest how mechanical stress may implicate integrin deformation and/or actin dynamics to signal through a Ras homolog gene family member A protein (RhoA)-striated muscle activator of Rho signaling (STARS) axis or implicate deformation of Notch to affect Bone Morphogenetic Protein (BMP) signaling through a small mother of decapentaplegic (Smad) axis.
Collapse
Affiliation(s)
- Emil Rindom
- Section of Sport Science, Department of Public Health, Aarhus UniversityAarhus, Denmark; Department of Biomedicine, Aarhus UniversityAarhus, Denmark
| | - Kristian Vissing
- Section of Sport Science, Department of Public Health, Aarhus University Aarhus, Denmark
| |
Collapse
|
43
|
Abstract
The skeletal muscle phenotype is subject to considerable malleability depending on use as well as internal and external cues. In humans, low-load endurance-type exercise leads to qualitative changes of muscle tissue characterized by an increase in structures supporting oxygen delivery and consumption, such as capillaries and mitochondria. High-load strength-type exercise leads to growth of muscle fibers dominated by an increase in contractile proteins. In endurance exercise, stress-induced signaling leads to transcriptional upregulation of genes, with Ca(2+) signaling and the energy status of the muscle cells sensed through AMPK being major input determinants. Several interrelated signaling pathways converge on the transcriptional co-activator PGC-1α, perceived to be the coordinator of much of the transcriptional and post-transcriptional processes. Strength training is dominated by a translational upregulation controlled by mTORC1. mTORC1 is mainly regulated by an insulin- and/or growth-factor-dependent signaling cascade as well as mechanical and nutritional cues. Muscle growth is further supported by DNA recruitment through activation and incorporation of satellite cells. In addition, there are several negative regulators of muscle mass. We currently have a good descriptive understanding of the molecular mechanisms controlling the muscle phenotype. The topology of signaling networks seems highly conserved among species, with the signaling outcome being dependent on the particular way individual species make use of the options offered by the multi-nodal networks. As a consequence, muscle structural and functional modifications can be achieved by an almost unlimited combination of inputs and downstream signaling events.
Collapse
Affiliation(s)
- Hans Hoppeler
- Emeritus Department of Anatomy, University of Bern, Baltzerstrasse 2, Bern 9 CH-3000, Switzerland
| |
Collapse
|
44
|
Camera DM, Smiles WJ, Hawley JA. Exercise-induced skeletal muscle signaling pathways and human athletic performance. Free Radic Biol Med 2016; 98:131-143. [PMID: 26876650 DOI: 10.1016/j.freeradbiomed.2016.02.007] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2015] [Revised: 01/28/2016] [Accepted: 02/03/2016] [Indexed: 12/18/2022]
Abstract
Skeletal muscle is a highly malleable tissue capable of altering its phenotype in response to external stimuli including exercise. This response is determined by the mode, (endurance- versus resistance-based), volume, intensity and frequency of exercise performed with the magnitude of this response-adaptation the basis for enhanced physical work capacity. However, training-induced adaptations in skeletal muscle are variable and unpredictable between individuals. With the recent application of molecular techniques to exercise biology, there has been a greater understanding of the multiplicity and complexity of cellular networks involved in exercise responses. This review summarizes the molecular and cellular events mediating adaptation processes in skeletal muscle in response to exercise. We discuss established and novel cell signaling proteins mediating key physiological responses associated with enhanced exercise performance and the capacity for reactive oxygen and nitrogen species to modulate training adaptation responses. We also examine the molecular bases underpinning heterogeneous responses to resistance and endurance exercise and the dissociation between molecular 'markers' of training adaptation and subsequent exercise performance.
Collapse
Affiliation(s)
- Donny M Camera
- Centre for Exercise and Nutrition, Mary MacKillop Institute for Health Research, Australian Catholic University, Melbourne, Vic. 3065, Australia
| | - William J Smiles
- Centre for Exercise and Nutrition, Mary MacKillop Institute for Health Research, Australian Catholic University, Melbourne, Vic. 3065, Australia
| | - John A Hawley
- Centre for Exercise and Nutrition, Mary MacKillop Institute for Health Research, Australian Catholic University, Melbourne, Vic. 3065, Australia; Research Institute for Sport and Exercise Sciences, Liverpool John Moores University, Liverpool L3 3AF, United Kingdom.
| |
Collapse
|
45
|
Qaisar R, Bhaskaran S, Van Remmen H. Muscle fiber type diversification during exercise and regeneration. Free Radic Biol Med 2016; 98:56-67. [PMID: 27032709 DOI: 10.1016/j.freeradbiomed.2016.03.025] [Citation(s) in RCA: 132] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2015] [Revised: 03/01/2016] [Accepted: 03/24/2016] [Indexed: 01/15/2023]
Abstract
The plasticity of skeletal muscle can be traced down to extensive metabolic, structural and molecular remodeling at the single fiber level. Skeletal muscle is comprised of different fiber types that are the basis of muscle plasticity in response to various functional demands. Resistance and endurance exercises are two external stimuli that differ in their duration and intensity of contraction and elicit markedly different responses in muscles adaptation. Further, eccentric contractions that are associated with exercise-induced injuries, elicit varied muscle adaptation and regenerative responses. Most adaptive changes are fiber type-specific and are highly influenced by diverse structural, metabolic and functional characteristics of individual fiber types. Regulation of signaling pathways by reactive oxygen species (ROS) and oxidative stress also plays an important role in muscle fiber adaptation during exercise. This review focuses on cellular and molecular responses that regulate the adaptation of skeletal muscle to exercise and exercise-related injuries.
Collapse
Affiliation(s)
- Rizwan Qaisar
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, 825 NE 13th Street, Oklahoma City, OK 73104, USA
| | - Shylesh Bhaskaran
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, 825 NE 13th Street, Oklahoma City, OK 73104, USA
| | - Holly Van Remmen
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, 825 NE 13th Street, Oklahoma City, OK 73104, USA.
| |
Collapse
|
46
|
Zheng X, Bao W, Yang J, Zhang T, Sun D, Liang Y, Li S, Wang Y, Feng X, Hao H, Wang Z. Focal Adhesion Kinase Directly Interacts with TSC2 Through Its FAT Domain and Regulates Cell Proliferation in Cashmere Goat Fetal Fibroblasts. DNA Cell Biol 2016; 35:480-8. [PMID: 27380318 DOI: 10.1089/dna.2015.3033] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Focal adhesion kinase (FAK) is a cytoplasmic nonreceptor tyrosine kinase that senses a variety of extracellular signals, such as growth factors and integrins, to control the process of cell proliferation and metabolism. We cloned three goat FAK transcript variants (KM655805, KM658268, and KM658269) that encode 1052, 1006, and 962 amino-acid residue proteins. Bioinformatics analysis indicated that the putative FAK protein contains an FERM domain, a PTK domain, two Proline-rich regions, and a focal adhesion-targeting (FAT) domain. All the three transcript variants of FAK were detected in seven different goat tissues, and variant 1 had the most accumulation whereas variant 2 and variant 3 had lower accumulation. Treatment of goat fetal fibroblasts (GFbs) with a specific FAK inhibitor, TAE226, inhibited cell proliferation (p < 0.05) and induced damage to the cell morphology in a dose- and time-dependent manner. Further research demonstrated that FAK directly interacted with TSC2 (Tuberous sclerosis 2) tuberin domain through its C-terminus, which contains the complete FAT domain. In conclusion, our results indicated that FAK may be widely expressed in Cashmere goat tissues and its products participate in the mammalian target of rapamycin signaling pathway and cell proliferation through a direct interaction with TSC2 in GFBs.
Collapse
Affiliation(s)
- Xu Zheng
- 1 College of Life Sciences, Inner Mongolia University , Hohhot, People's Republic of China .,2 Hulunbeier Municipal People's Hospital , Hailaer, People's Republic of China
| | - Wenlei Bao
- 1 College of Life Sciences, Inner Mongolia University , Hohhot, People's Republic of China
| | - Jiaofu Yang
- 1 College of Life Sciences, Inner Mongolia University , Hohhot, People's Republic of China
| | - Tao Zhang
- 1 College of Life Sciences, Inner Mongolia University , Hohhot, People's Republic of China
| | - Dongsheng Sun
- 3 Department of Oncology, Kailuan General Hospital , Tangshan, People's Republic of China
| | - Yan Liang
- 1 College of Life Sciences, Inner Mongolia University , Hohhot, People's Republic of China
| | - Shuyu Li
- 1 College of Life Sciences, Inner Mongolia University , Hohhot, People's Republic of China
| | - Yanfeng Wang
- 1 College of Life Sciences, Inner Mongolia University , Hohhot, People's Republic of China
| | - Xue Feng
- 1 College of Life Sciences, Inner Mongolia University , Hohhot, People's Republic of China
| | - Huifang Hao
- 1 College of Life Sciences, Inner Mongolia University , Hohhot, People's Republic of China
| | - Zhigang Wang
- 1 College of Life Sciences, Inner Mongolia University , Hohhot, People's Republic of China
| |
Collapse
|
47
|
The hibernating South American marsupial, Dromiciops gliroides, displays torpor-sensitive microRNA expression patterns. Sci Rep 2016; 6:24627. [PMID: 27090740 PMCID: PMC4835794 DOI: 10.1038/srep24627] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2016] [Accepted: 03/31/2016] [Indexed: 01/05/2023] Open
Abstract
When faced with adverse environmental conditions, the marsupial Dromiciops gliroides uses either daily or seasonal torpor to support survival and is the only known hibernating mammal in South America. As the sole living representative of the ancient Order Microbiotheria, this species can provide crucial information about the evolutionary origins and biochemical mechanisms of hibernation. Hibernation is a complex energy-saving strategy that involves changes in gene expression that are elicited in part by microRNAs. To better elucidate the role of microRNAs in orchestrating hypometabolism, a modified stem-loop technique and quantitative PCR were used to characterize the relative expression levels of 85 microRNAs in liver and skeletal muscle of control and torpid D. gliroides. Thirty-nine microRNAs were differentially regulated during torpor; of these, 35 were downregulated in liver and 11 were differentially expressed in skeletal muscle. Bioinformatic analysis predicted that the downregulated liver microRNAs were associated with activation of MAPK, PI3K-Akt and mTOR pathways, suggesting their importance in facilitating marsupial torpor. In skeletal muscle, hibernation-responsive microRNAs were predicted to regulate focal adhesion, ErbB, and mTOR pathways, indicating a promotion of muscle maintenance mechanisms. These tissue-specific responses suggest that microRNAs regulate key molecular pathways that facilitate hibernation, thermoregulation, and prevention of muscle disuse atrophy.
Collapse
|
48
|
Riedl I, Osler ME, Björnholm M, Egan B, Nader GA, Chibalin AV, Zierath JR. AMPKγ3 is dispensable for skeletal muscle hypertrophy induced by functional overload. Am J Physiol Endocrinol Metab 2016; 310:E461-72. [PMID: 26758685 PMCID: PMC4796264 DOI: 10.1152/ajpendo.00387.2015] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2015] [Accepted: 01/06/2016] [Indexed: 12/25/2022]
Abstract
Mechanisms regulating skeletal muscle growth involve a balance between the activity of serine/threonine protein kinases, including the mammalian target of rapamycin (mTOR) and 5'-AMP-activated protein kinase (AMPK). The contribution of different AMPK subunits to the regulation of cell growth size remains inadequately characterized. Using AMPKγ3 mutant-overexpressing transgenic Tg-Prkag3(225Q) and AMPKγ3-knockout (Prkag3(-/-)) mice, we investigated the requirement for the AMPKγ3 isoform in functional overload-induced muscle hypertrophy. Although the genetic disruption of the γ3 isoform did not impair muscle growth, control sham-operated AMPKγ3-transgenic mice displayed heavier plantaris muscles in response to overload hypertrophy and underwent smaller mass gain and lower Igf1 expression compared with wild-type littermates. The mTOR signaling pathway was upregulated with functional overload but unchanged between genetically modified animals and wild-type littermates. Differences in AMPK-related signaling pathways between transgenic, knockout, and wild-type mice did not impact muscle hypertrophy. Glycogen content was increased following overload in wild-type mice. In conclusion, our functional, transcriptional, and signaling data provide evidence against the involvement of the AMPKγ3 isoform in the regulation of skeletal muscle hypertrophy. Thus, the AMPKγ3 isoform is dispensable for functional overload-induced muscle growth. Mechanical loading can override signaling pathways that act as negative effectors of mTOR signaling and consequently promote skeletal muscle hypertrophy.
Collapse
Affiliation(s)
- Isabelle Riedl
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - Megan E Osler
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - Marie Björnholm
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - Brendan Egan
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden; Institute for Sport and Health, School of Public Health, Physiotherapy, and Population Science, University College Dublin, Belfield, Dublin, Ireland
| | - Gustavo A Nader
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden; and
| | - Alexander V Chibalin
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - Juleen R Zierath
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden; Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden; and
| |
Collapse
|
49
|
Brook MS, Wilkinson DJ, Phillips BE, Perez-Schindler J, Philp A, Smith K, Atherton PJ. Skeletal muscle homeostasis and plasticity in youth and ageing: impact of nutrition and exercise. Acta Physiol (Oxf) 2016; 216:15-41. [PMID: 26010896 PMCID: PMC4843955 DOI: 10.1111/apha.12532] [Citation(s) in RCA: 114] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2014] [Revised: 11/10/2014] [Accepted: 05/18/2015] [Indexed: 12/18/2022]
Abstract
Skeletal muscles comprise a substantial portion of whole body mass and are integral for locomotion and metabolic health. Increasing age is associated with declines in both muscle mass and function (e.g. strength‐related performance, power) with declines in muscle function quantitatively outweighing those in muscle volume. The mechanisms behind these declines are multi‐faceted involving both intrinsic age‐related metabolic dysregulation and environmental influences such as nutritional and physical activity. Ageing is associated with a degree of ‘anabolic resistance’ to these key environmental inputs, which likely accelerates the intrinsic processes driving ageing. On this basis, strategies to sensitize and/or promote anabolic responses to nutrition and physical activity are likely to be imperative in alleviating the progression and trajectory of sarcopenia. Both resistance‐ and aerobic‐type exercises are likely to confer functional and health benefits in older age, and a clutch of research suggests that enhancement of anabolic responsiveness to exercise and/or nutrition may be achieved by optimizing modifications of muscle‐loading paradigms (workload, volume, blood flow restriction) or nutritional support (e.g. essential amino acid/leucine) patterns. Nonetheless, more work is needed in which a more holistic view in ageing studies is taken into account. This should include improved characterization of older study recruits, that is physical activity/nutritional behaviours, to limit confounding variables influencing whether findings are attributable to age, or other environmental influences. Nonetheless, on balance, ageing is associated with declines in muscle mass and function and a partially related decline in aerobic capacity. There is also good evidence that metabolic flexibility is impaired in older age.
Collapse
Affiliation(s)
- M. S. Brook
- MRC-ARUK Centre of Excellence for Musculoskeletal Ageing Research, Clinical Metabolic and Molecular Physiology; University of Nottingham; Royal Derby Hospital Centre; Derby UK
| | - D. J. Wilkinson
- MRC-ARUK Centre of Excellence for Musculoskeletal Ageing Research, Clinical Metabolic and Molecular Physiology; University of Nottingham; Royal Derby Hospital Centre; Derby UK
| | - B. E. Phillips
- MRC-ARUK Centre of Excellence for Musculoskeletal Ageing Research, Clinical Metabolic and Molecular Physiology; University of Nottingham; Royal Derby Hospital Centre; Derby UK
| | - J. Perez-Schindler
- MRC-ARUK Centre of Excellence for Musculoskeletal Ageing Research, School of Sport, Exercise and Rehabilitation Sciences; University of Birmingham; Birmingham UK
| | - A. Philp
- MRC-ARUK Centre of Excellence for Musculoskeletal Ageing Research, School of Sport, Exercise and Rehabilitation Sciences; University of Birmingham; Birmingham UK
| | - K. Smith
- MRC-ARUK Centre of Excellence for Musculoskeletal Ageing Research, Clinical Metabolic and Molecular Physiology; University of Nottingham; Royal Derby Hospital Centre; Derby UK
| | - P. J. Atherton
- MRC-ARUK Centre of Excellence for Musculoskeletal Ageing Research, Clinical Metabolic and Molecular Physiology; University of Nottingham; Royal Derby Hospital Centre; Derby UK
| |
Collapse
|
50
|
Graham ZA, Gallagher PM, Cardozo CP. Focal adhesion kinase and its role in skeletal muscle. J Muscle Res Cell Motil 2015; 36:305-15. [PMID: 26142360 PMCID: PMC4659753 DOI: 10.1007/s10974-015-9415-3] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2015] [Accepted: 06/15/2015] [Indexed: 10/23/2022]
Abstract
Skeletal muscle has a remarkable ability to respond to different physical stresses. Loading muscle through exercise, either anaerobic or aerobic, can lead to increases in muscle size and function while, conversely, the absence of muscle loading stimulates rapid decreases in size and function. A principal mediator of this load-induced change is focal adhesion kinase (FAK), a downstream non-receptor tyrosine kinase that translates the cytoskeletal stress and strain signals transmitted across the cytoplasmic membrane by integrins to activate multiple anti-apoptotic and cell growth pathways. Changes in FAK expression and phosphorylation have been found to correlate to specific developmental states in myoblast differentiation, muscle fiber formation and muscle size in response to loading and unloading. With the capability to regulate costamere formation, hypertrophy and glucose metabolism, FAK is a molecule with diverse functions that are important in regulating muscle cell health.
Collapse
Affiliation(s)
- Zachary A Graham
- Center of Excellence for the Medical Consequences of Spinal Cord Injury, James J. Peters Veterans Affairs Medical Center, 130 W. Kingsbridge Rd., Bronx, NY, 10468, USA
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | - Christopher P Cardozo
- Center of Excellence for the Medical Consequences of Spinal Cord Injury, James J. Peters Veterans Affairs Medical Center, 130 W. Kingsbridge Rd., Bronx, NY, 10468, USA.
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Department of Rehabilitation Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Department of Pharmacology and Systems Therapeutics, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|