51
|
Jacko D, Schaaf K, Masur L, Windoffer H, Aussieker T, Schiffer T, Zacher J, Bloch W, Gehlert S. Repeated and Interrupted Resistance Exercise Induces the Desensitization and Re-Sensitization of mTOR-Related Signaling in Human Skeletal Muscle Fibers. Int J Mol Sci 2022; 23:ijms23105431. [PMID: 35628242 PMCID: PMC9141560 DOI: 10.3390/ijms23105431] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 05/07/2022] [Accepted: 05/09/2022] [Indexed: 02/06/2023] Open
Abstract
The acute resistance exercise (RE)-induced phosphorylation of mTOR-related signaling proteins in skeletal muscle can be blunted after repeated RE. The time frame in which the phosphorylation (p) of mTORS2448, p70S6kT421/S424, and rpS6S235/236 will be reduced during an RE training period in humans and whether progressive (PR) loading can counteract such a decline has not been described. (1) To enclose the time frame in which pmTORS2448, prpS6S235/236, and pp70S6kT421/S424 are acutely reduced after RE occurs during repeated RE. (2) To test whether PR will prevent that reduction compared to constant loading (CO) and (3) whether 10 days without RE may re-increase blunted signaling. Fourteen healthy males (24 ± 2.8 yrs.; 1.83 ± 0.1 cm; 79.3 ± 8.5 kg) were subjected to RE with either PR (n = 8) or CO (n = 6) loading. Subjects performed RE thrice per week, conducting three sets with 10−12 repetitions on a leg press and leg extension machine. Muscle biopsies were collected at rest (T0), 45 min after the first (T1), seventh (T7), 13th (T13), and 14th (X-T14) RE session. No differences were found between PR and CO for any parameter. Thus, the groups were combined, and the results show the merged values. prpS6S235/236 and pp70s6kT421/S424 were increased at T1, but were already reduced at T7 and up to T13 compared to T1. Ten days without RE re-increased prpS6S235/236 and pp70S6kT421/S424 at X-T14 to a level comparable to that of T1. pmTORS2448 was increased from T1 to X-T14 and did not decline over the training period. Single-fiber immunohistochemistry revealed a reduction in prpS6S235/236 in type I fibers from T1 to T13 and a re-increase at X-T14, which was more augmented in type II fibers at T13 (p < 0.05). The entity of myofibers revealed a high heterogeneity in the level of prpS6S235/236, possibly reflecting individual contraction-induced stress during RE. The type I and II myofiber diameter increased from T0 and T1 to T13 and X-T14 (p < 0.05) prpS6S235/236 and pp70s6kT421/S424 reflect RE-induced states of desensitization and re-sensitization in dependency on frequent loading by RE, but also by its cessation.
Collapse
Affiliation(s)
- Daniel Jacko
- Department of Molecular and Cellular Sports Medicine, Institute of Cardiovascular Research and Sports Medicine, German Sport University Cologne, 50933 Cologne, Germany; (D.J.); (K.S.); (L.M.); (H.W.); (T.A.); (W.B.)
- Olympic Base Center NRW/Rhineland, 50933 Cologne, Germany
| | - Kirill Schaaf
- Department of Molecular and Cellular Sports Medicine, Institute of Cardiovascular Research and Sports Medicine, German Sport University Cologne, 50933 Cologne, Germany; (D.J.); (K.S.); (L.M.); (H.W.); (T.A.); (W.B.)
| | - Lukas Masur
- Department of Molecular and Cellular Sports Medicine, Institute of Cardiovascular Research and Sports Medicine, German Sport University Cologne, 50933 Cologne, Germany; (D.J.); (K.S.); (L.M.); (H.W.); (T.A.); (W.B.)
| | - Hannes Windoffer
- Department of Molecular and Cellular Sports Medicine, Institute of Cardiovascular Research and Sports Medicine, German Sport University Cologne, 50933 Cologne, Germany; (D.J.); (K.S.); (L.M.); (H.W.); (T.A.); (W.B.)
| | - Thorben Aussieker
- Department of Molecular and Cellular Sports Medicine, Institute of Cardiovascular Research and Sports Medicine, German Sport University Cologne, 50933 Cologne, Germany; (D.J.); (K.S.); (L.M.); (H.W.); (T.A.); (W.B.)
| | - Thorsten Schiffer
- Outpatient Clinic for Sports Traumatology and Public Health Consultation, German Sport University Cologne, 50933 Cologne, Germany;
| | - Jonas Zacher
- Department ofPreventative and Rehabilitative Sports and Performance Medicine, Institute of Cardiology and Sports Medicine, German Sports University Cologne, 50933 Cologne, Germany;
| | - Wilhelm Bloch
- Department of Molecular and Cellular Sports Medicine, Institute of Cardiovascular Research and Sports Medicine, German Sport University Cologne, 50933 Cologne, Germany; (D.J.); (K.S.); (L.M.); (H.W.); (T.A.); (W.B.)
- German Research Centre of Elite Sport (Momentum), German Sport University Cologne, 50933 Cologne, Germany
| | - Sebastian Gehlert
- Department of Molecular and Cellular Sports Medicine, Institute of Cardiovascular Research and Sports Medicine, German Sport University Cologne, 50933 Cologne, Germany; (D.J.); (K.S.); (L.M.); (H.W.); (T.A.); (W.B.)
- Institute of Sport Science, Biosciences of Sports, University of Hildesheim, 31141 Hildesheim, Germany
- Correspondence: ; Tel.: +49-(0)-5121-883-580; Fax: +49-(0)-5121-883-591
| |
Collapse
|
52
|
Myofibrillar protein synthesis rates are increased in chronically exercised skeletal muscle despite decreased anabolic signaling. Sci Rep 2022; 12:7553. [PMID: 35534615 PMCID: PMC9085756 DOI: 10.1038/s41598-022-11621-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Accepted: 03/23/2022] [Indexed: 01/05/2023] Open
Abstract
The molecular responses to acute resistance exercise are well characterized. However, how cellular signals change over time to modulate chronic adaptations to more prolonged exercise training is less well understood. We investigated anabolic signaling and muscle protein synthesis rates at several time points after acute and chronic eccentric loading. Adult rat tibialis anterior muscle was stimulated for six sets of ten repetitions, and the muscle was collected at 0 h, 6 h, 18 h and 48 h. In the last group of animals, 48 h after the first exercise bout a second bout was conducted, and the muscle was collected 6 h later (54 h total). In a second experiment, rats were exposed to four exercise sessions over the course of 2 weeks. Anabolic signaling increased robustly 6 h after the first bout returning to baseline between 18 and 48 h. Interestingly, 6 h after the second bout mTORC1 activity was significantly lower than following the first bout. In the chronically exercised rats, we found baseline anabolic signaling was decreased, whereas myofibrillar protein synthesis (MPS) was substantially increased, 48 h after the last bout of exercise. The increase in MPS occurred in the absence of changes to muscle fiber size or mass. In conclusion, we find that anabolic signaling is already diminished after the second bout of acute resistance type exercise. Further, chronic exposure to resistance type exercise training results in decreased basal anabolic signaling but increased overall MPS rates.
Collapse
|
53
|
Buerger AN, Parente CE, Harris JP, Watts EG, Wormington AM, Bisesi JH. Impacts of diethylhexyl phthalate and overfeeding on physical fitness and lipid mobilization in Danio rerio (zebrafish). CHEMOSPHERE 2022; 295:133703. [PMID: 35066078 DOI: 10.1016/j.chemosphere.2022.133703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 01/04/2022] [Accepted: 01/18/2022] [Indexed: 06/14/2023]
Abstract
As the prevalence of obesity has steadily increased on a global scale, research has shifted to explore potential contributors to this pandemic beyond overeating and lack of exercise. Environmental chemical contaminants, known as obesogens, alter metabolic processes and exacerbate the obese phenotype. Diethylhexyl phthalate (DEHP) is a common chemical plasticizer found in medical supplies, food packaging, and polyvinyl materials, and has been identified as a probable obesogen. This study investigated the hypothesis that co-exposure to DEHP and overfeeding would result in decreased lipid mobilization and physical fitness in Danio rerio (zebrafish). Four treatment groups were randomly assigned: Regular Fed (control, 10 mg/fish/day with 0 mg/kg DEHP), Overfed (20 mg/fish/day with 0 mg/kg DEHP), Regular Fed + DEHP (10 mg/fish/day with 3 mg/kg DEHP), Overfed + DEHP (20 mg/fish/day with 3 mg/kg DEHP). After 24 weeks, swim tunnel assays were conducted on half of the zebrafish from each treatment to measure critical swimming speeds (Ucrit); the other fish were euthanized without swimming. Body mass index (BMI) was measured, and tissues were collected for blood lipid characterization and gene expression analyses. Co-exposure to DEHP and overfeeding decreased swim performance as measured by Ucrit. While no differences in blood lipids were observed with DEHP exposure, differential expression of genes related to lipid metabolism and utilization in the gastrointestinal and liver tissue suggests alterations in metabolism and lipid packaging, which may impact utilization and ability to mobilize lipid reserves during physical activity following chronic exposures.
Collapse
Affiliation(s)
- Amanda N Buerger
- Department of Environmental and Global Health, University of Florida, Gainesville, FL, USA; Center for Environmental and Human Toxicology, University of Florida, Gainesville, FL, USA
| | - Caitlyn E Parente
- Center for Environmental and Human Toxicology, University of Florida, Gainesville, FL, USA; Department of Chemistry, University of Florida, Gainesville, FL, USA
| | - Jason P Harris
- Center for Environmental and Human Toxicology, University of Florida, Gainesville, FL, USA; Department of Biology, University of Florida, Gainesville, FL, USA
| | - Emily G Watts
- Center for Environmental and Human Toxicology, University of Florida, Gainesville, FL, USA; Department of Chemistry, University of Florida, Gainesville, FL, USA
| | - Alexis M Wormington
- Center for Environmental and Human Toxicology, University of Florida, Gainesville, FL, USA
| | - Joseph H Bisesi
- Department of Environmental and Global Health, University of Florida, Gainesville, FL, USA; Center for Environmental and Human Toxicology, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
54
|
Wirth K, Keiner M, Fuhrmann S, Nimmerichter A, Haff GG. Strength Training in Swimming. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:ijerph19095369. [PMID: 35564764 PMCID: PMC9100337 DOI: 10.3390/ijerph19095369] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 04/22/2022] [Accepted: 04/24/2022] [Indexed: 11/16/2022]
Abstract
This narrative review deals with the topic of strength training in swimming, which has been a controversial issue for decades. It is not only about the importance for the performance at start, turn and swim speed, but also about the question of how to design a strength training program. Different approaches are discussed in the literature, with two aspects in the foreground. On the one hand is the discussion about the optimal intensity in strength training and, on the other hand, is the question of how specific strength training should be designed. In addition to a summary of the current state of research regarding the importance of strength training for swimming, the article shows which physiological adaptations should be achieved in order to be able to increase performance in the long term. Furthermore, an attempt is made to explain why some training contents seem to be rather unsuitable when it comes to increasing strength as a basis for higher performance in the start, turn and clean swimming. Practical training consequences are then derived from this. Regardless of the athlete's performance development, preventive aspects should also be considered in the discussion. The article provides a critical overview of the abovementioned key issues. The most important points when designing a strength training program for swimming are a sufficiently high-load intensity to increase maximum strength, which in turn is the basis for power, year-round strength training, parallel to swim training and working on the transfer of acquired strength skills in swim training, and not through supposedly specific strength training exercises on land or in the water.
Collapse
Affiliation(s)
- Klaus Wirth
- Faculty of Training and Sports Sciences, University of Applied Sciences Wiener Neustadt, 2700 Wiener Neustadt, Austria;
- Correspondence:
| | - Michael Keiner
- Department of Sport Science, University of Health and Sports, 85737 Ismaning, Germany;
| | - Stefan Fuhrmann
- Olympic Training and Testing Centre Hamburg/Schleswig-Holstein, 22049 Hamburg, Germany;
| | - Alfred Nimmerichter
- Faculty of Training and Sports Sciences, University of Applied Sciences Wiener Neustadt, 2700 Wiener Neustadt, Austria;
| | - G. Gregory Haff
- School of Medical and Health Sciences, Edith Cowan University, Joondalup, WA 6027, Australia;
| |
Collapse
|
55
|
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: 22] [Impact Index Per Article: 7.3] [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
|
56
|
Chang SW, Yoshihara T, Tsuzuki T, Natsume T, Kakigi R, Machida S, Naito H. Circadian rhythms modulate the effect of eccentric exercise on rat soleus muscles. PLoS One 2022; 17:e0264171. [PMID: 35213577 PMCID: PMC8880858 DOI: 10.1371/journal.pone.0264171] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2021] [Accepted: 02/04/2022] [Indexed: 11/23/2022] Open
Abstract
We investigated whether time-of-day dependent changes in the rat soleus (SOL) muscle size, after eccentric exercises, operate via the mechanistic target of rapamycin (mTOR) signaling pathway. For our first experiment, we assigned 9-week-old male Wistar rats randomly into four groups: light phase (zeitgeber time; ZT6) non-trained control, dark phase (ZT18) non-trained control, light phase-trained, and dark phase-trained. Trained animals performed 90 min of downhill running once every 3 d for 8 weeks. The second experiment involved dividing 9-week-old male Wistar rats to control and exercise groups. The latter were subjected to 15 min of downhill running at ZT6 and ZT18. The absolute (+12.8%) and relative (+9.4%) SOL muscle weights were higher in the light phase-trained group. p70S6K phosphorylation ratio was 42.6% higher in the SOL muscle of rats that had exercised only in light (non-trained ZT6). Collectively, the degree of muscle hypertrophy in SOL is time-of-day dependent, perhaps via the mTOR/p70S6K signaling.
Collapse
Affiliation(s)
- Shuo-wen Chang
- Graduate School of Health and Sports Science, Juntendo University, Chiba, Japan
- Department of Physical Education, National University of Tainan, Tainan, Taiwan
| | - Toshinori Yoshihara
- Graduate School of Health and Sports Science, Juntendo University, Chiba, Japan
| | - Takamasa Tsuzuki
- Graduate School of Health and Sports Science, Juntendo University, Chiba, Japan
- Faculty of Pharmacy, Meijo University, Nagoya, Aichi, Japan
| | - Toshiharu Natsume
- Graduate School of Health and Sports Science, Juntendo University, Chiba, Japan
- School of Medicine, Tokai University, Hiratsuka, Kanagawa, Japan
| | - Ryo Kakigi
- Faculty of Management & Information Sciences, Josai International University, Chiba, Japan
| | - Shuichi Machida
- Graduate School of Health and Sports Science, Juntendo University, Chiba, Japan
| | - Hisashi Naito
- Graduate School of Health and Sports Science, Juntendo University, Chiba, Japan
- * E-mail:
| |
Collapse
|
57
|
Nishimura Y, Jensen M, Bülow J, Thomsen TT, Arimitsu T, van Hall G, Fujita S, Holm L. Co-ingestion of cluster dextrin carbohydrate does not increase exogenous protein-derived amino acid release or myofibrillar protein synthesis following a whole-body resistance exercise in moderately trained younger males: a double-blinded randomized controlled crossover trial. Eur J Nutr 2022; 61:2475-2491. [PMID: 35182194 PMCID: PMC9279228 DOI: 10.1007/s00394-021-02782-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Accepted: 12/09/2021] [Indexed: 11/12/2022]
Abstract
Purpose This study investigates if co-ingestion of cluster dextrin (CDX) augments the appearance of intrinsically labeled meat protein hydrolysate-derived amino acid (D5-phenylalanine), Akt/mTORC1 signaling, and myofibrillar protein fractional synthetic rate (FSR). Methods Ten moderately trained healthy males (age: 21.5 ± 2.1 years, body mass: 75.7 ± 7.6 kg, body mass index (BMI): 22.9 ± 2.1 kg/m2) were included for a double-blinded randomized controlled crossover trial. Either 75 g of CDX or glucose (GLC) was given in conjunction with meat protein hydrolysate (0.6 g protein * FFM−1) following a whole-body resistance exercise. A primed-continuous intravenous infusion of L-[15N]-phenylalanine with serial muscle biopsies and venous blood sampling was performed. Results A time × group interaction effect was found for serum D5-phenylalanine enrichment (P < 0.01). Serum EAA and BCAA concentrations showed a main effect for group (P < 0.05). Tmax serum BCAA was greater in CDX as compared to GLC (P < 0.05). However, iAUC of all serum parameters did not differ between CDX and GLC (P > 0.05). Tmax serum EAA showed a trend towards a statistical significance favoring CDX over GLC. The phosphorylation of p70S6KThr389, rpS6Ser240/244, ERK1/2Thr202/Tyr204 was greater in CDX compared to GLC (P < 0.05). However, postprandial myofibrillar FSR did not differ between CDX and GLC (P = 0.17). Conclusion In moderately trained younger males, co-ingestion of CDX with meat protein hydrolysate does not augment the postprandial amino acid availability or myofibrillar FSR as compared to co-ingestion of GLC during the recovery from a whole-body resistance exercise despite an increased intramuscular signaling. Trial registration ClinicalTrials.gov ID: NCT03303729 (registered on October 3, 2017).
Collapse
Affiliation(s)
- Yusuke Nishimura
- School of Sport, Exercise and Rehabilitation Sciences, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK.,Department of Sport and Health Science, Ritsumeikan University, Shiga, Japan
| | - Mikkel Jensen
- Institute of Sports Medicine Copenhagen, Department of Orthopedic Surgery M, Bispebjerg Hospital, Copenhagen, Denmark
| | - Jacob Bülow
- Institute of Sports Medicine Copenhagen, Department of Orthopedic Surgery M, Bispebjerg Hospital, Copenhagen, Denmark
| | - Thomas Tagmose Thomsen
- Institute of Sports Medicine Copenhagen, Department of Orthopedic Surgery M, Bispebjerg Hospital, Copenhagen, Denmark
| | - Takuma Arimitsu
- Department of Sport and Health Science, Ritsumeikan University, Shiga, Japan
| | - Gerrit van Hall
- Clinical Metabolomics Core Facility, Department of Clinical Biochemistry, Rigshospitalet, Copenhagen, Denmark.,Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Satoshi Fujita
- Department of Sport and Health Science, Ritsumeikan University, Shiga, Japan
| | - Lars Holm
- School of Sport, Exercise and Rehabilitation Sciences, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK. .,Institute of Sports Medicine Copenhagen, Department of Orthopedic Surgery M, Bispebjerg Hospital, Copenhagen, Denmark. .,Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
58
|
Blood Flow Restriction Therapy and Its Use for Rehabilitation and Return to Sport: Physiology, Application, and Guidelines for Implementation. Arthrosc Sports Med Rehabil 2022; 4:e71-e76. [PMID: 35141538 PMCID: PMC8811521 DOI: 10.1016/j.asmr.2021.09.025] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Accepted: 09/22/2021] [Indexed: 12/26/2022] Open
Abstract
Blood flow restriction (BFR) is an expanding rehabilitation modality that uses a tourniquet to reduce arterial inflow and occlude venous outflow in the setting of resistance training or exercise. Initially, this technique was seen as a way to stimulate muscular development, but improved understanding of its physiologic benefits and mechanism of action has allowed for innovative clinical applications. BFR represents a way to decrease stress placed on the joints without compromising improvements in strength, whereas for postoperative, injured, or load-compromised individuals BFR represents a way to accelerate recovery and prevent atrophy. There is also growing evidence to suggest that it augments cardiovascular fitness and attenuates pain. The purpose of this review is to highlight the physiology and evidence behind the various applications of BFR, with a focus on postoperative rehabilitation. While much remains to be learned, it is clear that blood flow restriction therapy stimulates muscle hypertrophy via a synergistic response to metabolic stress and mechanical tension, with supplemental benefits on cardiovascular fitness and pain. New forms of BFR and expanding applications in postoperative patients and athletes hold promise for expedited recovery. Continued adherence to rehabilitation guidelines and exploration of BFRs physiology and various applications will help optimize its effect and prescription. Level of Evidence V, expert opinion.
Collapse
|
59
|
Wackerhage H, Vechetti IJ, Baumert P, Gehlert S, Becker L, Jaspers RT, de Angelis MH. Does a Hypertrophying Muscle Fibre Reprogramme its Metabolism Similar to a Cancer Cell? Sports Med 2022; 52:2569-2578. [PMID: 35460513 PMCID: PMC9584876 DOI: 10.1007/s40279-022-01676-1] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/17/2022] [Indexed: 02/01/2023]
Abstract
In 1924, Otto Warburg asked "How does the metabolism of a growing tissue differ from that of a non-growing tissue?" Currently, we know that proliferating healthy and cancer cells reprogramme their metabolism. This typically includes increased glucose uptake, glycolytic flux and lactate synthesis. A key function of this reprogramming is to channel glycolytic intermediates and other metabolites into anabolic reactions such as nucleotide-RNA/DNA synthesis, amino acid-protein synthesis and the synthesis of, for example, acetyl and methyl groups for epigenetic modification. In this review, we discuss evidence that a hypertrophying muscle similarly takes up more glucose and reprogrammes its metabolism to channel energy metabolites into anabolic pathways. We specifically discuss the functions of the cancer-associated enzymes phosphoglycerate dehydrogenase and pyruvate kinase muscle 2 in skeletal muscle. In addition, we ask whether increased glucose uptake by a hypertrophying muscle explains why muscularity is often negatively associated with type 2 diabetes mellitus and obesity.
Collapse
Affiliation(s)
- Henning Wackerhage
- Exercise Biology Group, Department of Health and Sports Sciences, Technical University of Munich, Munich, Germany
| | - Ivan J. Vechetti
- Department of Nutrition and Health Sciences, College of Education and Human Sciences, University of Nebraska-Lincoln, Lincoln, NE USA
| | - Philipp Baumert
- Exercise Biology Group, Department of Health and Sports Sciences, Technical University of Munich, Munich, Germany
| | - Sebastian Gehlert
- Department of Biosciences of Sports, Institute for Sports Science, University of Hildesheim, Hildesheim, Germany
| | - Lore Becker
- Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Munich, Germany
| | - Richard T. Jaspers
- Laboratory for Myology, Behavioural and Movement Sciences, Amsterdam Movement Sciences, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Martin Hrabě de Angelis
- Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Munich, Germany ,German Center for Diabetes Research (DZD), Neuherberg, Germany ,Chair of Experimental Genetics, TUM School of Life Sciences, Technische Universität München, Freising, Germany
| |
Collapse
|
60
|
Li Z, Du X, Wen L, Li Y, Qin J, Chen Z, Huang Y, Wu X, Luo H, Lin Y, Ye H. Transcriptome analysis reveals the involvement of ubiquitin-proteasome pathway in the regulation of muscle growth of rice flower carp. COMPARATIVE BIOCHEMISTRY AND PHYSIOLOGY. PART D, GENOMICS & PROTEOMICS 2021; 41:100948. [PMID: 34942523 DOI: 10.1016/j.cbd.2021.100948] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Revised: 11/24/2021] [Accepted: 11/26/2021] [Indexed: 12/13/2022]
Abstract
Growth mechanism of economically important aquaculture species has aroused widespread interest among scholars. Rice flower carp (Cyprinus carpio), commonly cultured in rice-fish farming systems, shows wide variation in body mass at the same age, which limits the development of commercial aquaculture. In this study, muscle tissues from 20-month-old fish of different sizes were used for transcriptome analysis and muscle histological studies. The muscle histological analysis showed the muscle growth in rice flower carp main depends on the hypertrophic growth of muscle fibers. A total of 30,590 unigenes were generated by muscle trancriptome analysis, including 403 differentially expressed genes (DEGs). Of these, 157 DEGs were upregulated and 246 DEGs were downregulated. Nine unigenes related to the ubiquitin-proteasome pathway were identified using differential expression analysis. This study initially revealed that the differences in growth of rice flower carp could be due to hypertrophic growth of muscle fibers caused by higher protein deposition, and the ubiquitin-proteasome pathway was an important factor affecting the growth rate of rice flower carp. E3 ubiquitin-protein ligase ari7, g2e3, Neurl1 and rnf144ab were upregulated in the slow-growing fish, indicating the binding of ubiquitin to target protein was enhanced. Foxo3 was upregulated in the slow-growing fish, which could promote the muscle loss. Eif4a2 was upregulated in the fast-growing fish, increasing protein translation efficiency. Some genes related to active muscle contraction such as actb, actg, camk2a, and camk2b were upregulated in the fast-growing rice flower carp muscle. In summary, these results provide valuable information about the key genes for use as biomarkers of growth in selective breeding programs for rice flower carp and provide novel insights into the regulatory mechanisms of muscle growth.
Collapse
Affiliation(s)
- Zhe Li
- Key Laboratory of Freshwater Fish Reproduction and Development (Ministry of Education), College of Fisheries, Southwest University, Chongqing 402460, China
| | - Xuesong Du
- Guangxi Key Laboratory of Aquatic Genetic Breeding and Healthy Aquaculture, Guangxi Academy of Fisheries Science, Nanning 530021, China
| | - Luting Wen
- Guangxi Key Laboratory of Aquatic Genetic Breeding and Healthy Aquaculture, Guangxi Academy of Fisheries Science, Nanning 530021, China
| | - Yu Li
- Key Laboratory of Freshwater Fish Reproduction and Development (Ministry of Education), College of Fisheries, Southwest University, Chongqing 402460, China
| | - Junqi Qin
- Guangxi Key Laboratory of Aquatic Genetic Breeding and Healthy Aquaculture, Guangxi Academy of Fisheries Science, Nanning 530021, China
| | - Zhong Chen
- Guangxi Key Laboratory of Aquatic Genetic Breeding and Healthy Aquaculture, Guangxi Academy of Fisheries Science, Nanning 530021, China
| | - Yin Huang
- Guangxi Key Laboratory of Aquatic Genetic Breeding and Healthy Aquaculture, Guangxi Academy of Fisheries Science, Nanning 530021, China
| | - Xia Wu
- College of Life Science, Guangxi Normal University, Guilin 541006, China
| | - Hui Luo
- Key Laboratory of Freshwater Fish Reproduction and Development (Ministry of Education), College of Fisheries, Southwest University, Chongqing 402460, China.
| | - Yong Lin
- Guangxi Key Laboratory of Aquatic Genetic Breeding and Healthy Aquaculture, Guangxi Academy of Fisheries Science, Nanning 530021, China.
| | - Hua Ye
- Key Laboratory of Freshwater Fish Reproduction and Development (Ministry of Education), College of Fisheries, Southwest University, Chongqing 402460, China.
| |
Collapse
|
61
|
Multiple Applications of Different Exercise Modalities with Rodents. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:3898710. [PMID: 34868454 PMCID: PMC8639251 DOI: 10.1155/2021/3898710] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 09/14/2021] [Accepted: 11/12/2021] [Indexed: 12/29/2022]
Abstract
A large proportion of chronic diseases can be derived from a sedentary lifestyle. Raising physical activity awareness is indispensable, as lack of exercise is the fourth most common cause of death worldwide. Animal models in different research fields serve as important tools in the study of acute or chronic noncommunicable disorders. With the help of animal-based exercise research, exercise-mediated complex antioxidant and inflammatory pathways can be explored, which knowledge can be transferred to human studies. Whereas sustained physical activity has an enormous number of beneficial effects on many organ systems, these animal models are easily applicable in several research areas. This review is aimed at providing an overall picture of scientific research studies using animal models with a focus on different training modalities. Without wishing to be exhaustive, the most commonly used forms of exercise are presented.
Collapse
|
62
|
Leser JM, Harriot A, Buck HV, Ward CW, Stains JP. Aging, Osteo-Sarcopenia, and Musculoskeletal Mechano-Transduction. FRONTIERS IN REHABILITATION SCIENCES 2021; 2:782848. [PMID: 36004321 PMCID: PMC9396756 DOI: 10.3389/fresc.2021.782848] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Accepted: 11/10/2021] [Indexed: 11/13/2022]
Abstract
The decline in the mass and function of bone and muscle is an inevitable consequence of healthy aging with early onset and accelerated decline in those with chronic disease. Termed osteo-sarcopenia, this condition predisposes the decreased activity, falls, low-energy fractures, and increased risk of co-morbid disease that leads to musculoskeletal frailty. The biology of osteo-sarcopenia is most understood in the context of systemic neuro-endocrine and immune/inflammatory alterations that drive inflammation, oxidative stress, reduced autophagy, and cellular senescence in the bone and muscle. Here we integrate these concepts to our growing understanding of how bone and muscle senses, responds and adapts to mechanical load. We propose that age-related alterations in cytoskeletal mechanics alter load-sensing and mechano-transduction in bone osteocytes and muscle fibers which underscores osteo-sarcopenia. Lastly, we examine the evidence for exercise as an effective countermeasure to osteo-sarcopenia.
Collapse
Affiliation(s)
| | | | | | | | - Joseph P. Stains
- Department of Orthopaedics, University of Maryland School of Medicine, Baltimore, MD, United States
| |
Collapse
|
63
|
Valentino T, Figueiredo VC, Mobley CB, McCarthy JJ, Vechetti IJ. Evidence of myomiR regulation of the pentose phosphate pathway during mechanical load-induced hypertrophy. Physiol Rep 2021; 9:e15137. [PMID: 34889054 PMCID: PMC8661100 DOI: 10.14814/phy2.15137] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Revised: 10/13/2021] [Accepted: 11/10/2021] [Indexed: 12/29/2022] Open
Abstract
Many of the molecular and cellular mechanisms discovered to regulate skeletal muscle hypertrophy were first identified using the rodent synergist ablation model. This model reveals the intrinsic capability and necessary pathways of skeletal muscle growth in response to mechanical overload (MOV). Reminiscent of the rapid cellular growth observed with cancer, we hypothesized that in response to MOV, skeletal muscle would undergo metabolic programming to sustain increased demands to support hypertrophy. To test this hypothesis, we analyzed the gene expression of specific metabolic pathways taken from transcriptomic microarray data of a MOV time course. We found an upregulation of genes involved in the oxidative branch of the pentose phosphate pathways (PPP) and mitochondrial branch of the folate cycle suggesting an increase in the production of NADPH. In addition, we sought to determine the potential role of skeletal muscle-enriched microRNA (myomiRs) and satellite cells in the regulation of the metabolic pathways that changed during MOV. We observed an inverse pattern in gene expression between muscle-enriched myomiR-1 and its known target gene glucose-6-phosphate dehydrogenase, G6pdx, suggesting myomiR regulation of PPP activation in response to MOV. Satellite cell fusion had a significant but modest impact on PPP gene expression. These transcriptomic findings suggest the robust muscle hypertrophy induced by MOV requires enhanced redox metabolism via PPP production of NADPH which is potentially regulated by a myomiR network.
Collapse
Affiliation(s)
- Taylor Valentino
- Department of PhysiologyCollege of MedicineLexingtonKentuckyUSA
- Center for Muscle BiologyUniversity of KentuckyLexingtonKentuckyUSA
| | - Vandre C. Figueiredo
- Center for Muscle BiologyUniversity of KentuckyLexingtonKentuckyUSA
- Department of Physical TherapyCollege of Health SciencesUniversity of KentuckyLexingtonKentuckyUSA
| | | | - John J. McCarthy
- Department of PhysiologyCollege of MedicineLexingtonKentuckyUSA
- Center for Muscle BiologyUniversity of KentuckyLexingtonKentuckyUSA
| | - Ivan J. Vechetti
- Department of Nutrition and Health SciencesCollege of Education and Human SciencesUniversity of Nebraska‐LincolnLincolnNebraskaUSA
| |
Collapse
|
64
|
Shirai T, Uemichi K, Hidaka Y, Kitaoka Y, Takemasa T. Effect of lactate administration on mouse skeletal muscle under calorie restriction. Curr Res Physiol 2021; 4:202-208. [PMID: 34746839 PMCID: PMC8562144 DOI: 10.1016/j.crphys.2021.09.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 08/25/2021] [Accepted: 09/03/2021] [Indexed: 12/18/2022] Open
Abstract
Calorie restriction (CR) involves a reductions of calorie intake without altering the nutritional balance, and has many beneficial effects, such as improving oxidative metabolism and extending lifespan. However, CR decreases in skeletal muscle mass and fat mass in correlation with the reduction in food intake. Lactate is known to have potential as a signaling molecule rather than a metabolite during exercise. In this study, we examined the effects of the combination of caloric restriction and lactate administration on skeletal muscle adaptation in order to elucidate a novel role of lactate. We first demonstrated that daily lactate administration (equivalent to 1 g/kg of body weight) for 2 weeks suppressed CR-induced muscle atrophy by activating mammalian/mechanistic target of rapamycin (mTOR) signaling, a muscle protein synthesis pathway, and inhibited autophagy-induced muscle degradation. Next, we found that lactate administration under calorie restriction enhanced mitochondrial enzyme activity (citrate synthase and succinate dehydrogenase) and the expression of oxidative phosphorylation (OXPHOS) protein expression. Our results suggest that lactate administration under caloric restriction not only suppresses muscle atrophy but also improves mitochondrial function.
Collapse
Affiliation(s)
- Takanaga Shirai
- Graduate School of Comprehensive Human Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, 305-8574, Ibaraki, Japan.,Faculty of Health and Sport Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, 305-8577, Ibaraki, Japan.,Research Fellow of Japan Society for Promotion Science, Japan
| | - Kazuki Uemichi
- Graduate School of Comprehensive Human Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, 305-8574, Ibaraki, Japan
| | - Yuki Hidaka
- School of Physical Education, Health and Sport Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, 305-8574, Ibaraki, Japan
| | - Yu Kitaoka
- Department of Human Sciences, Kanagawa University, 3-27-1 Rokkakubashi, Kanagawa-ku, Yokohama-shi, Kanagawa, 221-8686 Japan
| | - Tohru Takemasa
- Faculty of Health and Sport Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, 305-8577, Ibaraki, Japan
| |
Collapse
|
65
|
Shirai T, Uemichi K, Kubota K, Yamauchi Y, Takemasa T. Maslinic Acid Promotes Hypertrophy Induced by Functional Overload in Mouse Skeletal Muscle. J Nutr Sci Vitaminol (Tokyo) 2021; 67:317-322. [PMID: 34719617 DOI: 10.3177/jnsv.67.317] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Nutritional supplements are sometimes important for athletes to improve their sports performance and maintain their condition. Maslinic acid (MA) is a type of compound with a pentacyclic triterpene structure extracted from olives, and has a strong anti-inflammatory effect and improves metabolic function. This study aimed to investigate the effects of MA on muscle hypertrophy by functional overload using an animal model. Mice plantaris muscles were overloaded by synergist ablation surgery with/without MA and they were sampled at 4, 7, and 14 d after the operation. We demonstrated that MA significantly increased plantaris' cross-sectional area and activated the mechanistic target of rapamycin (mTOR) signaling compared with the non-supplemented group (main effect of MA, p<0.05). In addition, MA also significantly reduced catabolic proteins compared with the non-supplemented group. MA supplementation increased muscle fiber size and promoted muscle hypertrophy via mTOR signaling. Our results indicate that MA supplementation may be useful for promoting hypertrophy of skeletal muscle.
Collapse
Affiliation(s)
- Takanaga Shirai
- Graduate School of Comprehensive Human Sciences, University of Tsukuba.,Reserch Fellow in Japan Society for Promotion Science
| | - Kazuki Uemichi
- Graduate School of Comprehensive Human Sciences, University of Tsukuba
| | - Kakeru Kubota
- School of Physical Education, Health and Sport Sciences, University of Tsukuba
| | - Yuki Yamauchi
- Tsukuba Life Science Innovation Program (T-LSI), University of Tsukuba
| | - Tohru Takemasa
- Faculty of Health and Sport Sciences, University of Tsukuba
| |
Collapse
|
66
|
Takegaki J, Sase K, Kono Y, Nakano D, Fujita T, Konishi S, Fujita S. Intramuscular injection of mesenchymal stem cells activates anabolic and catabolic systems in mouse skeletal muscle. Sci Rep 2021; 11:21224. [PMID: 34707171 PMCID: PMC8551189 DOI: 10.1038/s41598-021-00627-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Accepted: 10/01/2021] [Indexed: 01/06/2023] Open
Abstract
Skeletal muscle mass is critical for good quality of life. Mesenchymal stem cells (MSCs) are multipotent stem cells distributed across various tissues. They are characterized by the capacity to secrete growth factors and differentiate into skeletal muscle cells. These capabilities suggest that MSCs might be beneficial for muscle growth. Nevertheless, little is known regarding the effects on muscle protein anabolic and catabolic systems of intramuscular injection of MSCs into skeletal muscle. Therefore, in the present study, we measured changes in mechanistic target of rapamycin complex 1 (mTORC1) signaling, the ubiquitin–proteasome system, and autophagy-lysosome system-related factors after a single intramuscular injection of MSCs with green fluorescence protein (GFP) into mouse muscles. The intramuscularly-injected MSCs were retained in the gastrocnemius muscle for 7 days after the injection, indicated by detection of GFP and expression of platelet-derived growth factor receptor-alpha. The injection of MSCs increased the expression of satellite cell-related genes, activated mTORC1 signaling and muscle protein synthesis, and increased protein ubiquitination and autophagosome formation (indicated by the expression of microtubule-associated protein 1 light chain 3-II). These results suggest that the intramuscular injection of MSCs activated muscle anabolic and catabolic systems and accelerated muscle protein turnover.
Collapse
Affiliation(s)
- Junya Takegaki
- Research Organization of Science and Technology, Ritsumeikan University, 1-1-1 Noji-Higashi, Kusatsu, Shiga, 525-8577, Japan.,Ritsumeikan Global Innovation Research Organization, Ritsumeikan University, 1-1-1 Noji-Higashi, Kusatsu, Shiga, 525-8577, Japan
| | - Kohei Sase
- Faculty of Sport and Health Science, Ritsumeikan University, 1-1-1 Noji-Higashi, Kusatsu, Shiga, 525-8577, Japan
| | - Yusuke Kono
- Ritsumeikan Global Innovation Research Organization, Ritsumeikan University, 1-1-1 Noji-Higashi, Kusatsu, Shiga, 525-8577, Japan
| | - Daiki Nakano
- Ritsumeikan Global Innovation Research Organization, Ritsumeikan University, 1-1-1 Noji-Higashi, Kusatsu, Shiga, 525-8577, Japan
| | - Takuya Fujita
- College of Pharmaceutical Sciences, Ritsumeikan University, 1-1-1 Noji-Higashi, Kusatsu, Shiga, 525-8577, Japan
| | - Satoshi Konishi
- Faculty of Science and Engineering, Ritsumeikan University, 1-1-1 Noji-Higashi, Kusatsu, Shiga, 525-8577, Japan
| | - Satoshi Fujita
- Faculty of Sport and Health Science, Ritsumeikan University, 1-1-1 Noji-Higashi, Kusatsu, Shiga, 525-8577, Japan.
| |
Collapse
|
67
|
Kamada Y, Toyama S, Arai Y, Inoue H, Nakagawa S, Fujii Y, Kaihara K, Kishida T, Mazda O, Takahashi K. Treadmill running prevents atrophy differently in fast- versus slow-twitch muscles in a rat model of rheumatoid arthritis. J Muscle Res Cell Motil 2021; 42:429-441. [PMID: 34687403 DOI: 10.1007/s10974-021-09610-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Accepted: 10/13/2021] [Indexed: 10/20/2022]
Abstract
To investigate the effects of treadmill running on two different types of skeletal muscle, we established a rat model of collagen-induced arthritis (CIA). The skeletal muscles studied were the extensor digitorum longus (EDL), which is rich in fast-twitch muscle fibers, and the soleus, which is rich in slow-twitch muscle fibers. The histological and transcriptional changes in these muscles at 14 and 44 days after immunosensitization were compared between rats that were forced to exercise (CIA ex group) and free-reared CIA rats (CIA no group). Change in protein expression was examined on day 14 after a single bout of treadmill running. Treadmill running had different effects on the relative muscle weight and total and fiber cross-sectional areas in each muscle type. In the soleus, it prevented muscle atrophy. Transcriptional analysis revealed increased eukaryotic translation initiation factor 4E (Eif4e) expression on day 14 and increased Atrogin-1 and peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC-1α) expression on day 44 in the soleus in the CIA ex group, suggesting an interaction between muscle type and exercise. A single bout of treadmill running increased the level of Eif4e and p70S6K and decreased that of Atrogin-1 in the soleus on day 14. Treadmill running prevented muscle atrophy in the soleus in a rat model of rheumatoid arthritis via activation of mitochondrial function, as evidenced by increased PGC-1α expression.
Collapse
Affiliation(s)
- Yoichiro Kamada
- Department of Orthopaedics, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto, Japan
| | - Shogo Toyama
- Department of Orthopaedics, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto, Japan
| | - Yuji Arai
- Department of Sports and Para-Sports Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto, Japan.
| | - Hiroaki Inoue
- Department of Orthopaedics, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto, Japan
| | - Shuji Nakagawa
- Department of Sports and Para-Sports Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto, Japan
| | - Yuta Fujii
- Department of Orthopaedics, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto, Japan
| | - Kenta Kaihara
- Department of Orthopaedics, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto, Japan
| | - Tsunao Kishida
- Department of Immunology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto, Japan
| | - Osam Mazda
- Department of Immunology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto, Japan
| | - Kenji Takahashi
- Department of Orthopaedics, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto, Japan
| |
Collapse
|
68
|
Davids CJ, Næss TC, Moen M, Cumming KT, Horwath O, Psilander N, Ekblom B, Coombes JS, Peake JM, Raastad T, Roberts LA. Acute cellular and molecular responses and chronic adaptations to low-load blood flow restriction and high-load resistance exercise in trained individuals. J Appl Physiol (1985) 2021; 131:1731-1749. [PMID: 34554017 DOI: 10.1152/japplphysiol.00464.2021] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Blood flow restriction (BFR) with low-load resistance exercise (RE) is often used as a surrogate to traditional high-load RE to stimulate muscular adaptations, such as hypertrophy and strength. However, it is not clear whether such adaptations are achieved through similar cellular and molecular processes. We compared changes in muscle function, morphology and signaling pathways between these differing training protocols. Twenty-one males and females (mean ± SD: 24.3 ± 3.1 years) experienced with resistance training (4.9 ± 2.6 years) performed nine weeks of resistance training (three times per week) with either high-loads (75-80% 1RM; HL-RT), or low-loads with BFR (30-40% 1RM; LL-BFR). Before and after the training intervention, resting muscle biopsies were collected, and quadricep cross-sectional area (CSA), muscular strength and power were measured. Approximately 5 days following the intervention, the same individuals performed an additional 'acute' exercise session under the same conditions, and serial muscle biopsies were collected to assess hypertrophic- and ribosomal-based signaling stimuli. Quadricep CSA increased with both LL-BFR (7.4±4.3%) and HL-RT (4.6±2.9%), with no significant differences between training groups (p=0.37). Muscular strength also increased in both training groups, but with superior gains in squat 1RM occurring with HL-RT (p<0.01). Acute phosphorylation of several key proteins involved in hypertrophy signaling pathways, and expression of ribosomal RNA transcription factors occurred to a similar degree with LL-BFR and HL-RT (all p>0.05 for between-group comparisons). Together, these findings validate low-load resistance training with continuous BFR as an effective alternative to traditional high-load resistance training for increasing muscle hypertrophy in trained individuals.
Collapse
Affiliation(s)
- Charlie J Davids
- School of Human Movement and Nutrition Sciences, University of Queensland, Brisbane, Australia.,Queensland Academy of Sport, Nathan, Australia
| | - Tore C Næss
- Department of Physical Performance, Norwegian School of Sport Science, Oslo, Norway
| | - Maria Moen
- Department of Physical Performance, Norwegian School of Sport Science, Oslo, Norway
| | | | - Oscar Horwath
- Åstrand Laboratory, Department of Physiology, Nutrition and Biomechanics, Swedish School of Sport and Health Sciences, Stockholm, Sweden
| | - Niklas Psilander
- Åstrand Laboratory, Department of Physiology, Nutrition and Biomechanics, Swedish School of Sport and Health Sciences, Stockholm, Sweden
| | - Björn Ekblom
- Åstrand Laboratory, Department of Physiology, Nutrition and Biomechanics, Swedish School of Sport and Health Sciences, Stockholm, Sweden
| | - Jeff S Coombes
- School of Human Movement and Nutrition Sciences, University of Queensland, Brisbane, Australia
| | - Jonathan M Peake
- Queensland Academy of Sport, Nathan, Australia.,Queensland University of Technology, School of Biomedical Science, Brisbane, Australia
| | - Truls Raastad
- Department of Physical Performance, Norwegian School of Sport Science, Oslo, Norway
| | - Llion Arwyn Roberts
- School of Human Movement and Nutrition Sciences, University of Queensland, Brisbane, Australia.,Queensland Academy of Sport, Nathan, Australia.,Griffith Sports Science, School of Health Sciences and Social Work, Griffith University, Gold Coast, Australia
| |
Collapse
|
69
|
Turner DC, Gorski PP, Seaborne RA, Viggars M, Murphy M, Jarvis JC, Martin NR, Stewart CE, Sharples AP. Mechanical loading of bioengineered skeletal muscle in vitro recapitulates gene expression signatures of resistance exercise in vivo. J Cell Physiol 2021; 236:6534-6547. [PMID: 33586196 PMCID: PMC8653897 DOI: 10.1002/jcp.30328] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Revised: 01/17/2021] [Accepted: 02/03/2021] [Indexed: 11/10/2022]
Abstract
Understanding the role of mechanical loading and exercise in skeletal muscle (SkM) is paramount for delineating the molecular mechanisms that govern changes in muscle mass. However, it is unknown whether loading of bioengineered SkM in vitro adequately recapitulates the molecular responses observed after resistance exercise (RE) in vivo. To address this, the transcriptional and epigenetic (DNA methylation) responses were compared after mechanical loading in bioengineered SkM in vitro and after RE in vivo. Specifically, genes known to be upregulated/hypomethylated after RE in humans were analyzed. Ninety-three percent of these genes demonstrated similar changes in gene expression post-loading in the bioengineered muscle when compared to acute RE in humans. Furthermore, similar differences in gene expression were observed between loaded bioengineered SkM and after programmed RT in rat SkM tissue. Hypomethylation occurred for only one of the genes analysed (GRIK2) post-loading in bioengineered SkM. To further validate these findings, DNA methylation and mRNA expression of known hypomethylated and upregulated genes post-acute RE in humans were also analyzed at 0.5, 3, and 24 h post-loading in bioengineered muscle. The largest changes in gene expression occurred at 3 h, whereby 82% and 91% of genes responded similarly when compared to human and rodent SkM respectively. DNA methylation of only a small proportion of genes analyzed (TRAF1, MSN, and CTTN) significantly increased post-loading in bioengineered SkM alone. Overall, mechanical loading of bioengineered SkM in vitro recapitulates the gene expression profile of human and rodent SkM after RE in vivo. Although some genes demonstrated differential DNA methylation post-loading in bioengineered SkM, such changes across the majority of genes analyzed did not closely mimic the epigenetic response to acute-RE in humans.
Collapse
Affiliation(s)
- Daniel C. Turner
- Institute for Science and Technology in Medicine (ISTM), School of Pharmacy and BioengineeringKeele UniversityStaffordshireUK
- Stem Cells, Ageing and Molecular Physiology Unit (SCAMP), Exercise Metabolism and Adaptation Research Group (EMARG), Research Institute for Sport and Exercise Sciences (RISES)Liverpool John Moores UniversityLiverpoolUK
- Randall Centre for Cell and Molecular Biophysics, School of Basic and Medical BiosciencesKing's College LondonLondonUK
| | - Piotr P. Gorski
- Institute for Science and Technology in Medicine (ISTM), School of Pharmacy and BioengineeringKeele UniversityStaffordshireUK
- Institute for Physical PerformanceNorwegian School of Sport Sciences (NiH)OsloNorway
| | - Robert A. Seaborne
- Stem Cells, Ageing and Molecular Physiology Unit (SCAMP), Exercise Metabolism and Adaptation Research Group (EMARG), Research Institute for Sport and Exercise Sciences (RISES)Liverpool John Moores UniversityLiverpoolUK
- Center for Genomics and Child Health, Blizard Institute, Barts and the London School of Medicine and DentistryQueen Mary University of LondonLondonUK
| | - Mark Viggars
- Stem Cells, Ageing and Molecular Physiology Unit (SCAMP), Exercise Metabolism and Adaptation Research Group (EMARG), Research Institute for Sport and Exercise Sciences (RISES)Liverpool John Moores UniversityLiverpoolUK
| | - Mark Murphy
- School of Pharmacy and Biomolecular SciencesLiverpool John Moores UniversityLiverpoolUK
| | - Jonathan C. Jarvis
- Stem Cells, Ageing and Molecular Physiology Unit (SCAMP), Exercise Metabolism and Adaptation Research Group (EMARG), Research Institute for Sport and Exercise Sciences (RISES)Liverpool John Moores UniversityLiverpoolUK
| | - Neil R.W. Martin
- School of Sport, Exercise and Health SciencesLoughborough UniversityLoughboroughUK
| | - Claire E. Stewart
- Stem Cells, Ageing and Molecular Physiology Unit (SCAMP), Exercise Metabolism and Adaptation Research Group (EMARG), Research Institute for Sport and Exercise Sciences (RISES)Liverpool John Moores UniversityLiverpoolUK
| | - Adam P. Sharples
- Institute for Physical PerformanceNorwegian School of Sport Sciences (NiH)OsloNorway
| |
Collapse
|
70
|
Ahn J, Kim MJ, Yoo A, Ahn J, Ha TY, Jung CH, Seo HD, Jang YJ. Identifying Codium fragile extract components and their effects on muscle weight and exercise endurance. Food Chem 2021; 353:129463. [PMID: 33743428 DOI: 10.1016/j.foodchem.2021.129463] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 02/04/2021] [Accepted: 02/22/2021] [Indexed: 01/29/2023]
Abstract
Codium fragile (CF) is a type of green algae consumed as kimchi in Asia. UPLC-QTOF-MS/MS analysis showed that CF contain lysophosphatidyl choline, canthaxanthin, retinoic acid, α-tocopherol, and unsaturated fatty acids, which reportedly improve skeletal muscle health. However, the effect of CF on skeletal muscle mass and function remains to be elucidated. In mice fed with CF extracts, exercise endurance and muscle weight increased. CF extracts enhanced protein synthesis and myogenic differentiation through the mTORC1 pathway. CF extracts also promoted oxidative muscle fiber formation and mitochondrial biogenesis through the PGC-1α-related signaling pathway. Upregulation of PGC-1α by CF extracts was abolished by EX527 SIRT1 inhibitor treatment. Changed signaling molecules in the CF extracts were partially regulated by canthaxanthin, a new compound in CF extracts, suggesting that canthaxanthin contribute synergistically to the effect of CF extracts. Therefore, CF is a potential food source for sport nutrition or prevention of sarcopenia.
Collapse
Affiliation(s)
- Jisong Ahn
- Natural Materials and Metabolism Research Group, Korea Food Research Institute, Wanju-gun 55365, Republic of Korea; Department of Food Science and Technology, Chonbuk National University, Jeonju-si 54896, Republic of Korea
| | - Min Jung Kim
- Healthcare Research Group, Korea Food Research Institute, Wanju-gun 55365, Republic of Korea
| | - Ahyoung Yoo
- Natural Materials and Metabolism Research Group, Korea Food Research Institute, Wanju-gun 55365, Republic of Korea; Department of Food and Nutrition, Chungnam National University, Daejeon 34134, Republic of Korea
| | - Jiyun Ahn
- Natural Materials and Metabolism Research Group, Korea Food Research Institute, Wanju-gun 55365, Republic of Korea; Division of Food Biotechnology, University of Science and Technology, Daejeon 34113, Republic of Korea
| | - Tae Youl Ha
- Natural Materials and Metabolism Research Group, Korea Food Research Institute, Wanju-gun 55365, Republic of Korea; Division of Food Biotechnology, University of Science and Technology, Daejeon 34113, Republic of Korea
| | - Chang Hwa Jung
- Natural Materials and Metabolism Research Group, Korea Food Research Institute, Wanju-gun 55365, Republic of Korea; Division of Food Biotechnology, University of Science and Technology, Daejeon 34113, Republic of Korea
| | - Hyo Deok Seo
- Natural Materials and Metabolism Research Group, Korea Food Research Institute, Wanju-gun 55365, Republic of Korea
| | - Young Jin Jang
- Natural Materials and Metabolism Research Group, Korea Food Research Institute, Wanju-gun 55365, Republic of Korea; Major of Food Science & Technology, Seoul Women's University, Seoul 01797, Republic of Korea.
| |
Collapse
|
71
|
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
|
72
|
Nishimura Y, Musa I, Holm L, Lai YC. Recent advances in measuring and understanding the regulation of exercise-mediated protein degradation in skeletal muscle. Am J Physiol Cell Physiol 2021; 321:C276-C287. [PMID: 34038244 DOI: 10.1152/ajpcell.00115.2021] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Skeletal muscle protein turnover plays a crucial role in controlling muscle mass and protein quality control, including sarcomeric (structural and contractile) proteins. Protein turnover is a dynamic and continual process of protein synthesis and degradation. The ubiquitin proteasome system (UPS) is a key degradative system for protein degradation and protein quality control in skeletal muscle. UPS-mediated protein quality control is known to be impaired in aging and diseases. Exercise is a well-recognized, nonpharmacological approach to promote muscle protein turnover rates. Over the past decades, we have acquired substantial knowledge of molecular mechanisms of muscle protein synthesis after exercise. However, there have been considerable gaps in the mechanisms of how muscle protein degradation is regulated at the molecular level. The main challenge to understand muscle protein degradation is due in part to the lack of solid stable isotope tracer methodology to measure muscle protein degradation rate. Understanding the mechanisms of UPS with the concomitant measurement of protein degradation rate in skeletal muscle will help identify novel therapeutic strategies to ameliorate impaired protein turnover and protein quality control in aging and diseases. Thus, the goal of this present review was to highlight how recent advances in the field may help improve our understanding of exercise-mediated protein degradation. We discuss 1) the emerging roles of protein phosphorylation and ubiquitylation modifications in regulating proteasome-mediated protein degradation after exercise and 2) methodological advances to measure in vivo myofibrillar protein degradation rate using stable isotope tracer methods.
Collapse
Affiliation(s)
- Yusuke Nishimura
- School of Sport, Exercise and Rehabilitation Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Ibrahim Musa
- School of Sport, Exercise and Rehabilitation Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Lars Holm
- School of Sport, Exercise and Rehabilitation Sciences, University of Birmingham, Birmingham, United Kingdom
- MRC Versus Arthritis Centre for Musculoskeletal Ageing Research, University of Birmingham, Birmingham, United Kingdom
| | - Yu-Chiang Lai
- School of Sport, Exercise and Rehabilitation Sciences, University of Birmingham, Birmingham, United Kingdom
- MRC Versus Arthritis Centre for Musculoskeletal Ageing Research, University of Birmingham, Birmingham, United Kingdom
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, United Kingdom
| |
Collapse
|
73
|
Kotani T, Takegaki J, Tamura Y, Kouzaki K, Nakazato K, Ishii N. Repeated bouts of resistance exercise in rats alter mechanistic target of rapamycin complex 1 activity and ribosomal capacity but not muscle protein synthesis. Exp Physiol 2021; 106:1950-1960. [PMID: 34197668 DOI: 10.1113/ep089699] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Accepted: 06/30/2021] [Indexed: 01/03/2023]
Abstract
NEW FINDINGS What is the central question of this study? Is muscle protein synthesis (MPS) additionally activated following exercise when ribosomal capacity is increased after repeated bouts of resistance exercise (RE)? What is the main finding and its importance? Skeletal muscles with increased ribosome content through repeated RE bouts showed sufficient activation of MPS with lower mechanistic target of rapamycin complex 1 signalling. Thus, repeated bouts of RE possibly change the translational capacity and efficiency to optimize translation activation following RE. ABSTRACT Resistance exercise (RE) activates ribosome biogenesis and increases ribosome content in skeletal muscles. However, it is unclear whether the increase in ribosome content subsequently causes an increase in RE-induced activation of muscle protein synthesis (MPS). Thus, this study aimed to investigate the relationship between ribosome content and MPS after exercise using a rat RE model. Male Sprague-Dawley rats were categorized into three groups (n = 6 for each group): sedentary (SED) and RE trained with one bout (1B) or three bouts (3B). The RE stimulus was applied to the right gastrocnemius muscle by transcutaneous electrical stimulation under isoflurane anaesthesia. The 3B group underwent stimulation every other day. Our results revealed that 6 h after the last bout of RE, muscles in the 3B group showed an increase in total RNA and 18S+28S rRNA content per muscle weight compared with the SED and 1B groups. In both the 1B and 3B groups, MPS, estimated by puromycin incorporation in proteins, was higher than that in the SED group 6 h after exercise; however, no significant difference was observed between the 1B and 3B groups. In the 1B and 3B groups, phosphorylated p70S6K at Thr-389 increased, indicating mechanistic target of rapamycin complex 1 (mTORC1) activity. p70S6K phosphorylation level was lower in the 3B group than in the 1B group. Finally, protein synthesis per ribosome (indicator of translation efficiency) was lower in the 3B group than in the 1B group. Thus, three bouts of RE changed the ribosome content and mTORC1 activation, but not the degree of RE-induced global MPS activation.
Collapse
Affiliation(s)
- Takaya Kotani
- Research Institute for Sport Science, Nippon Sport Science University, Tokyo, Japan
| | - Junya Takegaki
- Ritsumeikan Global Innovation Research Organization, Ritsumeikan University, Shiga, Japan
| | - Yuki Tamura
- Research Institute for Sport Science, Nippon Sport Science University, Tokyo, Japan.,Graduate School of Health and Sport Science, Nippon Sport Science University, Tokyo, Japan.,Faculty of Sport Science, Nippon Sport Science University, Tokyo, Japan
| | - Karina Kouzaki
- Research Institute for Sport Science, Nippon Sport Science University, Tokyo, Japan.,Graduate School of Medical and Health Science, Nippon Sport Science University, Tokyo, Japan.,Faculty of Medical Science, Nippon Sport Science University, Tokyo, Japan
| | - Koichi Nakazato
- Research Institute for Sport Science, Nippon Sport Science University, Tokyo, Japan.,Graduate School of Health and Sport Science, Nippon Sport Science University, Tokyo, Japan.,Graduate School of Medical and Health Science, Nippon Sport Science University, Tokyo, Japan.,Faculty of Medical Science, Nippon Sport Science University, Tokyo, Japan
| | - Naokata Ishii
- Graduate School or Arts and Sciences, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
74
|
McKendry J, Stokes T, Mcleod JC, Phillips SM. Resistance Exercise, Aging, Disuse, and Muscle Protein Metabolism. Compr Physiol 2021; 11:2249-2278. [PMID: 34190341 DOI: 10.1002/cphy.c200029] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Skeletal muscle is the organ of locomotion, its optimal function is critical for athletic performance, and is also important for health due to its contribution to resting metabolic rate and as a site for glucose uptake and storage. Numerous endogenous and exogenous factors influence muscle mass. Much of what is currently known regarding muscle protein turnover is owed to the development and use of stable isotope tracers. Skeletal muscle mass is determined by the meal- and contraction-induced alterations of muscle protein synthesis and muscle protein breakdown. Increased loading as resistance training is the most potent nonpharmacological strategy by which skeletal muscle mass can be increased. Conversely, aging (sarcopenia) and muscle disuse lead to the development of anabolic resistance and contribute to the loss of skeletal muscle mass. Nascent omics-based technologies have significantly improved our understanding surrounding the regulation of skeletal muscle mass at the gene, transcript, and protein levels. Despite significant advances surrounding the mechanistic intricacies that underpin changes in skeletal muscle mass, these processes are complex, and more work is certainly needed. In this article, we provide an overview of the importance of skeletal muscle, describe the influence that resistance training, aging, and disuse exert on muscle protein turnover and the molecular regulatory processes that contribute to changes in muscle protein abundance. © 2021 American Physiological Society. Compr Physiol 11:2249-2278, 2021.
Collapse
Affiliation(s)
- James McKendry
- Exercise Metabolism Research Group, Department of Kinesiology, McMaster University, Hamilton, Ontario, Canada
| | - Tanner Stokes
- Exercise Metabolism Research Group, Department of Kinesiology, McMaster University, Hamilton, Ontario, Canada
| | - Jonathan C Mcleod
- Exercise Metabolism Research Group, Department of Kinesiology, McMaster University, Hamilton, Ontario, Canada
| | - Stuart M Phillips
- Exercise Metabolism Research Group, Department of Kinesiology, McMaster University, Hamilton, Ontario, Canada
| |
Collapse
|
75
|
Abstract
Since ancient times, the health benefits of regular physical activity/exercise have been recognized and the classic studies of Morris and Paffenbarger provided the epidemiological evidence in support of such an association. Cardiorespiratory fitness, often measured by maximal oxygen uptake, and habitual physical activity levels are inversely related to mortality. Thus, studies exploring the biological bases of the health benefits of exercise have largely focused on the cardiovascular system and skeletal muscle (mass and metabolism), although there is increasing evidence that multiple tissues and organ systems are influenced by regular exercise. Communication between contracting skeletal muscle and multiple organs has been implicated in exercise benefits, as indeed has other interorgan "cross-talk." The application of molecular biology techniques and "omics" approaches to questions in exercise biology has opened new lines of investigation to better understand the beneficial effects of exercise and, in so doing, inform the optimization of exercise regimens and the identification of novel therapeutic strategies to enhance health and well-being.
Collapse
Affiliation(s)
- Mark Hargreaves
- Department of Anatomy & Physiology, The University of Melbourne, Melbourne, Victoria, Australia
| |
Collapse
|
76
|
Kotani T, Takegaki J, Tamura Y, Kouzaki K, Nakazato K, Ishii N. The effect of repeated bouts of electrical stimulation-induced muscle contractions on proteolytic signaling in rat skeletal muscle. Physiol Rep 2021; 9:e14842. [PMID: 33991444 PMCID: PMC8123562 DOI: 10.14814/phy2.14842] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Accepted: 03/16/2021] [Indexed: 11/24/2022] Open
Abstract
Mechanistic target of rapamycin complex 1 (mTORC1) plays a central role in muscle protein synthesis and repeated bouts of resistance exercise (RE) blunt mTORC1 activation. However, the changes in the proteolytic signaling when recurrent RE bouts attenuate mTORC1 activation are unclear. Using a RE model of electrically stimulated rat skeletal muscle, this study aimed to clarify the effect of repeated RE bouts on acute proteolytic signaling, particularly the calpain, autophagy‐lysosome, and ubiquitin‐proteasome pathway. p70S6K and rpS6 phosphorylation, indicators of mTORC1 activity, were attenuated by repeated RE bouts. Calpain 3 protein was decreased at 6 h post‐RE in all exercised groups regardless of the bout number. Microtubule‐associated protein 1 light chain 3 beta‐II, an indicator of autophagosome formation, was increased at 3 h and repeated RE bouts increased at 6 h, post‐RE. Ubiquitinated proteins were increased following RE, but these increases were independent of the number of RE bouts. These results suggest that the magnitude of autophagosome formation was increased following RE when mTORC1 activity was attenuated with repeated bouts of RE.
Collapse
Affiliation(s)
- Takaya Kotani
- Department of Life Sciences, Graduate School of Arts and Sciences, The University of Tokyo, Tokyo, Japan.,Graduate School of Health and Sport Science, Nippon Sport Science University, Tokyo, Japan
| | - Junya Takegaki
- Ritsumeikan Global Innovation Research Organization, Ritsumeikan University, Shiga, Japan
| | - Yuki Tamura
- Graduate School of Health and Sport Science, Nippon Sport Science University, Tokyo, Japan
| | - Karina Kouzaki
- Graduate School of Health and Sport Science, Nippon Sport Science University, Tokyo, Japan
| | - Koichi Nakazato
- Graduate School of Health and Sport Science, Nippon Sport Science University, Tokyo, Japan
| | - Naokata Ishii
- Department of Life Sciences, Graduate School of Arts and Sciences, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
77
|
Saud A, Luiz RS, Leite APO, Muller CR, Visona I, Reinecke N, Silva WH, Gloria MA, Razvickas CV, Casarini DE, Schor N. Resistance exercise training ameliorates chronic kidney disease outcomes in a 5/6 nephrectomy model. Life Sci 2021; 275:119362. [PMID: 33741414 DOI: 10.1016/j.lfs.2021.119362] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Revised: 03/03/2021] [Accepted: 03/08/2021] [Indexed: 11/15/2022]
Abstract
Chronic kidney disease (CKD) is defined by decreased glomerular filtration rate (GFR) or increased albumin excretion leading to renal injury. However, exercise training is an important non-pharmacological intervention that ameliorates and protects against Diabetes Mellitus, cardiovascular disease, and CKD. AIM Our aim was to evaluate the capability of resistance exercise training (RET) to improve CKD outcomes and the contribution of the renal and muscular Akt/mTOR signaling pathway for RET beneficial effects on a CKD model. MAIN METHODS Male Wistar rats were subjected to RET, followed for 10 weeks, and randomly divided into 5 groups: Sham: Sham-operated; sedentary and nephrectomy (5/6Nx) (SNS); exercising post-5/6Nx (SNE); exercising pre-5/6Nx (ENS); exercising pre- and post-5/6Nx (ENE). The systolic blood pressure (BP) was measured. Creatinine, proteinuria, and blood urea nitrogen (BUN) were evaluated. After euthanasia Renal and muscular Akt/mTOR signaling pathways were analyzed. KEY FINDING Our study showed that the SNS presented renal injury, hypertension, weight and muscular mass loss and a higher mortality rate. SNS group also decreased renal IL-10 and increased TNF-alfa and TGF-Beta. Renal AKT, mTOR, and rpS6 pathway were increased, PTEN was decreased on SNS. And muscular Akt and mTOR were decreased on SNS. SIGNIFICANCE The RET before and after the 5/6Nx ameliorates all these parameters mentioned above, suggesting that RET is a good non-pharmacological approach to diminish complications frequently found in CKD. We also suggest that the AKT-m-TOR pathway can play an important role in these beneficial outcomes of RET on the CKD animal model.
Collapse
Affiliation(s)
- A Saud
- Departamento de Medicina, Disciplina de Nefrologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, SP, Brazil
| | - R S Luiz
- Departamento de Medicina, Disciplina de Nefrologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, SP, Brazil
| | - A P O Leite
- Departamento de Medicina, Disciplina de Nefrologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, SP, Brazil; Tulane University, Department of Physiology & Medicine, New Orleans, LA, United States
| | - C R Muller
- Department of Bioengineering, University of California San Diego, La Jolla, CA, United States
| | - I Visona
- Departamento de Patologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, SP, Brazil
| | - N Reinecke
- Departamento de Medicina, Disciplina de Nefrologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, SP, Brazil
| | - W H Silva
- Departamento de Medicina, Disciplina de Nefrologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, SP, Brazil
| | - M A Gloria
- Departamento de Medicina, Disciplina de Nefrologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, SP, Brazil
| | - C V Razvickas
- Departamento de Medicina, Disciplina de Nefrologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, SP, Brazil
| | - D E Casarini
- Departamento de Medicina, Disciplina de Nefrologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, SP, Brazil.
| | - N Schor
- Departamento de Medicina, Disciplina de Nefrologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, SP, Brazil
| |
Collapse
|
78
|
Solsona R, Pavlin L, Bernardi H, Sanchez AMJ. Molecular Regulation of Skeletal Muscle Growth and Organelle Biosynthesis: Practical Recommendations for Exercise Training. Int J Mol Sci 2021; 22:2741. [PMID: 33800501 PMCID: PMC7962973 DOI: 10.3390/ijms22052741] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 03/04/2021] [Accepted: 03/04/2021] [Indexed: 12/18/2022] Open
Abstract
The regulation of skeletal muscle mass and organelle homeostasis is dependent on the capacity of cells to produce proteins and to recycle cytosolic portions. In this investigation, the mechanisms involved in skeletal muscle mass regulation-especially those associated with proteosynthesis and with the production of new organelles-are presented. Thus, the critical roles of mammalian/mechanistic target of rapamycin complex 1 (mTORC1) pathway and its regulators are reviewed. In addition, the importance of ribosome biogenesis, satellite cells involvement, myonuclear accretion, and some major epigenetic modifications related to protein synthesis are discussed. Furthermore, several studies conducted on the topic of exercise training have recognized the central role of both endurance and resistance exercise to reorganize sarcomeric proteins and to improve the capacity of cells to build efficient organelles. The molecular mechanisms underlying these adaptations to exercise training are presented throughout this review and practical recommendations for exercise prescription are provided. A better understanding of the aforementioned cellular pathways is essential for both healthy and sick people to avoid inefficient prescriptions and to improve muscle function with emergent strategies (e.g., hypoxic training). Finally, current limitations in the literature and further perspectives, notably on epigenetic mechanisms, are provided to encourage additional investigations on this topic.
Collapse
Affiliation(s)
- Robert Solsona
- Laboratoire Interdisciplinaire Performance Santé Environnement de Montagne (LIPSEM), Faculty of Sports Sciences, University of Perpignan Via Domitia, UR 4640, 7 Avenue Pierre de Coubertin, 66120 Font-Romeu, France;
| | - Laura Pavlin
- DMEM, University of Montpellier, INRAE UMR866, 2 Place Pierre Viala, 34060 Montpellier, France; (L.P.); (H.B.)
| | - Henri Bernardi
- DMEM, University of Montpellier, INRAE UMR866, 2 Place Pierre Viala, 34060 Montpellier, France; (L.P.); (H.B.)
| | - Anthony MJ Sanchez
- Laboratoire Interdisciplinaire Performance Santé Environnement de Montagne (LIPSEM), Faculty of Sports Sciences, University of Perpignan Via Domitia, UR 4640, 7 Avenue Pierre de Coubertin, 66120 Font-Romeu, France;
| |
Collapse
|
79
|
Steinert ND, Potts GK, Wilson GM, Klamen AM, Lin KH, Hermanson JB, McNally RM, Coon JJ, Hornberger TA. Mapping of the contraction-induced phosphoproteome identifies TRIM28 as a significant regulator of skeletal muscle size and function. Cell Rep 2021; 34:108796. [PMID: 33657380 PMCID: PMC7967290 DOI: 10.1016/j.celrep.2021.108796] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Revised: 01/11/2021] [Accepted: 02/05/2021] [Indexed: 12/25/2022] Open
Abstract
Mechanical signals, such as those evoked by maximal-intensity contractions (MICs), can induce an increase in muscle mass. Rapamycin-sensitive signaling events are widely implicated in the regulation of this process; however, recent studies indicate that rapamycin-insensitive signaling events are also involved. Thus, to identify these events, we generate a map of the MIC-regulated and rapamycin-sensitive phosphoproteome. In total, we quantify more than 10,000 unique phosphorylation sites and find that more than 2,000 of these sites are significantly affected by MICs, but remarkably, only 38 of the MIC-regulated events are mediated through a rapamycin-sensitive mechanism. Further interrogation of the rapamycin-insensitive phosphorylation events identifies the S473 residue on Tripartite Motif-Containing 28 (TRIM28) as one of the most robust MIC-regulated phosphorylation sites, and extensive follow-up studies suggest that TRIM28 significantly contributes to the homeostatic regulation of muscle size and function as well as the hypertrophy that occurs in response to increased mechanical loading.
Collapse
Affiliation(s)
- Nathaniel D Steinert
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI, USA; School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI, USA
| | - Gregory K Potts
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI, USA
| | - Gary M Wilson
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI, USA
| | - Amelia M Klamen
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI, USA; School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI, USA
| | - Kuan-Hung Lin
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI, USA; School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI, USA
| | - Jake B Hermanson
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI, USA; School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI, USA
| | - Rachel M McNally
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI, USA; School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI, USA
| | - Joshua J Coon
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI, USA; Genome Center of Wisconsin, University of Wisconsin-Madison, Madison, WI, USA; Morgridge Institute for Research, Madison, WI, USA; Department of Biomolecular Chemistry, University of Wisconsin-Madison, Madison, WI, USA
| | - Troy A Hornberger
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI, USA; School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI, USA.
| |
Collapse
|
80
|
Langer HT, Mossakowski AA, Pathak S, Mascal M, Baar K. Cannabidiol Does Not Impair Anabolic Signaling Following Eccentric Contractions in Rats. Int J Sport Nutr Exerc Metab 2021; 31:93-100. [PMID: 33621949 DOI: 10.1123/ijsnem.2020-0270] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Revised: 11/17/2020] [Accepted: 11/17/2020] [Indexed: 11/18/2022]
Abstract
Cannabidiol (CBD) has proven clinical benefits in the treatment of seizures, inflammation, and pain. The recent legalization of CBD in many countries has caused increased interest in the drug as an over-the-counter treatment for athletes looking to improve recovery. However, no data on the effects of CBD on the adaptive response to exercise in muscle are available. To address this gap, we eccentrically loaded the tibialis anterior muscle of 14 rats, injected them with a vehicle (n = 7) or 100 mg/kg CBD (n = 7), and measured markers of injury, inflammation, anabolic signaling, and autophagy 18 hr later. Pro-inflammatory signaling through nuclear factor kappa B (NF-kB) (Ser536) increased with loading in both groups; however, the effect was significantly greater (36%) in the vehicle group (p < .05). Simultaneously, anabolic signaling through ribosomal protein S6 kinase beta-1 (S6K1) (Thr389) increased after eccentric contractions in both groups with no difference between vehicle and CBD (p = .66). The ribosomal protein S6 phosphorylation (240/244) increased with stimulation (p < .001) and tended to be higher in the CBD group (p = .09). The ubiquitin-binding protein p62 levels were not modulated by stimulation (p = .6), but they were 46% greater in the CBD compared with the vehicle group (p = .01). Although liver weight did not differ between the groups (p = .99) and levels of proteins associated with stress were similar, we did observe serious side effects in one animal. In conclusion, an acute dose of CBD decreased pro-inflammatory signaling in the tibialis anterior without blunting the anabolic response to exercise in rats. Future research should determine whether these effects translate to improved recovery without altering adaptation in humans.
Collapse
Affiliation(s)
| | | | | | | | - Keith Baar
- University of California
- VA Northern California Health Care System
| |
Collapse
|
81
|
Khodabukus A. Tissue-Engineered Skeletal Muscle Models to Study Muscle Function, Plasticity, and Disease. Front Physiol 2021; 12:619710. [PMID: 33716768 PMCID: PMC7952620 DOI: 10.3389/fphys.2021.619710] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Accepted: 01/25/2021] [Indexed: 12/20/2022] Open
Abstract
Skeletal muscle possesses remarkable plasticity that permits functional adaptations to a wide range of signals such as motor input, exercise, and disease. Small animal models have been pivotal in elucidating the molecular mechanisms regulating skeletal muscle adaptation and plasticity. However, these small animal models fail to accurately model human muscle disease resulting in poor clinical success of therapies. Here, we review the potential of in vitro three-dimensional tissue-engineered skeletal muscle models to study muscle function, plasticity, and disease. First, we discuss the generation and function of in vitro skeletal muscle models. We then discuss the genetic, neural, and hormonal factors regulating skeletal muscle fiber-type in vivo and the ability of current in vitro models to study muscle fiber-type regulation. We also evaluate the potential of these systems to be utilized in a patient-specific manner to accurately model and gain novel insights into diseases such as Duchenne muscular dystrophy (DMD) and volumetric muscle loss. We conclude with a discussion on future developments required for tissue-engineered skeletal muscle models to become more mature, biomimetic, and widely utilized for studying muscle physiology, disease, and clinical use.
Collapse
Affiliation(s)
- Alastair Khodabukus
- Department of Biomedical Engineering, Duke University, Durham, NC, United States
| |
Collapse
|
82
|
Dunlap KR, Steiner JL, Rossetti ML, Kimball SR, Gordon BS. A clinically relevant decrease in contractile force differentially regulates control of glucocorticoid receptor translocation in mouse skeletal muscle. J Appl Physiol (1985) 2021; 130:1052-1063. [PMID: 33600283 DOI: 10.1152/japplphysiol.01064.2020] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Muscle atrophy decreases physical function and overall health. Increased glucocorticoid production and/or use of prescription glucocorticoids can significantly induce muscle atrophy by activating the glucocorticoid receptor, thereby transcribing genes that shift protein balance in favor of net protein degradation. Although mechanical overload can blunt glucocorticoid-induced atrophy in young muscle, those affected by glucocorticoids generally have impaired force generation. It is unknown whether contractile force alters the ability of resistance exercise to mitigate glucocorticoid receptor translocation and induce a desirable shift in protein balance when glucocorticoids are elevated. In the present study, mice were subjected to a single bout of unilateral, electrically induced muscle contractions by stimulating the sciatic nerve at 100 Hz or 50 Hz frequencies to elicit high or moderate force contractions of the tibialis anterior, respectively. Dexamethasone was used to activate the glucocorticoid receptor. Dexamethasone increased glucocorticoid signaling, including nuclear translocation of the receptor, but this was mitigated only by high force contractions. The ability of high force contractions to mitigate glucocorticoid receptor translocation coincided with a contraction-mediated increase in muscle protein synthesis, which did not occur in the dexamethasone-treated mice subjected to moderate force contractions. Though moderate force contractions failed to increase protein synthesis following dexamethasone treatment, both high and moderate force contractions blunted the glucocorticoid-mediated increase in LC3 II:I marker of autophagy. Thus, these data show that force generation is important for the ability of resistance exercise to mitigate glucocorticoid receptor translocation and promote a desirable shift in protein balance when glucocorticoids are elevated.NEW & NOTEWORTHY Glucocorticoids induce significant skeletal muscle atrophy by activating the glucocorticoid receptor. Our work shows that muscle contractile force dictates glucocorticoid receptor nuclear translocation. We also show that blunting nuclear translocation by high force contractions coincides with the ability of muscle to mount an anabolic response characterized by increased muscle protein synthesis. This work further defines the therapeutic parameters of skeletal muscle contractions to blunt glucocorticoid-induced atrophy.
Collapse
Affiliation(s)
- Kirsten R Dunlap
- Department of Nutrition, Food and Exercise Science, Florida State University, Tallahassee, Florida
| | - Jennifer L Steiner
- Department of Nutrition, Food and Exercise Science, Florida State University, Tallahassee, Florida.,Institute of Sports Sciences and Medicine, Florida State University, Tallahassee, Florida
| | - Michael L Rossetti
- Department of Nutrition, Food and Exercise Science, Florida State University, Tallahassee, Florida
| | - Scot R Kimball
- Department of Cellular and Molecular Physiology, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania
| | - Bradley S Gordon
- Department of Nutrition, Food and Exercise Science, Florida State University, Tallahassee, Florida.,Institute of Sports Sciences and Medicine, Florida State University, Tallahassee, Florida
| |
Collapse
|
83
|
Suginohara T, Wakabayashi K, Ato S, Ogasawara R. Effect of 2-deoxyglucose-mediated inhibition of glycolysis on the regulation of mTOR signaling and protein synthesis before and after high-intensity muscle contraction. Metabolism 2021; 114:154419. [PMID: 33161019 DOI: 10.1016/j.metabol.2020.154419] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Revised: 10/29/2020] [Accepted: 11/02/2020] [Indexed: 02/07/2023]
Abstract
BACKGROUND Glycolysis controls mTORC1 signaling and protein synthesis. In skeletal muscle, glucose metabolism increases with both exercise/contraction intensity and volume, and therefore, high-intensity muscle contraction (HiMC) such as resistance exercise facilitates glycolysis including glucose uptake and glycogen breakdown. However, it is unknown whether glycolysis regulates HiMC-induced mTORC1 activation and increase in protein synthesis. METHODS To determine whether glycolysis regulates basal and HiMC-induced mTORC1 signaling and protein synthesis, we employed 2-deoxyglucose (2-DG) to inhibit glycolysis and isometrically contracted the gastrocnemius muscle of Sprague Dawley rats using percutaneous electrical stimulation. RESULTS Inhibition of glycolysis by 2-DG inhibited basal phosphorylation of p70S6K and 4E-BP1 (downstream targets of mTORC1) and protein synthesis (all P < 0.05) independent of AMPK phosphorylation. AMPK phosphorylation was comparably increased after HiMC at 0 h post HiMC and returned to basal levels 6 h post HiMC in both vehicle- and 2-DG-treated groups. Glycolysis inhibition attenuated muscle contraction-induced phosphorylation of 4E-BP1 at 6 h post HiMC (P < 0.05) but not p70S6K phosphorylation and protein synthesis. CONCLUSION Although glycolysis is involved in basal but not HiMC-induced muscle protein synthesis, it regulates both basal and HiMC-induced mTORC1 signaling, and may play key roles in skeletal muscle adaptation to HiMC.
Collapse
Affiliation(s)
- Takeshi Suginohara
- Department of Life Science and Applied Chemistry, Nagoya Institute of Technology, Nagoya, Japan
| | - Koki Wakabayashi
- Department of Life Science and Applied Chemistry, Nagoya Institute of Technology, Nagoya, Japan
| | - Satoru Ato
- Department of Life Science and Applied Chemistry, Nagoya Institute of Technology, Nagoya, Japan
| | - Riki Ogasawara
- Department of Life Science and Applied Chemistry, Nagoya Institute of Technology, Nagoya, Japan.
| |
Collapse
|
84
|
Rindom E, Herskind J, Blaauw B, Overgaard K, Vissing K, Paoli FV. Concomitant excitation and tension development are required for myocellular gene expression and protein synthesis in rat skeletal muscle. Acta Physiol (Oxf) 2021; 231:e13540. [PMID: 32687678 DOI: 10.1111/apha.13540] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 07/12/2020] [Accepted: 07/14/2020] [Indexed: 02/06/2023]
Abstract
AIM Loading-induced tension development is often assumed to constitute an independent cue to initiate muscle protein synthesis following resistance exercise. However, with traditional physiological models of resistance exercise, changes in loading-induced tension development also reflect changes in neural activation patterns, and direct evidence for a mechanosensitive mechanism is therefore limited. Here, we sought to examine the importance of excitation and tension development per se on initiation of signalling, gene transcription and protein synthesis in rat skeletal muscle. METHODS Isolated rat extensor digitorum longus muscles were allocated to the following interventions: (a) Excitation-induced eccentric contractions (ECC); (b) Passive stretching without excitation (PAS); (c) Excitation with inhibition of contractions (STIM + IMA ) and; (d) Excitation in combination with both inhibition of contractions and PAS (STIM + IMA + PAS). Assessment of transcriptional and translational signalling, gene transcription and acute muscle protein synthesis was compared in stimulated vs contra-lateral non-stimulated control muscle. RESULTS Protein synthesis increased solely in muscles subjected to a combination of excitation and tension development (ECC and STIM + IMA + PAS). The same pattern was true for p38 mitogen-activated protein kinase signalling for gene transcription as well as for gene transcription of immediate early genes FOS and JUN. In contrast, mechanistic target of rapamycin Complex 1 signalling for translation initiation increased in all muscles subjected to increased tension development (ECC and STIM + IMA + PAS as well as PAS). CONCLUSIONS The current study suggests that exercise-induced increases in protein synthesis as well as transcriptional signalling is dependent on the concomitant effect of excitation and tension development, whereas signalling for translation initiation is only dependent of tension development per se.
Collapse
Affiliation(s)
- Emil Rindom
- Department of Biomedicine Aarhus University Aarhus Denmark
| | - Jon Herskind
- Section for Sport Science Department of Public Health Aarhus University Aarhus Denmark
| | - Bert Blaauw
- Department of Biomedical Sciences University of Padova Padova Italy
| | - Kristian Overgaard
- Section for Sport Science Department of Public Health Aarhus University Aarhus Denmark
| | - Kristian Vissing
- Section for Sport Science Department of Public Health Aarhus University Aarhus Denmark
| | - Frank V. Paoli
- Department of Biomedicine Aarhus University Aarhus Denmark
| |
Collapse
|
85
|
Exercise as a therapy for cancer-induced muscle wasting. SPORTS MEDICINE AND HEALTH SCIENCE 2020; 2:186-194. [PMID: 35782998 PMCID: PMC9219331 DOI: 10.1016/j.smhs.2020.11.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 11/12/2020] [Accepted: 11/17/2020] [Indexed: 12/17/2022] Open
Abstract
Cancer cachexia is a progressive disorder characterized by body weight, fat, and muscle loss. Cachexia induces metabolic disruptions that can be analogous and distinct from those observed in cancer, obscuring both diagnosis and treatment options. Inflammation, hypogonadism, and physical inactivity are widely investigated as systemic mediators of cancer-induced muscle wasting. At the cellular level, dysregulation of protein turnover and energy metabolism can negatively impact muscle mass and function. Exercise is well known for its anti-inflammatory effects and potent stimulation of anabolic signaling. Emerging evidence suggests the potential for exercise to rescue muscle's sensitivity to anabolic stimuli, reduce wasting through protein synthesis modulation, myokine release, and subsequent downregulation of proteolytic factors. To date, there is no recommendation for exercise in the management of cachexia. Given its complex nature, a multimodal approach incorporating exercise offers promising potential for cancer cachexia treatment. This review's primary objective is to summarize the growing body of research examining exercise regulation of cancer cachexia. Furthermore, we will provide evidence for exercise interactions with established systemic and cellular regulators of cancer-induced muscle wasting.
Collapse
|
86
|
Strength Training Reduces Fat Accumulation and Improves Blood Lipid Profile Even in the Absence of Skeletal Muscle Hypertrophy in High-Fat Diet-Induced Obese Condition. J Obes 2020. [DOI: 10.1155/2020/8010784] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
The aim was to investigate the effect of strength training on skeletal muscle morphology and metabolic adaptations in obese rats fed with unsaturated high-fat diet (HFD). The hypothesis was that strength training induces positive metabolic adaptations in obese rats despite impaired muscle hypertrophy. Male Wistar rats (n = 58) were randomized into two groups and fed a standard diet or a high-fat diet (HFD) containing 49.2% of fat. After induction and maintenance to obesity, the rats were divided into four groups: animals distributed in sedentary control (CS), control submitted to strength training protocol (CT), obese sedentary (ObS), and obese submitted to strength training protocol (ObT). The exercise protocol consisted of 10 weeks of training on a vertical ladder (three times a week) with a load attached to the animal’s tail. At the end of 10 weeks, strength training promoted positive changes in the body composition and metabolic parameters in obese animals. Specifically, ObT animals presented a reduction of 22.6% and 14.3% in body fat and adiposity index when compared to ObS, respectively. Furthermore, these rats had lower levels of triglycerides (ObT = 23.1 ± 9.5 vs. ObS = 30.4 ± 6.9 mg/dL) and leptin (ObT = 13.2 ± 7.2 vs. ObS = 20.5 ± 4.3 ng/mL). Training (ObT and CT) induced a greater strength gain when compared with the respective control groups. In addition, the weight of the flexor hallucis longus (FHL) muscle was higher in the ObT group than in the CT group, representing an increase of 26.1%. However, training did not promote hypertrophy as observed by a similar cross-sectional area of the FHL and plantar muscles. Based on these results, high-intensity strength training promoted an improvement of body composition and metabolic profile in obese rats that were fed a high-fat diet without skeletal muscle adaptations, becoming a relevant complementary strategy for the treatment of obesity.
Collapse
|
87
|
Preserved Skeletal Muscle Mitochondrial Function, Redox State, Inflammation and Mass in Obese Mice with Chronic Heart Failure. Nutrients 2020; 12:nu12113393. [PMID: 33158222 PMCID: PMC7694273 DOI: 10.3390/nu12113393] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 10/30/2020] [Accepted: 10/31/2020] [Indexed: 12/18/2022] Open
Abstract
Background: Skeletal muscle (SM) mitochondrial dysfunction, oxidative stress, inflammation and muscle mass loss may worsen prognosis in chronic heart failure (CHF). Diet-induced obesity may also cause SM mitochondrial dysfunction as well as oxidative stress and inflammation, but obesity per se may be paradoxically associated with high SM mass and mitochondrial adenosine triphosphate (ATP) production, as well as with enhanced survival in CHF. Methods: We investigated interactions between myocardial infarction(MI)-induced CHF and diet-induced obesity (12-wk 60% vs. standard 10% fat) in modulating gastrocnemius muscle (GM) mitochondrial ATP and tissue superoxide generation, oxidized glutathione (GSSG), cytokines and insulin signalling activation in 10-wk-old mice in the following groups: lean sham-operated, lean CHF (LCHF), obese CHF (ObCHF; all n = 8). The metabolic impact of obesity per se was investigated by pair-feeding ObCHF to standard diet with stabilized excess body weight until sacrifice at wk 8 post-MI. Results: Compared to sham, LCHF had low GM mass, paralleled by low mitochondrial ATP production and high mitochondrial reative oxygen species (ROS) production, pro-oxidative redox state, pro-inflammatory cytokine changes and low insulin signaling (p < 0.05). In contrast, excess body weight in pair-fed ObCHF was associated with high GM mass, preserved mitochondrial ATP and mitochondrial ROS production, unaltered redox state, tissue cytokines and insulin signaling (p = non significant vs. Sham, p < 0.05 vs. LCHF) despite higher superoxide generation from non-mitochondrial sources. Conclusions: CHF disrupts skeletal muscle mitochondrial function in lean rodents with low ATP and high mitochondrial ROS production, associated with tissue pro-inflammatory cytokine profile, low insulin signaling and muscle mass loss. Following CHF onset, obesity per se is associated with high skeletal muscle mass and preserved tissue ATP production, mitochondrial ROS production, redox state, cytokines and insulin signaling. These paradoxical and potentially favorable obesity-associated metabolic patterns could contribute to reported obesity-induced survival advantage in CHF.
Collapse
|
88
|
MiniVStimA: A miniaturized easy to use implantable electrical stimulator for small laboratory animals. PLoS One 2020; 15:e0241638. [PMID: 33125415 PMCID: PMC7598460 DOI: 10.1371/journal.pone.0241638] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Accepted: 10/16/2020] [Indexed: 01/17/2023] Open
Abstract
According to PubMed, roughly 10% of the annually added publications are describing findings from the small animal model (mice and rats), including investigations in the field of muscle physiology and training. A subset of this research requires neural stimulation with flexible adjustments of stimulation parameters, highlighting the need for reliable implantable electrical stimulators, small enough (~1 cm3), that even mice can tolerate them without impairing their movement. The MiniVStimA is a battery-powered implant for nerve stimulation with an outer diameter of 15 mm and an encapsulated volume of 1.2 cm3 in its smallest variation. It can be pre-programmed according to the experimental protocol and controlled after implantation with a magnet. It delivers constant current charge-balanced monophasic rectangular pulses up to 2 mA and 1 ms phase width (1 kΩ load). The circuitry is optimized for small volume and energy efficiency. Due to the variation of the internal oscillator (31 kHz ± 10%), calibration measures must be implemented during the manufacturing process, which can reduce the deviation of the frequency related parameters down to ± 1%. The expected lifetime of the smaller (larger) version is 100 (480) days for stimulation with 7 Hz all day and 10 (48) days for stimulation with 100 Hz. Devices with complex stimulation patterns for nerve stimulation have been successfully used in two in-vivo studies, lasting up to nine weeks. The implant worked fully self-contained while the animal stayed in its familiar environment. External components are not required during the entire time.
Collapse
|
89
|
Counts BR, Hardee JP, Fix DK, Vanderveen BN, Montalvo RN, Carson JA. Cachexia Disrupts Diurnal Regulation of Activity, Feeding, and Muscle Mechanistic Target of Rapamycin Complex 1 in Mice. Med Sci Sports Exerc 2020; 52:577-587. [PMID: 32058469 DOI: 10.1249/mss.0000000000002166] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
INTRODUCTION Cancer cachexia is characterized by severe skeletal muscle mass loss, which is driven by decreased muscle protein synthesis and increased protein degradation. Daily physical activity and feeding behaviors exhibit diurnal fluctuations in mice that can impact the systemic environment and skeletal muscle signaling. PURPOSE We investigated the effect of cancer cachexia on the diurnal regulation of feeding, physical activity, and skeletal muscle mechanistic target of rapamycin complex 1 (mTORC1) signaling in tumor-bearing mice. We also examined the impact of increased physical activity on diurnal behaviors and skeletal muscle mTROC1 signaling in the cancer environment. METHODS Physical activity and feeding behaviors were measured for four consecutive days before sacrifice in male C57BL/6 (B6; n = 24) and Apc (MIN; n = 22) mice at 7:00 AM and 7:00 PM under ad libitum condition. A subset of B6 (n = 16) and MIN (n = 19) mice were given wheel access for 2 wk before diurnal behavior measurements. Gastrocnemius muscle protein expression was examined. RESULTS The MIN mice demonstrated altered diurnal fluctuations in feeding and activity compared with the B6. Interestingly, cachexia did not alter MIN total food intake, but dramatically reduced cage physical activity. As a measurement of mTORC1 activity, 4E-BP1 phosphorylation increased after the dark cycle in B6 and precachectic MIN mice, whereas rpS6 phosphorylation was only increased after the dark cycle in MIN mice. MIN 4E-BP1 phosphorylation at the end of the light cycle was significantly correlated with cachexia progression and reduced physical activity. Voluntary wheel running increased light cycle MIN 4E-BP1 phosphorylation and attenuated muscle mass loss. CONCLUSIONS The cancer environment can alter diurnal feeding and physical activity behaviors in tumor-bearing mice, which are linked to the progression of cachexia and muscle wasting. Furthermore, suppressed physical activity during cachexia is associated with decreased skeletal muscle mTORC1 signaling.
Collapse
Affiliation(s)
- Brittany R Counts
- Integrative Muscle Biology Laboratory, Division of Rehabilitation Sciences, College of Health Professions, University of Tennessee Health Science Center, Memphis TN
| | - Justin P Hardee
- Integrative Muscle Biology Laboratory, Department of Exercise Science, University of South Carolina, Columbia, SC
| | - Dennis K Fix
- Integrative Muscle Biology Laboratory, Department of Exercise Science, University of South Carolina, Columbia, SC
| | - Brandon N Vanderveen
- Integrative Muscle Biology Laboratory, Department of Exercise Science, University of South Carolina, Columbia, SC
| | - Ryan N Montalvo
- Integrative Muscle Biology Laboratory, Department of Exercise Science, University of South Carolina, Columbia, SC
| | - James A Carson
- Integrative Muscle Biology Laboratory, Division of Rehabilitation Sciences, College of Health Professions, University of Tennessee Health Science Center, Memphis TN
| |
Collapse
|
90
|
Verbrugge SAJ, Gehlert S, Stadhouders LEM, Jacko D, Aussieker T, M. J. de Wit G, Vogel ISP, Offringa C, Schönfelder M, Jaspers RT, Wackerhage H. PKM2 Determines Myofiber Hypertrophy In Vitro and Increases in Response to Resistance Exercise in Human Skeletal Muscle. Int J Mol Sci 2020; 21:E7062. [PMID: 32992783 PMCID: PMC7583908 DOI: 10.3390/ijms21197062] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2020] [Revised: 09/21/2020] [Accepted: 09/23/2020] [Indexed: 12/22/2022] Open
Abstract
Nearly 100 years ago, Otto Warburg investigated the metabolism of growing tissues and discovered that tumors reprogram their metabolism. It is poorly understood whether and how hypertrophying muscle, another growing tissue, reprograms its metabolism too. Here, we studied pyruvate kinase muscle (PKM), which can be spliced into two isoforms (PKM1, PKM2). This is of interest, because PKM2 redirects glycolytic flux towards biosynthetic pathways, which might contribute to muscle hypertrophy too. We first investigated whether resistance exercise changes PKM isoform expression in growing human skeletal muscle and found that PKM2 abundance increases after six weeks of resistance training, whereas PKM1 decreases. Second, we determined that Pkm2 expression is higher in fast compared to slow fiber types in rat skeletal muscle. Third, by inducing hypertrophy in differentiated C2C12 cells and by selectively silencing Pkm1 and/or Pkm2 with siRNA, we found that PKM2 limits myotube growth. We conclude that PKM2 contributes to hypertrophy in C2C12 myotubes and indicates a changed metabolic environment within hypertrophying human skeletal muscle fibers. PKM2 is preferentially expressed in fast muscle fibers and may partly contribute to the increased potential for hypertrophy in fast fibers.
Collapse
Affiliation(s)
- Sander A. J. Verbrugge
- Department for Sport and Health Sciences, Technical University of Munich, Georg-Brauchle-Ring 60/62, 80992 München/Munich, Germany; (S.A.J.V.); (M.S.)
| | - Sebastian Gehlert
- Department for the Biosciences of Sports, Institute of Sports Science, University of Hildesheim, Universitätsplatz 1, 31141 Hildesheim, Germany
- Department for Molecular and Cellular Sports Medicine, German Sport University Cologne, 50933 Cologne, Germany; (D.J.); (T.A.)
| | - Lian E. M. Stadhouders
- Laboratory for Myology, Department of Human Movement Sciences, Faculty of Behavioural and Movement Sciences, Vrije Universiteit Amsterdam, Amsterdam Movement Sciences, De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands; (L.E.M.S.); (G.M.J.d.W.); (I.S.P.V.); (C.O.)
| | - Daniel Jacko
- Department for Molecular and Cellular Sports Medicine, German Sport University Cologne, 50933 Cologne, Germany; (D.J.); (T.A.)
| | - Thorben Aussieker
- Department for Molecular and Cellular Sports Medicine, German Sport University Cologne, 50933 Cologne, Germany; (D.J.); (T.A.)
| | - Gerard M. J. de Wit
- Laboratory for Myology, Department of Human Movement Sciences, Faculty of Behavioural and Movement Sciences, Vrije Universiteit Amsterdam, Amsterdam Movement Sciences, De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands; (L.E.M.S.); (G.M.J.d.W.); (I.S.P.V.); (C.O.)
| | - Ilse S. P. Vogel
- Laboratory for Myology, Department of Human Movement Sciences, Faculty of Behavioural and Movement Sciences, Vrije Universiteit Amsterdam, Amsterdam Movement Sciences, De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands; (L.E.M.S.); (G.M.J.d.W.); (I.S.P.V.); (C.O.)
| | - Carla Offringa
- Laboratory for Myology, Department of Human Movement Sciences, Faculty of Behavioural and Movement Sciences, Vrije Universiteit Amsterdam, Amsterdam Movement Sciences, De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands; (L.E.M.S.); (G.M.J.d.W.); (I.S.P.V.); (C.O.)
| | - Martin Schönfelder
- Department for Sport and Health Sciences, Technical University of Munich, Georg-Brauchle-Ring 60/62, 80992 München/Munich, Germany; (S.A.J.V.); (M.S.)
| | - Richard T. Jaspers
- Laboratory for Myology, Department of Human Movement Sciences, Faculty of Behavioural and Movement Sciences, Vrije Universiteit Amsterdam, Amsterdam Movement Sciences, De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands; (L.E.M.S.); (G.M.J.d.W.); (I.S.P.V.); (C.O.)
| | - Henning Wackerhage
- Department for Sport and Health Sciences, Technical University of Munich, Georg-Brauchle-Ring 60/62, 80992 München/Munich, Germany; (S.A.J.V.); (M.S.)
| |
Collapse
|
91
|
Pirkmajer S, Bezjak K, Matkovič U, Dolinar K, Jiang LQ, Miš K, Gros K, Milovanova K, Pirkmajer KP, Marš T, Kapilevich L, Chibalin AV. Ouabain Suppresses IL-6/STAT3 Signaling and Promotes Cytokine Secretion in Cultured Skeletal Muscle Cells. Front Physiol 2020; 11:566584. [PMID: 33101052 PMCID: PMC7544989 DOI: 10.3389/fphys.2020.566584] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Accepted: 08/25/2020] [Indexed: 12/16/2022] Open
Abstract
The cardiotonic steroids (CTS), such as ouabain and marinobufagenin, are thought to be adrenocortical hormones secreted during exercise and the stress response. The catalytic α-subunit of Na,K-ATPase (NKA) is a CTS receptor, whose largest pool is located in skeletal muscles, indicating that muscles are a major target for CTS. Skeletal muscles contribute to adaptations to exercise by secreting interleukin-6 (IL-6) and plethora of other cytokines, which exert paracrine and endocrine effects in muscles and non-muscle tissues. Here, we determined whether ouabain, a prototypical CTS, modulates IL-6 signaling and secretion in the cultured human skeletal muscle cells. Ouabain (2.5–50 nM) suppressed the abundance of STAT3, a key transcription factor downstream of the IL-6 receptor, as well as its basal and IL-6-stimulated phosphorylation. Conversely, ouabain (50 nM) increased the phosphorylation of ERK1/2, Akt, p70S6K, and S6 ribosomal protein, indicating activation of the ERK1/2 and the Akt-mTOR pathways. Proteasome inhibitor MG-132 blocked the ouabain-induced suppression of the total STAT3, but did not prevent the dephosphorylation of STAT3. Ouabain (50 nM) suppressed hypoxia-inducible factor-1α (HIF-1α), a modulator of STAT3 signaling, but gene silencing of HIF-1α and/or its partner protein HIF-1β did not mimic effects of ouabain on the phosphorylation of STAT3. Ouabain (50 nM) failed to suppress the phosphorylation of STAT3 and HIF-1α in rat L6 skeletal muscle cells, which express the ouabain-resistant α1-subunit of NKA. We also found that ouabain (100 nM) promoted the secretion of IL-6, IL-8, GM-CSF, and TNF-α from the skeletal muscle cells of healthy subjects, and the secretion of GM-CSF from cells of subjects with the type 2 diabetes. Marinobufagenin (10 nM), another important CTS, did not alter the secretion of these cytokines. In conclusion, our study shows that ouabain suppresses the IL-6 signaling via STAT3, but promotes the secretion of IL-6 and other cytokines, which might represent a negative feedback in the IL-6/STAT3 pathway. Collectively, our results implicate a role for CTS and NKA in regulation of the IL-6 signaling and secretion in skeletal muscle.
Collapse
Affiliation(s)
- Sergej Pirkmajer
- Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Katja Bezjak
- Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Urška Matkovič
- Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Klemen Dolinar
- Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Lake Q Jiang
- Integrative Physiology, Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - Katarina Miš
- Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Katarina Gros
- Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Kseniya Milovanova
- Department of Sports and Health Tourism, Sports Physiology and Medicine, National Research Tomsk State University, Tomsk, Russia
| | - Katja Perdan Pirkmajer
- Department of Rheumatology, University Medical Centre Ljubljana, Ljubljana, Slovenia.,Department of Internal Medicine, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Tomaž Marš
- Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Leonid Kapilevich
- Department of Sports and Health Tourism, Sports Physiology and Medicine, National Research Tomsk State University, Tomsk, Russia.,Central Scientific Laboratory, Siberian State Medical University, Tomsk, Russia
| | - Alexander V Chibalin
- Integrative Physiology, Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
92
|
Solagna F, Nogara L, Dyar KA, Greulich F, Mir AA, Türk C, Bock T, Geremia A, Baraldo M, Sartori R, Farup J, Uhlenhaut H, Vissing K, Krüger M, Blaauw B. Exercise-dependent increases in protein synthesis are accompanied by chromatin modifications and increased MRTF-SRF signalling. Acta Physiol (Oxf) 2020; 230:e13496. [PMID: 32408395 PMCID: PMC7507144 DOI: 10.1111/apha.13496] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Revised: 05/06/2020] [Accepted: 05/06/2020] [Indexed: 12/13/2022]
Abstract
AIM Resistance exercise increases muscle mass over time. However, the early signalling events leading to muscle growth are not yet well-defined. Here, we aim to identify new signalling pathways important for muscle remodelling after exercise. METHODS We performed a phosphoproteomics screen after a single bout of exercise in mice. As an exercise model we used unilateral electrical stimulation in vivo and treadmill running. We analysed muscle biopsies from human subjects to verify if our findings in murine muscle also translate to exercise in humans. RESULTS We identified a new phosphorylation site on Myocardin-Related Transcription Factor B (MRTF-B), a co-activator of serum response factor (SRF). Phosphorylation of MRTF-B is required for its nuclear translocation after exercise and is accompanied by the transcription of the SRF target gene Fos. In addition, high-intensity exercise also remodels chromatin at specific SRF target gene loci through the phosphorylation of histone 3 on serine 10 in myonuclei of both mice and humans. Ablation of the MAP kinase member MSK1/2 is sufficient to prevent this histone phosphorylation, reduce induction of SRF-target genes, and prevent increases in protein synthesis after exercise. CONCLUSION Our results identify a new exercise signalling fingerprint in vivo, instrumental for exercise-induced protein synthesis and potentially muscle growth.
Collapse
Affiliation(s)
| | - Leonardo Nogara
- Venetian Institute of Molecular Medicine (VIMM) Padova Italy
- Department of Biomedical Sciences University of Padova Padova Italy
| | - Kenneth A. Dyar
- Molecular Endocrinology, Institute for Diabetes and Cancer (IDC) Helmholz Zentrum MunichHelmholtz Diabetes Center (HMGU) Munich Germany
| | - Franziska Greulich
- Molecular Endocrinology, Institute for Diabetes and Cancer (IDC) Helmholz Zentrum MunichHelmholtz Diabetes Center (HMGU) Munich Germany
| | - Ashfaq A. Mir
- Molecular Endocrinology, Institute for Diabetes and Cancer (IDC) Helmholz Zentrum MunichHelmholtz Diabetes Center (HMGU) Munich Germany
| | - Clara Türk
- Research laboratory for Biochemical Pathology Department of Clinical Medicine & Department of Biomedicine Aarhus University Aarhus Denmark
| | - Theresa Bock
- Research laboratory for Biochemical Pathology Department of Clinical Medicine & Department of Biomedicine Aarhus University Aarhus Denmark
| | - Alessia Geremia
- Venetian Institute of Molecular Medicine (VIMM) Padova Italy
- Department of Biomedical Sciences University of Padova Padova Italy
| | - Martina Baraldo
- Venetian Institute of Molecular Medicine (VIMM) Padova Italy
- Department of Biomedical Sciences University of Padova Padova Italy
| | - Roberta Sartori
- Venetian Institute of Molecular Medicine (VIMM) Padova Italy
- Department of Biomedical Sciences University of Padova Padova Italy
| | - Jean Farup
- Research laboratory for Biochemical Pathology Department of Clinical Medicine & Department of Biomedicine Aarhus University Aarhus Denmark
| | - Henriette Uhlenhaut
- Molecular Endocrinology, Institute for Diabetes and Cancer (IDC) Helmholz Zentrum MunichHelmholtz Diabetes Center (HMGU) Munich Germany
- Chair for Metabolic Programming TUM School of Life SciencesZIEL‐Institute for Food & Health Freising Germany
| | - Kristian Vissing
- Department of Public Health, Section for Sport Science Aarhus University Aarhus Denmark
| | - Marcus Krüger
- Institute for Genetics Cologne Excellence Cluster on Cellular Stress Responses in Aging‐Associated Diseases (CECAD)University of Cologne Cologne Germany
| | - Bert Blaauw
- Venetian Institute of Molecular Medicine (VIMM) Padova Italy
- Department of Biomedical Sciences University of Padova Padova Italy
| |
Collapse
|
93
|
MacDonald TL, Pattamaprapanont P, Pathak P, Fernandez N, Freitas EC, Hafida S, Mitri J, Britton SL, Koch LG, Lessard SJ. Hyperglycaemia is associated with impaired muscle signalling and aerobic adaptation to exercise. Nat Metab 2020; 2:902-917. [PMID: 32694831 PMCID: PMC8278496 DOI: 10.1038/s42255-020-0240-7] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Accepted: 06/12/2020] [Indexed: 12/11/2022]
Abstract
Increased aerobic exercise capacity, as a result of exercise training, has important health benefits. However, some individuals are resistant to improvements in exercise capacity, probably due to undetermined genetic and environmental factors. Here, we show that exercise-induced improvements in aerobic capacity are blunted and aerobic remodelling of skeletal muscle is impaired in several animal models associated with chronic hyperglycaemia. Our data point to chronic hyperglycaemia as a potential negative regulator of aerobic adaptation, in part, via glucose-mediated modifications of the extracellular matrix, impaired vascularization and aberrant mechanical signalling in muscle. We also observe low exercise capacity and enhanced c-Jun N-terminal kinase activation in response to exercise in humans with impaired glucose tolerance. Our work indicates that current shifts in dietary and metabolic health, associated with increasing incidence of hyperglycaemia, might impair muscular and organismal adaptations to exercise training, including aerobic capacity as one of its key health outcomes.
Collapse
Affiliation(s)
- Tara L MacDonald
- Research Division, Joslin Diabetes Center, Boston, MA, USA
- Harvard Medical School, Harvard University, Boston, MA, USA
- Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Pattarawan Pattamaprapanont
- Research Division, Joslin Diabetes Center, Boston, MA, USA
- Harvard Medical School, Harvard University, Boston, MA, USA
- Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Prerana Pathak
- Research Division, Joslin Diabetes Center, Boston, MA, USA
| | | | - Ellen C Freitas
- School of Physical Education and Sport, University of São Paulo, Ribeirão Preto, Brazil
| | - Samar Hafida
- Research Division, Joslin Diabetes Center, Boston, MA, USA
- Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Joanna Mitri
- Research Division, Joslin Diabetes Center, Boston, MA, USA
- Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Steven L Britton
- Department of Molecular and Integrative Physiology, and Department of Anesthesiology, University of Michigan, Ann Arbor, MI, USA
| | - Lauren G Koch
- Department of Physiology and Pharmacology, The University of Toledo, Toledo, OH, USA
| | - Sarah J Lessard
- Research Division, Joslin Diabetes Center, Boston, MA, USA.
- Harvard Medical School, Harvard University, Boston, MA, USA.
- Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA.
| |
Collapse
|
94
|
Sase K, Kido K, Ato S, Fujita S. Effect of resistance training on rat skeletal muscle during severe food restriction. TRANSLATIONAL SPORTS MEDICINE 2020. [DOI: 10.1002/tsm2.162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Affiliation(s)
- Kohei Sase
- Faculty of Sport and Health Science Ritsumeikan University Kusatsu Shiga Japan
| | - Kohei Kido
- Faculty of Sport and Health Science Ritsumeikan University Kusatsu Shiga Japan
| | - Satoru Ato
- Faculty of Sport and Health Science Ritsumeikan University Kusatsu Shiga Japan
| | - Satoshi Fujita
- Faculty of Sport and Health Science Ritsumeikan University Kusatsu Shiga Japan
| |
Collapse
|
95
|
Bodine SC. Edward F. Adolph Distinguished Lecture. Skeletal muscle atrophy: Multiple pathways leading to a common outcome. J Appl Physiol (1985) 2020; 129:272-282. [PMID: 32644910 PMCID: PMC7473948 DOI: 10.1152/japplphysiol.00381.2020] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 07/02/2020] [Accepted: 07/03/2020] [Indexed: 12/14/2022] Open
Abstract
Skeletal muscle atrophy continues to be a serious consequence of many diseases and conditions for which there is no treatment. Our understanding of the mechanisms regulating skeletal muscle mass has improved considerably over the past two decades. For many years it was known that skeletal muscle atrophy resulted from an imbalance between protein synthesis and protein breakdown, with the net balance shifting toward protein breakdown. However, the molecular and cellular mechanisms underlying the increased breakdown of myofibrils was unknown. Over the past two decades, numerous reports have identified novel genes and signaling pathways that are upregulated and activated in response to stimuli such as disuse, inflammation, metabolic stress, starvation and others that induce muscle atrophy. This review summarizes the discovery efforts performed in the identification of several pathways involved in the regulation of skeletal muscle mass: the mammalian target of rapamycin (mTORC1) and the ubiquitin proteasome pathway and the E3 ligases, MuRF1 and MAFbx. While muscle atrophy is a common outcome of many diseases, it is doubtful that a single gene or pathway initiates or mediates the breakdown of myofibrils. Interestingly, however, is the observation that upregulation of the E3 ligases, MuRF1 and MAFbx, is a common feature of many divergent atrophy conditions. The challenge for the field of muscle biology is to understand how all of the various molecules, transcription factors, and signaling pathways interact to produce muscle atrophy and to identify the critical factors for intervention.
Collapse
Affiliation(s)
- Sue C Bodine
- Department of Internal Medicine/Endocrinology and Metabolism, University of Iowa Carver College of Medicine, Iowa City, Iowa
| |
Collapse
|
96
|
Murach KA, McCarthy JJ, Peterson CA, Dungan CM. Making Mice Mighty: recent advances in translational models of load-induced muscle hypertrophy. J Appl Physiol (1985) 2020; 129:516-521. [PMID: 32673155 DOI: 10.1152/japplphysiol.00319.2020] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
The ability to genetically manipulate mice allows for gain- and loss-of-function in vivo, making them an ideal model for elucidating mechanisms of skeletal muscle mass regulation. Combining genetic models with mechanical muscle loading enables identification of specific factors involved in the hypertrophic response as well as the ability to test the requirement of those factors for adaptation, thereby informing performance and therapeutic interventions. Until recently, approaches for inducing mechanically mediated muscle hypertrophy (i.e., resistance-training analogs) have been limited and considered "nontranslatable" to humans. This mini-review outlines recent translational advances in loading-mediated strategies for inducing muscle hypertrophy in mice, and highlights the advantages and disadvantages of each method. The skeletal muscle field is poised for new breakthroughs in understanding mechanisms regulating load-induced muscle growth given the numerous murine tools that have very recently been described.
Collapse
Affiliation(s)
- Kevin A Murach
- The Center for Muscle Biology, University of Kentucky, Lexington, Kentucky.,Department of Physical Therapy, College of Health Sciences, University of Kentucky, Lexington, Kentucky
| | - John J McCarthy
- The Center for Muscle Biology, University of Kentucky, Lexington, Kentucky.,Department of Physiology, College of Medicine, University of Kentucky, Lexington, Kentucky
| | - Charlotte A Peterson
- The Center for Muscle Biology, University of Kentucky, Lexington, Kentucky.,Department of Physical Therapy, College of Health Sciences, University of Kentucky, Lexington, Kentucky.,Department of Physiology, College of Medicine, University of Kentucky, Lexington, Kentucky
| | - Cory M Dungan
- The Center for Muscle Biology, University of Kentucky, Lexington, Kentucky.,Department of Physical Therapy, College of Health Sciences, University of Kentucky, Lexington, Kentucky
| |
Collapse
|
97
|
Lawrence MM, Van Pelt DW, Confides AL, Hunt ER, Hettinger ZR, Laurin JL, Reid JJ, Peelor FF, Butterfield TA, Dupont-Versteegden EE, Miller BF. Massage as a mechanotherapy promotes skeletal muscle protein and ribosomal turnover but does not mitigate muscle atrophy during disuse in adult rats. Acta Physiol (Oxf) 2020; 229:e13460. [PMID: 32125770 PMCID: PMC7293583 DOI: 10.1111/apha.13460] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Revised: 02/25/2020] [Accepted: 02/26/2020] [Indexed: 12/12/2022]
Abstract
AIM Interventions that decrease atrophy during disuse are desperately needed to maintain muscle mass. We recently found that massage as a mechanotherapy can improve muscle regrowth following disuse atrophy. Therefore, we aimed to determine if massage has similar anabolic effects when applied during normal weight bearing conditions (WB) or during atrophy induced by hindlimb suspension (HS) in adult rats. METHODS Adult (10 months) male Fischer344-Brown Norway rats underwent either hindlimb suspension (HS, n = 8) or normal WB (WB, n = 8) for 7 days. Massage was applied using cyclic compressive loading (CCL) in WB (WBM, n = 9) or HS rats (HSM, n = 9) and included four 30-minute bouts of CCL applied to gastrocnemius muscle every other day. RESULTS Massage had no effect on any anabolic parameter measured under WB conditions (WBM). In contrast, massage during HS (HSM) stimulated protein turnover, but did not mitigate muscle atrophy. Atrophy from HS was caused by both lowered protein synthesis and higher degradation. HS and HSM had lowered total RNA compared with WB and this was the result of significantly higher ribosome degradation in HS that was attenuated in HSM, without differences in ribosomal biogenesis. Also, massage increased protein turnover in the non-massaged contralateral limb during HS. Finally, we determined that total RNA degradation primarily dictates loss of muscle ribosomal content during disuse atrophy. CONCLUSION We conclude that massage is an effective mechanotherapy to impact protein turnover during muscle disuse in both the massaged and non-massaged contralateral muscle, but it does not attenuate the loss of muscle mass.
Collapse
Affiliation(s)
- Marcus M. Lawrence
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA
| | - Douglas W. Van Pelt
- Department of Physical Therapy, University of Kentucky, Lexington, KY 40536, USA
- Center for Muscle Biology, University of Kentucky, Lexington, KY 40536, USA
| | - Amy L. Confides
- Department of Physical Therapy, University of Kentucky, Lexington, KY 40536, USA
- Center for Muscle Biology, University of Kentucky, Lexington, KY 40536, USA
| | - Emily R. Hunt
- Department of Physical Therapy, University of Kentucky, Lexington, KY 40536, USA
- Center for Muscle Biology, University of Kentucky, Lexington, KY 40536, USA
| | - Zachary R. Hettinger
- Department of Physical Therapy, University of Kentucky, Lexington, KY 40536, USA
- Center for Muscle Biology, University of Kentucky, Lexington, KY 40536, USA
| | - Jaime L. Laurin
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA
| | - Justin J. Reid
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA
| | - Frederick F. Peelor
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA
| | - Timothy A. Butterfield
- Center for Muscle Biology, University of Kentucky, Lexington, KY 40536, USA
- Department of Athletic Training and Clinical Nutrition, University of Kentucky, Lexington, KY 40536, USA
| | - Esther E. Dupont-Versteegden
- Department of Physical Therapy, University of Kentucky, Lexington, KY 40536, USA
- Center for Muscle Biology, University of Kentucky, Lexington, KY 40536, USA
| | - Benjamin F. Miller
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA
| |
Collapse
|
98
|
Asrani K, Murali S, Lam B, Na CH, Phatak P, Sood A, Kaur H, Khan Z, Noë M, Anchoori RK, Talbot CC, Smith B, Skaro M, Lotan TL. mTORC1 feedback to AKT modulates lysosomal biogenesis through MiT/TFE regulation. J Clin Invest 2020; 129:5584-5599. [PMID: 31527310 DOI: 10.1172/jci128287] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Accepted: 09/10/2019] [Indexed: 01/02/2023] Open
Abstract
The microphthalmia family of transcription factors (MiT/TFEs) controls lysosomal biogenesis and is negatively regulated by the nutrient sensor mTORC1. However, the mechanisms by which cells with constitutive mTORC1 signaling maintain lysosomal catabolism remain to be elucidated. Using the murine epidermis as a model system, we found that epidermal Tsc1 deletion resulted in a phenotype characterized by wavy hair and curly whiskers, and was associated with increased EGFR and HER2 degradation. Unexpectedly, constitutive mTORC1 activation with Tsc1 loss increased lysosomal content via upregulated expression and activity of MiT/TFEs, whereas genetic deletion of Rheb or Rptor or prolonged pharmacologic mTORC1 inactivation had the reverse effect. This paradoxical increase in lysosomal biogenesis by mTORC1 was mediated by feedback inhibition of AKT, and a resulting suppression of AKT-induced MiT/TFE downregulation. Thus, inhibiting hyperactive AKT signaling in the context of mTORC1 loss-of-function fully restored MiT/TFE expression and activity. These data suggest that signaling feedback loops work to restrain or maintain cellular lysosomal content during chronically inhibited or constitutively active mTORC1 signaling, respectively, and reveal a mechanism by which mTORC1 regulates upstream receptor tyrosine kinase signaling.
Collapse
Affiliation(s)
| | | | | | - Chan-Hyun Na
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Pornima Phatak
- Baltimore Veteran Affairs Medical Center, Baltimore, Maryland, USA
| | | | | | | | | | | | | | - Barbara Smith
- Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | | | | |
Collapse
|
99
|
Damiani APL, Caldas LC, Melo AB, Contreiro CDE, Estevam WM, Nogueira BV, Ferreira LG, Leopoldo AS, Leopoldo APL. RESISTANCE TRANING PROTOCOLS PROMOTE STRENGTH INCREASE WITHOUT MORPHOLOGICAL CHANGES. REV BRAS MED ESPORTE 2020. [DOI: 10.1590/1517-869220202603209955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
ABSTRACT Introduction Resistance training (RT) has been related to increased protein synthesis, and in the myocardium it triggers morphological adaptations that result in improved cardiac contractility. In skeletal muscle, RT promotes an improvement in functional capacity and in sarcopenia caused by aging. However, the efficacy of this training method in the cardiac and skeletal systems has not yet been clarified. Objective To investigate the effect of different vertical ladder RT protocols on cardiac and skeletal structure and morphology. Materials and Methods: Wistar rats (n = 28) were randomized into four groups: sedentary (C); RT protocol with 4 to 9 climbs, 3 sessions/week, 120 second interval and intensity of 50% to 100% of the maximum load (ML) with progressive addition of 30 g (RT1); RT protocol with 4 to 5 climbs, 3 sessions/week, 60 second interval and intensity of 50% to 100% of the ML, where a 30 g overload was added in the 5th climb (RT2); RT protocol with 4 to 5 climbs, 5 sessions/week, 60 second interval and intensity of 50% to 100% of the ML; the animals that completed the 4th climb underwent the 5th climb with 100% ML plus 30 g (RT3). RT protocols were performed for 9 weeks with a duration of 30 to 45 minutes/day. The nutritional profile and cardiac/skeletal muscle morphology were evaluated along with the cross sectional area and collagen fraction. Results RT did not promote adaptations in cardiac and musculoskeletal structure and morphology, nor was it able to reduce body weight and body fat deposits. However, RT brought about an increase in absolute and relative strength. Conclusion Vertical ladder RT protocols, regardless of weekly frequency, lead to increased muscle strength without cardiac and skeletal structural adaptations. Level of evidence I, Therapeutic studies - Investigating treatment results.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - André Soares Leopoldo
- Universidade Federal do Espírito Santo, Brazil; Universidade Federal do Espírito Santo, Brazil
| | - Ana Paula Lima Leopoldo
- Universidade Federal do Espírito Santo, Brazil; Universidade Federal do Espírito Santo, Brazil
| |
Collapse
|
100
|
Enhanced skeletal muscle insulin sensitivity after acute resistance-type exercise is upregulated by rapamycin-sensitive mTOR complex 1 inhibition. Sci Rep 2020; 10:8509. [PMID: 32444657 PMCID: PMC7244536 DOI: 10.1038/s41598-020-65397-z] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Accepted: 04/13/2020] [Indexed: 01/07/2023] Open
Abstract
Acute aerobic exercise (AE) increases skeletal muscle insulin sensitivity for several hours, caused by acute activation of AMP-activated protein kinase (AMPK). Acute resistance exercise (RE) also activates AMPK, possibly improving insulin-stimulated glucose uptake. However, RE-induced rapamycin-sensitive mechanistic target of rapamycin complex 1 (mTORC1) activation is higher and has a longer duration than after AE. In molecular studies, mTORC1 was shown to be upstream of insulin receptor substrate 1 (IRS-1) Ser phosphorylation residue, inducing insulin resistance. Therefore, we hypothesised that although RE increases insulin sensitivity through AMPK activation, prolonged mTORC1 activation after RE reduces RE-induced insulin sensitising effect. In this study, we used an electrical stimulation-induced RE model in rats, with rapamycin as an inhibitor of mTORC1 activation. Our results showed that RE increased insulin-stimulated glucose uptake following AMPK signal activation. However, mTORC1 activation and IRS-1 Ser632/635 and Ser612 phosphorylation were elevated 6 h after RE, with concomitant impairment of insulin-stimulated Akt signal activation. By contrast, rapamycin inhibited these prior exercise responses. Furthermore, increases in insulin-stimulated skeletal muscle glucose uptake 6 h after RE were higher in rats with rapamycin treatment than with placebo treatment. Our data suggest that mTORC1/IRS-1 signaling inhibition enhances skeletal muscle insulin-sensitising effect of RE.
Collapse
|