1
|
Paul S, Donath L, Hoppstädter J, Hecksteden A. Resistance but not endurance training suppresses glucocorticoid-induced leucine zipper (GILZ) expression in human skeletal muscle. Eur J Appl Physiol 2025; 125:1023-1036. [PMID: 39499305 DOI: 10.1007/s00421-024-05644-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Accepted: 10/14/2024] [Indexed: 11/07/2024]
Abstract
PURPOSE Within human skeletal muscle, statin treatment leads to elevated levels of the glucocorticoid-induced leucine zipper (GILZ). Further, GILZ mediates the muscle-related side effects of statins. Physical exercise leads to GILZ suppression, in a mechanosensitive manner. Given that statin treatment is rarely tolerated by habitually exercising individuals due to statin-associated muscle symptoms (SAMS), it appears that the opposing regulation of GILZ facilitates this detrimental interaction of two key measures of cardiovascular prevention, specifically for exercise modalities with high muscle strain. Similarly, opposing regulation of atrophy associated genes (atrogenes) may be a further mechanism. If confirmed, these results might have implications for the exercise prescription of statin-users. METHODS A systematic search of the Gene Expression Omnibus (GEO) repository for studies reporting the acute effects of either endurance (END), conventional resistance (RT), or eccentric resistance training (ECC) was conducted. GILZ, as well as the expression of pivotal atrogenes (e.g., muscle atrophy F-box, cathepsin L, etc.) were quantified. RESULTS 15 studies with 204 participants (22 females; 182 males) were included. RT resulted in the highest GILZ suppression, significantly differing from the expressional change after END ( - 0.46 ± 1.11 vs. - 0.07 ± 1.08), but not from ECC ( - 0.46 ± 1.11 vs. - 0.46 ± 0.95). Similar results were seen for various atrogenes. CONCLUSION Our results strengthen the assumption that mechanical loading can be considered a key mediator of exercise-induced changes in GILZ and atrogene expression.
Collapse
Affiliation(s)
- Sebastian Paul
- Department of Training Intervention Research, German Sport University Cologne, 50933, Cologne, Germany.
- Institute of Physiology, Medical University of Innsbruck, 6020, Innsbruck, Austria.
| | - Lars Donath
- Department of Training Intervention Research, German Sport University Cologne, 50933, Cologne, Germany
| | - Jessica Hoppstädter
- Department of Pharmacy, Pharmaceutical Biology, Saarland University, 66123, Saarbrücken, Germany
| | - Anne Hecksteden
- Institute of Sport Science, Universität of Innsbruck, 6020, Innsbruck, Austria
- Institute of Physiology, Medical University of Innsbruck, 6020, Innsbruck, Austria
| |
Collapse
|
2
|
Bartsch A, Sherman SL, Tramer J, Vel MS, Fredericson M. Preserving Knee Health and Delivering Specialized Care for Active Older Athletes. Sports Health 2025:19417381251326527. [PMID: 40145656 PMCID: PMC11951129 DOI: 10.1177/19417381251326527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/28/2025] Open
Abstract
CONTEXT Athletes differ from recreational exercisers in many characteristics and often require tailored treatments uniquely adapted to their situations and requirements. This practice is highlighted in young and middle-aged high-performance athletes. However, with advancing age and declining physical performance, age often outweighs athleticism, discounting the existing distinctions. This review focuses on physiological age-related processes in active older athletes and common knee conditions and elucidates the differences in preventing and treating knee injuries from the active adult population. EVIDENCE ACQUISITION Nonsystematic review with critical appraisal of existing literature. STUDY DESIGN Clinical review. LEVEL OF EVIDENCE Level 4. RESULTS Nonsteroidal anti-inflammatory drugs may interfere with the muscle hypertrophy mechanism in older athletes and it may be beneficial to adapt to other pharmacological interventions for knee osteoarthritis (OA). Arthroplasty is not typically compatible with high level sports activities; anterior cruciate ligament reconstruction surgery in the older athlete may be an effective option to improve function and enable return to sport, especially in the absence of OA. Chronic degenerative meniscal injuries can usually be treated conservatively, regardless of subjective mechanical symptoms. Acute traumatic meniscal tears in nonarthritic knees that cause effusions or reproducible mechanical symptoms may yet be considered for repair at any age. Conservative options are more dominant for patella tendinopathy, where platelet-rich plasma may be more effective than the classic extracorporeal shockwave therapy. CONCLUSION With the increase of the active older athletic population, prevention and injury treatment strategies must be balanced and tailored to their individual needs. Older athletes have various goals and demands in their respective sports, necessitating distinct prevention and treatment strategies.Strength of Recommendation Taxonomy (SORT):B.
Collapse
Affiliation(s)
- Anna Bartsch
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Redwood City, California
- Department of Physical Medicine and Rehabilitation, Stanford University School of Medicine, Redwood City, California
- Department of Orthopedics and Trauma Surgery, University Hospital Basel, Basel, Switzerland
| | - Seth Lawrence Sherman
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Redwood City, California
| | - Joseph Tramer
- Department of Orthopaedic Surgery, Cleveland Clinic Foundation, Garfield Heights, Ohio
| | - Monica Sri Vel
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Redwood City, California
| | - Michael Fredericson
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Redwood City, California
- Department of Physical Medicine and Rehabilitation, Stanford University School of Medicine, Redwood City, California
| |
Collapse
|
3
|
Voskoboynik Y, McCulloch AD, Sahoo D. Macrophages on the run: Exercise balances macrophage polarization for improved health. Mol Metab 2024; 90:102058. [PMID: 39476967 PMCID: PMC11585839 DOI: 10.1016/j.molmet.2024.102058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 10/11/2024] [Accepted: 10/24/2024] [Indexed: 11/09/2024] Open
Abstract
OBJECTIVE Exercise plays a crucial role in maintaining and improving human health. However, the precise molecular mechanisms that govern the body's response to exercise or/compared to periods of inactivity remain elusive. Current evidence appears to suggest that exercise exerts a seemingly dual influence on macrophage polarization states, inducing both pro-immune response M1 activation and cell-repair-focused M2 activation. To reconcile this apparent paradox, we leveraged a comprehensive meta-analysis of 75 diverse exercise and immobilization published datasets (7000+ samples), encompassing various exercise modalities, sampling techniques, and species. METHODS 75 exercise and immobilization expression datasets were identified and processed for analysis. The data was analyzed using boolean relationships which uses binary gene expression relationships in order to increase the signal to noise achieved from the data, allowing for the use of comparison across such a diverse set of datasets. We utilized a boolean relationship-aided macrophage gene model [1], to model the macrophage polarization state in pre and post exercise samples in both immediate exercise and long term training. RESULTS Our modeling uncovered a key temporal dynamic: exercise triggers an immediate M1 surge, while long term training transitions to sustained M2 activation. These patterns were consistent across different species (human vs mouse), sampling methods (blood vs muscle biopsy), and exercise type (resistance vs endurance), and routinely showed statistically significant results. Immobilization was shown to have the opposite effect of exercise by triggering an immediate M2 activation. Individual characteristics like gender, exercise intensity and age were found to impact the degree of polarization without changing the overall patterns. To model macrophages within the specific context of muscle tissue, we identified a focused gene set signature of muscle resident macrophage polarization, allowing for the precise measurement of macrophage activity in response to exercise within the muscle. CONCLUSIONS These consistent patterns across all 75 examined studies suggest that the long term health benefits of exercise stem from its ability to orchestrate a balanced and temporally-regulated interplay between pro-immune response (M1) and reparative macrophage activity (M2). Similarly, it suggests that an imbalance between pro-immune and cell repair responses could facilitate disease development. Our findings shed light on the intricate molecular choreography behind exercise-induced health benefits with a particular insight on its effect on the macrophages within the muscle.
Collapse
Affiliation(s)
- Yotam Voskoboynik
- Department of Bioinformatics and System Biology, Jacobs School of Engineering, University of California San Diego, San Diego, United States
| | - Andrew D McCulloch
- Department of Bioengineering, University of California San Diego, United States; Department of Medicine, University of California San Diego, United States
| | - Debashis Sahoo
- Department of Pediatrics, University of California San Diego, United States; Department of Computer Science and Engineering, Jacob's School of Engineering, University of California San Diego, United States.
| |
Collapse
|
4
|
Voss AC, Chambers TL, Gries KJ, Jemiolo B, Raue U, Minchev K, Begue G, Lee GA, Trappe TA, Trappe SW. Exercise microdosing for skeletal muscle health applications to spaceflight. J Appl Physiol (1985) 2024; 136:1040-1052. [PMID: 38205550 PMCID: PMC11365549 DOI: 10.1152/japplphysiol.00491.2023] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 12/21/2023] [Accepted: 01/03/2024] [Indexed: 01/12/2024] Open
Abstract
Findings from a recent 70-day bedrest investigation suggested intermittent exercise testing in the control group may have served as a partial countermeasure for skeletal muscle size, function, and fiber-type shifts. The purpose of the current study was to investigate the metabolic and skeletal muscle molecular responses to the testing protocols. Eight males (29 ± 2 yr) completed muscle power (6 × 4 s; peak muscle power: 1,369 ± 86 W) and V̇o2max (13 ± 1 min; 3.2 ± 0.2 L/min) tests on specially designed supine cycle ergometers during two separate trials. Blood catecholamines and lactate were measured pre-, immediately post-, and 4-h postexercise. Muscle homogenate and muscle fiber-type-specific [myosin heavy chain (MHC) I and MHC IIa] mRNA levels of exercise markers (myostatin, IκBα, myogenin, MuRF-1, ABRA, RRAD, Fn14, PDK4) and MHC I, IIa, and IIx were measured from vastus lateralis muscle biopsies obtained pre- and 4-h postexercise. The muscle power test altered (P ≤ 0.05) norepinephrine (+124%), epinephrine (+145%), lactate (+300%), and muscle homogenate mRNA (IκBα, myogenin, MuRF-1, RRAD, Fn14). The V̇o2max test altered (P ≤ 0.05) norepinephrine (+1,394%), epinephrine (+1,412%), lactate (+736%), and muscle homogenate mRNA (myostatin, IκBα, myogenin, MuRF-1, ABRA, RRAD, Fn14, PDK4). In general, both tests influenced MHC IIa muscle fibers more than MHC I with respect to the number of genes that responded and the magnitude of response. Both tests also influenced MHC mRNA expression in a muscle fiber-type-specific manner. These findings provide unique insights into the adaptive response of skeletal muscle to small doses of exercise and could help shape exercise dosing for astronauts and Earth-based individuals.NEW & NOTEWORTHY Declines in skeletal muscle health are a concern for astronauts on long-duration spaceflights. The current findings add to the growing body of exercise countermeasures data, suggesting that small doses of specific exercise can be beneficial for certain aspects of skeletal muscle health. This information can be used in conjunction with other components of existing exercise programs for astronauts and might translate to other areas focused on skeletal muscle health (e.g., sports medicine, rehabilitation, aging).
Collapse
Affiliation(s)
- Adam C Voss
- Human Performance Laboratory, Ball State University, Muncie, Indiana, United States
| | - Toby L Chambers
- Human Performance Laboratory, Ball State University, Muncie, Indiana, United States
| | - Kevin J Gries
- Human Performance Laboratory, Ball State University, Muncie, Indiana, United States
| | - Bozena Jemiolo
- Human Performance Laboratory, Ball State University, Muncie, Indiana, United States
| | - Ulrika Raue
- Human Performance Laboratory, Ball State University, Muncie, Indiana, United States
| | - Kiril Minchev
- Human Performance Laboratory, Ball State University, Muncie, Indiana, United States
| | - Gwenaelle Begue
- Human Performance Laboratory, Ball State University, Muncie, Indiana, United States
| | - Gary A Lee
- Human Performance Laboratory, Ball State University, Muncie, Indiana, United States
| | - Todd A Trappe
- Human Performance Laboratory, Ball State University, Muncie, Indiana, United States
| | - Scott W Trappe
- Human Performance Laboratory, Ball State University, Muncie, Indiana, United States
| |
Collapse
|
5
|
Baumann CW, Deane CS, Etheridge T, Szewczyk NJ, Willis CRG, Lowe DA. Adaptability to eccentric exercise training is diminished with age in female mice. J Appl Physiol (1985) 2023; 135:1135-1145. [PMID: 37823203 PMCID: PMC10979833 DOI: 10.1152/japplphysiol.00428.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 10/02/2023] [Accepted: 10/03/2023] [Indexed: 10/13/2023] Open
Abstract
The ability of skeletal muscle to adapt to eccentric contractions has been suggested to be blunted in older muscle. If eccentric exercise is to be a safe and efficient training mode for older adults, preclinical studies need to establish if older muscle can effectively adapt and if not, determine the molecular signatures that are causing this impairment. The purpose of this study was to quantify the extent age impacts functional adaptations of muscle and identify genetic signatures associated with adaptation (or lack thereof). The anterior crural muscles of young (4 mo) and older (28 mo) female mice performed repeated bouts of eccentric contractions in vivo (50 contractions/wk for 5 wk) and isometric torque was measured across the initial and final bouts. Transcriptomics was completed by RNA-sequencing 1 wk following the fifth bout to identify common and differentially regulated genes. When torques post eccentric contractions were compared after the first and fifth bouts, young muscle exhibited a robust ability to adapt, increasing isometric torque 20%-36%, whereas isometric torque of older muscle decreased up to 18% (P ≤ 0.047). Using differential gene expression, young and older muscles shared some common transcriptional changes in response to eccentric exercise training, whereas other transcripts appeared to be age dependent. That is, the ability to express particular genes after repeated bouts of eccentric contractions was not the same between ages. These molecular signatures may reveal, in part, why older muscles do not appear to be as adaptive to exercise training as young muscles.NEW & NOTEWORTHY The ability to adapt to exercise training may help prevent and combat sarcopenia. Here, we demonstrate young mouse muscles get stronger whereas older mouse muscles become weaker after repeated bouts of eccentric contractions, and that numerous genes were differentially expressed between age groups following training. These results highlight that molecular and functional plasticity is not fixed in skeletal muscle with advancing age, and the ability to handle or cope with physical stress may be impaired.
Collapse
Affiliation(s)
- Cory W Baumann
- Ohio Musculoskeletal and Neurological Institute (OMNI), Ohio University, Athens, Ohio, United States
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, Ohio, United States
| | - Colleen S Deane
- Faculty of Life Sciences, Department of Public Health and Sport Sciences, University of Exeter, Exeter, United Kingdom
- Faculty of Medicine, Department of Human Development & Health, University of Southampton, Southampton General Hospital, Southampton, United Kingdom
| | - Timothy Etheridge
- Faculty of Life Sciences, Department of Public Health and Sport Sciences, University of Exeter, Exeter, United Kingdom
| | - Nathaniel J Szewczyk
- Ohio Musculoskeletal and Neurological Institute (OMNI), Ohio University, Athens, Ohio, United States
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, Ohio, United States
| | - Craig R G Willis
- Ohio Musculoskeletal and Neurological Institute (OMNI), Ohio University, Athens, Ohio, United States
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, Ohio, United States
- Faculty of Life Sciences, School of Chemistry and Biosciences, University of Bradford, Bradford, United Kingdom
| | - Dawn A Lowe
- Divisions of Rehabilitation Science and Physical Therapy, Department of Rehabilitation Medicine, Medical School, University of Minnesota, Minneapolis, Minnesota, United States
| |
Collapse
|
6
|
Lee CJ, Nicoll JX. Time Course Evaluation of Mitogen-Activated Protein Kinase Phosphorylation to Resistance Exercise: A Systematic Review. J Strength Cond Res 2023; 37:710-725. [PMID: 36727997 DOI: 10.1519/jsc.0000000000004409] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
ABSTRACT Lee, CJ and Nicoll, JX. Time course evaluation of mitogen-activated protein kinase phosphorylation to resistance exercise: a systematic review. J Strength Cond Res 37(3): 710-725, 2023-Resistance exercise (RE) can increase the signaling activities of mitogen-activated protein kinases (MAPKs), specifically extracellular signal-regulated kinases 1/2 (ERK1/2), p90 ribosomal S6 kinases (p90RSK), c-Jun NH2-terminal kinases (JNK), and p38-MAPK. These RE-induced responses contribute to various intracellular processes modulating growth and development in skeletal muscles, playing an essential role in resistance training adaptations. The time course of MAPK phosphorylation to different RE conditions, such as training experience and varying loads, remains ambiguous. A systematic review was conducted to determine the effects of different post-RE recovery time points on the MAPK signaling cascade. In addition, the effects of loading and training statuses on MAPK responses were also investigated. The review was performed according to the preferred reporting items for systematic reviews and meta-analyses guidelines with a literature search incorporating 3 electronic databases. A modified version of the Downs and Black checklist was used to evaluate the methodological quality of the studies. The signaling responses were measured within a time range between immediately post-RE and >6 hours post-RE. Forty-four studies met the inclusion criteria, and all were classified as good-to-moderate methodological quality. Mitogen-activated protein kinase phosphorylation increased to different levels after RE, with the highest near the cessation of exercise. Although overall signaling was attenuated among trained individuals likely because of training adaptations, greater MAPK responses can be attributed to moderate loads of 65-85% 1RM regardless of the training experience. However, specific training-induced responses remain equivocal, and further investigations are required to determine the ideal training parameters to optimize anabolic intramuscular signaling, which may likely optimize resistance training adaptations.
Collapse
Affiliation(s)
- Christopher J Lee
- Department of Kinesiology, California State University, Northridge, Northridge, California
| | | |
Collapse
|
7
|
Lee B, Kim SK, Shin YJ, Son YH, Yang JW, Lee SM, Yang YR, Lee KP, Kwon KS. Genome-wide analysis of a cellular exercise model based on electrical pulse stimulation. Sci Rep 2022; 12:21251. [PMID: 36481702 PMCID: PMC9731977 DOI: 10.1038/s41598-022-25758-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Accepted: 12/05/2022] [Indexed: 12/13/2022] Open
Abstract
Skeletal muscle communicates with other organs via myokines, which are secreted by muscle during exercise and exert various effects. Despite much investigation of the exercise, the underlying molecular mechanisms are still not fully understood. Here, we applied an in vitro exercise model in which cultured C2C12 myotubes were subjected to electrical pulse stimulation (EPS), which mimics contracting muscle. Based on the significantly up- and down-regulated genes in EPS, we constructed an in silico model to predict exercise responses at the transcriptional level. The in silico model revealed similarities in the transcriptomes of the EPS and exercised animals. Comparative analysis of the EPS data and exercised mouse muscle identified putative biomarkers in exercise signaling pathways and enabled to discover novel exercise-induced myokines. Biochemical analysis of selected exercise signature genes in muscle from exercised mice showed that EPS mimics in vivo exercise, at least in part, at the transcriptional level. Consequently, we provide a novel myokine, Amphiregulin (AREG), up-regulated both in vitro and in vivo, that would be a potential target for exercise mimetics.
Collapse
Affiliation(s)
- Bora Lee
- grid.249967.70000 0004 0636 3099Aging Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141 Republic of Korea
| | - Seon Kyu Kim
- grid.249967.70000 0004 0636 3099Personalized Genomic Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141 Republic of Korea ,grid.412786.e0000 0004 1791 8264Department of Bioinformatics, KRIBB School of Bioscience, Korea University of Science and Technology (UST), Daejeon, 34113 Republic of Korea
| | - Yeo Jin Shin
- grid.249967.70000 0004 0636 3099Aging Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141 Republic of Korea
| | - Young Hoon Son
- grid.249967.70000 0004 0636 3099Aging Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141 Republic of Korea
| | - Jae Won Yang
- grid.249967.70000 0004 0636 3099Aging Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141 Republic of Korea ,grid.412786.e0000 0004 1791 8264Department of Biomolecular Science, KRIBB School of Bioscience, Korea University of Science and Technology (UST), Daejeon, 34113 Republic of Korea
| | - Seung-Min Lee
- grid.249967.70000 0004 0636 3099Aging Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141 Republic of Korea
| | - Yong Ryul Yang
- grid.249967.70000 0004 0636 3099Aging Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141 Republic of Korea
| | - Kwang-Pyo Lee
- grid.249967.70000 0004 0636 3099Aging Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141 Republic of Korea ,grid.412786.e0000 0004 1791 8264Department of Biomolecular Science, KRIBB School of Bioscience, Korea University of Science and Technology (UST), Daejeon, 34113 Republic of Korea ,Aventi Inc., Daejeon, 34141 Republic of Korea
| | - Ki-Sun Kwon
- grid.249967.70000 0004 0636 3099Aging Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141 Republic of Korea ,grid.412786.e0000 0004 1791 8264Department of Functional Genomics, KRIBB School of Bioscience, Korea University of Science and Technology (UST), Daejeon, 34113 Republic of Korea ,Aventi Inc., Daejeon, 34141 Republic of Korea
| |
Collapse
|
8
|
Orozco CA, González-Giraldo Y, Bonilla DA, Forero DA. An in silico analysis of genome-wide expression profiles of the effects of exhaustive exercise identifies heat shock proteins as the key players. Meta Gene 2022. [DOI: 10.1016/j.mgene.2022.101012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
|
9
|
Felipe SMDS, de Freitas RM, Penha EDDS, Pacheco C, Martins DL, Alves JO, Soares PM, Loureiro ACC, Lima T, Silveira LR, Ferraz ASM, de Souza JES, Leal-Cardoso JH, Carvalho DP, Ceccatto VM. Transcriptional profile in rat muscle: down-regulation networks in acute strenuous exercise. PeerJ 2021; 9:e10500. [PMID: 33859869 PMCID: PMC8020866 DOI: 10.7717/peerj.10500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Accepted: 11/15/2020] [Indexed: 11/20/2022] Open
Abstract
Background Physical exercise is a health promotion factor regulating gene expression and causing changes in phenotype, varying according to exercise type and intensity. Acute strenuous exercise in sedentary individuals appears to induce different transcriptional networks in response to stress caused by exercise. The objective of this research was to investigate the transcriptional profile of strenuous experimental exercise. Methodology RNA-Seq was performed with Rattus norvegicus soleus muscle, submitted to strenuous physical exercise on a treadmill with an initial velocity of 0.5 km/h and increments of 0.2 km/h at every 3 min until animal exhaustion. Twenty four hours post-physical exercise, RNA-seq protocols were performed with coverage of 30 million reads per sample, 100 pb read length, paired-end, with a list of counts totaling 12816 genes. Results Eighty differentially expressed genes (61 down-regulated and 19 up-regulated) were obtained. Reactome and KEGG database searches revealed the most significant pathways, for down-regulated gene set, were: PI3K-Akt signaling pathway, RAF-MAP kinase, P2Y receptors and Signaling by Erbb2. Results suggest PI3K-AKT pathway inactivation by Hbegf, Fgf1 and Fgr3 receptor regulation, leading to inhibition of cell proliferation and increased apoptosis. Cell signaling transcription networks were found in transcriptome. Results suggest some metabolic pathways which indicate the conditioning situation of strenuous exercise induced genes encoding apoptotic and autophagy factors, indicating cellular stress. Conclusion Down-regulated networks showed cell transduction and signaling pathways, with possible inhibition of cellular proliferation and cell degeneration. These findings reveal transitory and dynamic process in cell signaling transcription networks in skeletal muscle after acute strenuous exercise.
Collapse
Affiliation(s)
| | | | | | - Christina Pacheco
- Superior Institute of Biomedic Sciences, Universidade Estadual do Ceará, Fortaleza, Ceará, Brazil
| | - Danilo Lopes Martins
- Digital Metropolis Institute, Universidade Federal do Rio Grande do Norte, Natal, Rio Grande do Norte, Brazil
| | - Juliana Osório Alves
- Superior Institute of Biomedic Sciences, Universidade Estadual do Ceará, Fortaleza, Ceará, Brazil
| | - Paula Matias Soares
- Superior Institute of Biomedic Sciences, Universidade Estadual do Ceará, Fortaleza, Ceará, Brazil
| | | | - Tanes Lima
- Institute of Biology, Universidade Estadual de Campinas, Campinas, São Paulo, Brazil
| | - Leonardo R Silveira
- Institute of Biology, Universidade Estadual de Campinas, Campinas, São Paulo, Brazil
| | | | | | | | - Denise P Carvalho
- Carlos Chagas Filho Biophysics Institute, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Vania Marilande Ceccatto
- Superior Institute of Biomedic Sciences, Universidade Estadual do Ceará, Fortaleza, Ceará, Brazil
| |
Collapse
|
10
|
Pavis GF, Jameson TSO, Dirks ML, Lee BP, Abdelrahman DR, Murton AJ, Porter C, Alamdari N, Mikus CR, Wall BT, Stephens FB. Improved recovery from skeletal muscle damage is largely unexplained by myofibrillar protein synthesis or inflammatory and regenerative gene expression pathways. Am J Physiol Endocrinol Metab 2021; 320:E291-E305. [PMID: 33284089 PMCID: PMC8260377 DOI: 10.1152/ajpendo.00454.2020] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
The contribution of myofibrillar protein synthesis (MyoPS) to recovery from skeletal muscle damage in humans is unknown. Recreationally active men and women consumed a daily protein-polyphenol beverage targeted at increasing amino acid availability and reducing inflammation (PPB; n = 9), both known to affect MyoPS, or an isocaloric placebo (PLA; n = 9) during 168 h of recovery from 300 maximal unilateral eccentric contractions (EE). Muscle function was assessed daily. Muscle biopsies were collected for 24, 27, 36, 72, and 168 h for MyoPS measurements using 2H2O and expression of 224 genes using RT-qPCR and pathway analysis. PPB improved recovery of muscle function, which was impaired for 5 days after EE in PLA (interaction P < 0.05). Acute postprandial MyoPS rates were unaffected by nutritional intervention (24-27 h). EE increased overnight (27-36 h) MyoPS versus the control leg (PLA: 33 ± 19%; PPB: 79 ± 25%; leg P < 0.01), and PPB tended to increase this further (interaction P = 0.06). Daily MyoPS rates were greater with PPB between 72 and 168 h after EE, albeit after function had recovered. Inflammatory and regenerative signaling pathways were dramatically upregulated and clustered after EE but were unaffected by nutritional intervention. These results suggest that accelerated recovery from EE is not explained by elevated MyoPS or suppression of inflammation.NEW & NOTEWORTHY The present study investigated the contribution of myofibrillar protein synthesis (MyoPS) and associated gene signaling to recovery from 300 muscle-damaging, eccentric contractions. Measured with 2H2O, MyoPS rates were elevated during recovery and observed alongside expression of inflammatory and regenerative signaling pathways. A nutritional intervention accelerated recovery; however, MyoPS and gene signaling were unchanged compared with placebo. These data indicate that MyoPS and associated signaling do not explain accelerated recovery from muscle damage.
Collapse
Affiliation(s)
- George F Pavis
- Nutritional Physiology Group, Department of Sport and Health Sciences, College of Life and Environmental Sciences, University of Exeter, Exeter, United Kingdom
| | - Tom S O Jameson
- Nutritional Physiology Group, Department of Sport and Health Sciences, College of Life and Environmental Sciences, University of Exeter, Exeter, United Kingdom
| | - Marlou L Dirks
- Nutritional Physiology Group, Department of Sport and Health Sciences, College of Life and Environmental Sciences, University of Exeter, Exeter, United Kingdom
| | - Benjamin P Lee
- Institute of Biomedical and Clinical Sciences, University of Exeter Medical School, University of Exeter, Exeter, United Kingdom
| | - Doaa R Abdelrahman
- Department of Surgery, University of Texas Medical Branch, Galveston, Texas
| | - Andrew J Murton
- Department of Surgery, University of Texas Medical Branch, Galveston, Texas
| | - Craig Porter
- Department of Surgery, University of Texas Medical Branch, Galveston, Texas
| | | | | | - Benjamin T Wall
- Nutritional Physiology Group, Department of Sport and Health Sciences, College of Life and Environmental Sciences, University of Exeter, Exeter, United Kingdom
| | - Francis B Stephens
- Nutritional Physiology Group, Department of Sport and Health Sciences, College of Life and Environmental Sciences, University of Exeter, Exeter, United Kingdom
| |
Collapse
|
11
|
Transcriptomic profiling of skeletal muscle adaptations to exercise and inactivity. Nat Commun 2020; 11:470. [PMID: 31980607 PMCID: PMC6981202 DOI: 10.1038/s41467-019-13869-w] [Citation(s) in RCA: 263] [Impact Index Per Article: 52.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Accepted: 11/29/2019] [Indexed: 12/26/2022] Open
Abstract
The molecular mechanisms underlying the response to exercise and inactivity are not fully understood. We propose an innovative approach to profile the skeletal muscle transcriptome to exercise and inactivity using 66 published datasets. Data collected from human studies of aerobic and resistance exercise, including acute and chronic exercise training, were integrated using meta-analysis methods (www.metamex.eu). Here we use gene ontology and pathway analyses to reveal selective pathways activated by inactivity, aerobic versus resistance and acute versus chronic exercise training. We identify NR4A3 as one of the most exercise- and inactivity-responsive genes, and establish a role for this nuclear receptor in mediating the metabolic responses to exercise-like stimuli in vitro. The meta-analysis (MetaMEx) also highlights the differential response to exercise in individuals with metabolic impairments. MetaMEx provides the most extensive dataset of skeletal muscle transcriptional responses to different modes of exercise and an online interface to readily interrogate the database. The pathways that underlie the effects of exercise on metabolism remain incompletely described. Here, the authors perform a meta-analysis of transcriptomic data from 66 published datasets of human skeletal muscle. They identify pathways selectively activated by inactivity, aerobic or resistance exercise, and characterize NR4A3 as one of the genes responsive to inactivity.
Collapse
|
12
|
Valladares-Ide D, Peñailillo L, Collao N, Marambio H, Deldicque L, Zbinden-Foncea H. Activation of protein synthesis, regeneration, and MAPK signaling pathways following repeated bouts of eccentric cycling. Am J Physiol Endocrinol Metab 2019; 317:E1131-E1139. [PMID: 31593504 DOI: 10.1152/ajpendo.00216.2019] [Citation(s) in RCA: 9] [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] [Indexed: 12/21/2022]
Abstract
The aim of this study was to examine the activation of skeletal muscle signaling pathways related to protein synthesis and the gene expression of regeneration/degradation markers following repeated bouts of eccentric cycling. Nine untrained men (25.4 ± 1.9 yr) performed two 30-min eccentric cycling bouts (ECC1, ECC2) at 85% of maximal concentric workload, separated by 2 wk. Muscle biopsies were taken from the vastus lateralis before and 2 h after each bout. Indirect markers of muscle damage were assessed before and 24-48 h after exercise. Changes in the Akt/mammalian target of rapamycin (mTOR)/rbosomal protein S6 kinase 1 (S6K1)/ribosomal protein S6 (rpS6) and MAPK signaling pathways were measured by Western blot and changes in mRNA expression of IL-6 and IL-1β, and myogenic regulatory factors (MRFs) were measured by real-time PCR. ECC1 induced greater increases in indirect markers of muscle damage compared with ECC2. Phosphorylation of S6K1 and rpS6 increased after both exercise bouts (P < 0.05), whereas phosphorylation of mTOR increased after ECC2 only (P = 0.03). Atrogin-1 mRNA expression decreased after ECC1 and ECC2 (P < 0.05) without changes in muscle RING-finger protein-1 mRNA. Basal mRNA levels of myoblast determination protein-1 (MyoD), MRF4, and myogenin were higher 2 wk after ECC1 (P < 0.05). MRF4 mRNA increased after ECC1 and ECC2 (P < 0.05), whereas MyoD mRNA expression increased only after ECC1 (P = 0.03). Phosphorylation of JNK and p38 MAPK increased after both exercise bouts (P < 0.05), similar to IL-6 and IL-1β mRNA expression. All together, these results suggest that differential regulation of the mTOR pathway and MRF expression could mediate the repeated bout effect observed between an initial and secondary bout of eccentric exercise.
Collapse
Affiliation(s)
- Denisse Valladares-Ide
- Exercise Science Laboratory, School of Kinesiology, Faculty of Medicine, Universidad Finis Terrae, Santiago, Chile
| | - Luis Peñailillo
- Exercise Science Laboratory, School of Kinesiology, Faculty of Medicine, Universidad Finis Terrae, Santiago, Chile
| | - Nicolás Collao
- Exercise Science Laboratory, School of Kinesiology, Faculty of Medicine, Universidad Finis Terrae, Santiago, Chile
| | - Hugo Marambio
- Centro de Salud Deportiva, Clínica Santa María, Santiago, Chile
| | - Louise Deldicque
- Institute of Neuroscience, Université Catholique de Louvain, Louvain-la-Neuve, Belgium
| | - Hermann Zbinden-Foncea
- Exercise Science Laboratory, School of Kinesiology, Faculty of Medicine, Universidad Finis Terrae, Santiago, Chile
- Centro de Salud Deportiva, Clínica Santa María, Santiago, Chile
| |
Collapse
|
13
|
Latimer LE, Constantin D, Greening NJ, Calvert L, Menon MK, Steiner MC, Greenhaff PL. Impact of transcutaneous neuromuscular electrical stimulation or resistance exercise on skeletal muscle mRNA expression in COPD. Int J Chron Obstruct Pulmon Dis 2019; 14:1355-1364. [PMID: 31308645 PMCID: PMC6612952 DOI: 10.2147/copd.s189896] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Accepted: 03/05/2019] [Indexed: 12/11/2022] Open
Abstract
Background: Voluntary resistance exercise (RE) training increases muscle mass and strength in patients with chronic obstructive pulmonary disease (COPD). Nonvolitional transcutaneous neuromuscular electrical stimulation (NMES) may be an alternative strategy for reducing ambulatory muscle weakness in patients unable to perform RE training, but little comparative data are available. This study, therefore, investigated changes in muscle mRNA abundance of a number of gene targets in response to a single bout of NMES compared with RE. Methods: Twenty-six patients with stable COPD (15 male; FEV1, 43±18% predicted; age, 64±8 years; fat free mass index, 16.6±1.8 kg/m2) undertook 30 minutes of quadriceps NMES (50 Hz, current at the limit of tolerance) or 5×30 maximal voluntary isokinetic knee extensions. Vastus lateralis muscle biopsies were obtained at rest immediately before and 24 hours after intervention. Expression of 384 targeted mRNA transcripts was assessed by real time TaqMan PCR. Significant change in expression from baseline was determined using the ΔΔCT method with a false discovery rate (FDR) of <5%. Results: NMES and RE altered mRNA abundance of 18 and 68 genes, respectively (FDR <5%), of which 14 genes were common to both interventions and of the same magnitude of fold change. Biological functions of upregulated genes included inflammation, hypertrophy, muscle protein turnover, and muscle growth, whilst downregulated genes included mitochondrial and cell signaling functions. Conclusions: Compared with NMES, RE had a broader impact on mRNA abundance and, therefore, appears to be the superior intervention for maximizing transcriptional responses in the quadriceps of patients with COPD. However, if voluntary RE is not feasible in a clinical setting, NMES by modifying expression of genes known to impact upon muscle mass and strength may have a positive influence on muscle function.
Collapse
Affiliation(s)
- Lorna E Latimer
- Department of Respiratory Sciences, University of Leicester, Leicester, UK.,Institute for Lung Health, National Institute for Health Research (NIHR) Leicester Biomedical Research Centre - Respiratory, Glenfield Hospital, Leicester, UK
| | - Despina Constantin
- Medical Research Council/Arthritis Research UK (MRC/ARUK) Centre for Musculoskeletal Ageing Research, University of Nottingham, Nottingham, UK.,Centre for Sport, Exercise and Osteoarthritis Research, University of Nottingham, Nottingham, UK.,NIHR Nottingham Biomedical Research Centre, University of Nottingham, Nottingham, UK
| | - Neil J Greening
- Department of Respiratory Sciences, University of Leicester, Leicester, UK.,Institute for Lung Health, National Institute for Health Research (NIHR) Leicester Biomedical Research Centre - Respiratory, Glenfield Hospital, Leicester, UK
| | - Lori Calvert
- Peterborough and Stamford Hospitals NHS Foundation Trust, Peterborough City Hospital, Bretton, UK
| | - Manoj K Menon
- Barking, Havering and Redbridge University Hospitals NHS Trust, Chest Clinic, King George Hospital, Ilford, UK
| | - Michael C Steiner
- Department of Respiratory Sciences, University of Leicester, Leicester, UK.,Institute for Lung Health, National Institute for Health Research (NIHR) Leicester Biomedical Research Centre - Respiratory, Glenfield Hospital, Leicester, UK
| | - Paul L Greenhaff
- Medical Research Council/Arthritis Research UK (MRC/ARUK) Centre for Musculoskeletal Ageing Research, University of Nottingham, Nottingham, UK.,Centre for Sport, Exercise and Osteoarthritis Research, University of Nottingham, Nottingham, UK.,NIHR Nottingham Biomedical Research Centre, University of Nottingham, Nottingham, UK
| |
Collapse
|
14
|
Comparative Transcriptome and Methylome Analysis in Human Skeletal Muscle Anabolism, Hypertrophy and Epigenetic Memory. Sci Rep 2019; 9:4251. [PMID: 30862794 PMCID: PMC6414679 DOI: 10.1038/s41598-019-40787-0] [Citation(s) in RCA: 72] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Accepted: 02/22/2019] [Indexed: 02/07/2023] Open
Abstract
Transcriptome wide changes in human skeletal muscle after acute (anabolic) and chronic resistance exercise (RE) induced hypertrophy have been extensively determined in the literature. We have also recently undertaken DNA methylome analysis (850,000 + CpG sites) in human skeletal muscle after acute and chronic RE, detraining and retraining, where we identified an association between DNA methylation and epigenetic memory of exercise induced skeletal muscle hypertrophy. However, it is currently unknown as to whether all the genes identified in the transcriptome studies to date are also epigenetically regulated at the DNA level after acute, chronic or repeated RE exposure. We therefore aimed to undertake large scale bioinformatical analysis by pooling the publicly available transcriptome data after acute (110 samples) and chronic RE (181 samples) and comparing these large data sets with our genome-wide DNA methylation analysis in human skeletal muscle after acute and chronic RE, detraining and retraining. Indeed, after acute RE we identified 866 up- and 936 down-regulated genes at the expression level, with 270 (out of the 866 up-regulated) identified as being hypomethylated, and 216 (out of 936 downregulated) as hypermethylated. After chronic RE we identified 2,018 up- and 430 down-regulated genes with 592 (out of 2,018 upregulated) identified as being hypomethylated and 98 (out of 430 genes downregulated) as hypermethylated. After KEGG pathway analysis, genes associated with ‘cancer’ pathways were significantly enriched in both bioinformatic analysis of the pooled transcriptome and methylome datasets after both acute and chronic RE. This resulted in 23 (out of 69) and 28 (out of 49) upregulated and hypomethylated and 12 (out of 37) and 2 (out of 4) downregulated and hypermethylated ‘cancer’ genes following acute and chronic RE respectively. Within skeletal muscle tissue, these ‘cancer’ genes predominant functions were associated with matrix/actin structure and remodelling, mechano-transduction (e.g. PTK2/Focal Adhesion Kinase and Phospholipase D- following chronic RE), TGF-beta signalling and protein synthesis (e.g. GSK3B after acute RE). Interestingly, 51 genes were also identified to be up/downregulated in both the acute and chronic RE pooled transcriptome analysis as well as significantly hypo/hypermethylated after acute RE, chronic RE, detraining and retraining. Five genes; FLNB, MYH9, SRGAP1, SRGN, ZMIZ1 demonstrated increased gene expression in the acute and chronic RE transcriptome and also demonstrated hypomethylation in these conditions. Importantly, these 5 genes demonstrated retained hypomethylation even during detraining (following training induced hypertrophy) when exercise was ceased and lean mass returned to baseline (pre-training) levels, identifying them as genes associated with epigenetic memory in skeletal muscle. Importantly, for the first time across the transcriptome and epigenome combined, this study identifies novel differentially methylated genes associated with human skeletal muscle anabolism, hypertrophy and epigenetic memory.
Collapse
|
15
|
Reitzner SM, Norrbom J, Sundberg CJ, Gidlund E. Expression of striated activator of rho-signaling in human skeletal muscle following acute exercise and long-term training. Physiol Rep 2018; 6:e13624. [PMID: 29504288 PMCID: PMC5835521 DOI: 10.14814/phy2.13624] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Accepted: 01/31/2018] [Indexed: 12/20/2022] Open
Abstract
The striated activator of rho-signaling (STARS) protein acts as a link between external stimuli and exercise adaptation such as muscle hypertrophy. However, the acute and long-term adaptational response of STARS is still unclear. This study aimed at investigating the acute and long-term endurance training response on the mRNA and protein expression of STARS and its related upstream and downstream factors in human skeletal muscle. mRNA and protein levels of STARS and related factors were assessed in skeletal muscle of healthy young men and women following an acute bout of endurance exercise (n = 15) or 12 weeks of one-legged training (n = 23). Muscle biopsies were obtained before (acute and long-term), at 30 min, 2, and 6 h following acute exercise, and at 24 h following both acute exercise and long-term training. Following acute exercise, STARS mRNA was significantly elevated 3.9-fold at 30 min returning back to baseline 24 h after exercise. STARS protein levels were numerically but nonsignificantly increased 7.2-fold at 24 h. No changes in STARS or ERRα mRNA or STARS protein expression were seen following long-term training. PGC-1α mRNA increased 1.7-fold following long-term training. MRTF-A mRNA was increased both following acute exercise and long-term training, in contrast to SRF mRNA and protein which did not change. STARS mRNA is acutely upregulated with exercise, but there is no cumulative effect to long-term training as seen in PGC-1α mRNA expression. Exercise intensity might play a role in manifestation of protein expression, suggesting a more complex regulation of STARS.
Collapse
Affiliation(s)
- Stefan M. Reitzner
- Department of Physiology and PharmacologyKarolinska InstitutetStockholmSweden
| | - Jessica Norrbom
- Department of Physiology and PharmacologyKarolinska InstitutetStockholmSweden
| | - Carl Johan Sundberg
- Department of Physiology and PharmacologyKarolinska InstitutetStockholmSweden
- Department of Learning, Informatics, Management and EthicsKarolinska InstitutetStockholmSweden
| | - Eva‐Karin Gidlund
- Department of Physiology and PharmacologyKarolinska InstitutetStockholmSweden
| |
Collapse
|
16
|
Transcriptional and Post-Translational Targeting of Myocyte Stress Protein 1 (MS1) by the JNK Pathway in Cardiac Myocytes. J Mol Signal 2017; 12:3. [PMID: 30210579 PMCID: PMC5853832 DOI: 10.5334/1750-2187-12-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Myocyte Stress Protein 1 (MS1) is a muscle-specific, stress-responsive, regulator of gene expression. It was originally identified in embryonic mouse heart which showed increased expression in a rat model of left ventricular hypertrophy. To determine if MS1 was responsive to other stresses relevant to cardiac myocyte function, we tested if it could be induced by the metabolic stresses associated with ischaemia/reperfusion injury in cardiac myocytes. We found that metabolic stress increased MS1 expression, both at the mRNA and protein level, concurrent with activation of the c-Jun N-terminal Kinase (JNK) signalling pathway. MS1 induction by metabolic stress was blocked by both the transcription inhibitor actinomycin D and a JNK inhibitor, suggesting that activation of the JNK pathway during metabolic stress in cardiac myocytes leads to transcriptional induction of MS1. MS1 was also found to be an efficient JNK substrate in vitro, with a major JNK phosphorylation site identified at Thr-62. In addition, MS1 was found to co-precipitate with JNK, and inspection of the amino acid sequence upstream of the phosphorylation site, at Thr-62, revealed a putative Mitogen-Activated Protein Kinase (MAPK) binding site. Taken together, these data identify MS1 as a likely transcriptional and post-translational target for the JNK pathway in cardiac myocytes subjected to metabolic stress.
Collapse
|
17
|
Fernández-Verdejo R, Vanwynsberghe AM, Hai T, Deldicque L, Francaux M. Activating transcription factor 3 regulates chemokine expression in contracting C 2C 12 myotubes and in mouse skeletal muscle after eccentric exercise. Biochem Biophys Res Commun 2017; 492:249-254. [PMID: 28822763 DOI: 10.1016/j.bbrc.2017.08.059] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Accepted: 08/15/2017] [Indexed: 01/05/2023]
Abstract
Activating transcription factor (ATF) 3 regulates chemokine expression in various cell types and tissues. Herein, we studied this regulation in contracting muscle cells in vitro, and in skeletal muscle after muscle-damaging exercise in vivo. C2C12 myotubes with normal or low ATF3 levels (atf3_siRNA) were electrically stimulated (EPS). Also, ATF3-knockout (ATF3-KO) and control mice ran downhill until exhaustion, and muscles were analyzed post-exercise. EPS increased ATF3 levels in myotubes (P < 0.01). Chemokine C-C motif ligand (ccl) 2 mRNA increased post-EPS, but atf3_siRNA attenuated the response (P < 0.05). Atf3_siRNA up-regulated ccl6 basal mRNA, and down-regulated ccl9 and chemokine C-X-C motif ligand (cxcl) 1 basal mRNAs. Post-exercise, ATF3-KO mice showed exacerbated mRNA levels of ccl6 and ccl9 in soleus (P < 0.05), and similar trends were observed for ccl2 and interleukin (il) 1β (P < 0.09). In quadriceps, il6 mRNA level increased only in ATF3-KO (P < 0.05), and cxcl1 mRNA showed a similar trend (P = 0.082). Cluster of differentiation-68 (cd68) mRNA, a macrophage marker, increased in quadriceps and soleus independently of genotype (P < 0.001). Our data demonstrate that ATF3 regulates chemokine expression in muscle cells in vitro and skeletal muscle in vivo, but the regulation differs in each model. Cells other than myofibers may thus participate in the response observed in skeletal muscle. Our results also indicate that ATF3-independent mechanisms would regulate macrophage infiltration upon muscle-damaging exercise. The implications of chemokine regulation in skeletal muscle remain to be determined.
Collapse
Affiliation(s)
- R Fernández-Verdejo
- Institute of Neuroscience, Université Catholique de Louvain, Louvain-la-Neuve, Belgium
| | - A M Vanwynsberghe
- Institute of Neuroscience, Université Catholique de Louvain, Louvain-la-Neuve, Belgium
| | - T Hai
- Department of Biological Chemistry and Pharmacology, Ohio State University, Columbus, OH, USA
| | - L Deldicque
- Institute of Neuroscience, Université Catholique de Louvain, Louvain-la-Neuve, Belgium
| | - M Francaux
- Institute of Neuroscience, Université Catholique de Louvain, Louvain-la-Neuve, Belgium.
| |
Collapse
|
18
|
Russell AP, Wallace MA, Kalanon M, Zacharewicz E, Della Gatta PA, Garnham A, Lamon S. Striated muscle activator of Rho signalling (STARS) is reduced in ageing human skeletal muscle and targeted by miR-628-5p. Acta Physiol (Oxf) 2017; 220:263-274. [PMID: 27739650 DOI: 10.1111/apha.12819] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2016] [Revised: 09/12/2016] [Accepted: 10/11/2016] [Indexed: 12/12/2022]
Abstract
AIM The striated muscle activator of Rho signalling (STARS) is a muscle-specific actin-binding protein. The STARS signalling pathway is activated by resistance exercise and is anticipated to play a role in signal mechanotransduction. Animal studies have reported a negative regulation of STARS signalling with age, but such regulation has not been investigated in humans. METHODS Ten young (18-30 years) and 10 older (60-75 years) subjects completed an acute bout of resistance exercise. Gene and protein expression of members of the STARS signalling pathway and miRNA expression of a subset of miRNAs, predicted or known to target members of STARS signalling pathway, were measured in muscle biopsies collected pre-exercise and 2 h post-exercise. RESULTS For the first time, we report a significant downregulation of the STARS protein in older subjects. However, there was no effect of age on the magnitude of STARS activation in response to an acute bout of exercise. Finally, we established that miR-628-5p, a miRNA regulated by age and exercise, binds to the STARS 3'UTR to directly downregulate its transcription. CONCLUSION This study describes for the first time the resistance exercise-induced regulation of STARS signalling in skeletal muscle from older humans and identifies a new miRNA involved in the transcriptional control of STARS.
Collapse
Affiliation(s)
- A. P. Russell
- Institute for Physical Activity and Nutrition (IPAN); School of Exercise and Nutrition Sciences; Deakin University; Geelong Vic. Australia
| | - M. A. Wallace
- Institute for Physical Activity and Nutrition (IPAN); School of Exercise and Nutrition Sciences; Deakin University; Geelong Vic. Australia
| | - M. Kalanon
- Institute for Physical Activity and Nutrition (IPAN); School of Exercise and Nutrition Sciences; Deakin University; Geelong Vic. Australia
| | - E. Zacharewicz
- Institute for Physical Activity and Nutrition (IPAN); School of Exercise and Nutrition Sciences; Deakin University; Geelong Vic. Australia
| | - P. A. Della Gatta
- Institute for Physical Activity and Nutrition (IPAN); School of Exercise and Nutrition Sciences; Deakin University; Geelong Vic. Australia
| | - A. Garnham
- Institute for Physical Activity and Nutrition (IPAN); School of Exercise and Nutrition Sciences; Deakin University; Geelong Vic. Australia
| | - S. Lamon
- Institute for Physical Activity and Nutrition (IPAN); School of Exercise and Nutrition Sciences; Deakin University; Geelong Vic. Australia
| |
Collapse
|
19
|
Rindom E, Vissing K. Mechanosensitive Molecular Networks Involved in Transducing Resistance Exercise-Signals into Muscle Protein Accretion. Front Physiol 2016; 7:547. [PMID: 27909410 PMCID: PMC5112233 DOI: 10.3389/fphys.2016.00547] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2016] [Accepted: 10/31/2016] [Indexed: 02/05/2023] Open
Abstract
Loss of skeletal muscle myofibrillar protein with disease and/or inactivity can severely deteriorate muscle strength and function. Strategies to counteract wasting of muscle myofibrillar protein are therefore desirable and invite for considerations on the potential superiority of specific modes of resistance exercise and/or the adequacy of low load resistance exercise regimens as well as underlying mechanisms. In this regard, delineation of the potentially mechanosensitive molecular mechanisms underlying muscle protein synthesis (MPS), may contribute to an understanding on how differentiated resistance exercise can transduce a mechanical signal into stimulation of muscle accretion. Recent findings suggest specific upstream exercise-induced mechano-sensitive myocellular signaling pathways to converge on mammalian target of rapamycin complex 1 (mTORC1), to influence MPS. This may e.g. implicate mechanical activation of signaling through a diacylglycerol kinase (DGKζ)-phosphatidic acid (PA) axis or implicate integrin deformation to signal through a Focal adhesion kinase (FAK)-Tuberous Sclerosis Complex 2 (TSC2)-Ras homolog enriched in brain (Rheb) axis. Moreover, since initiation of translation is reliant on mRNA, it is also relevant to consider potentially mechanosensitive signaling pathways involved in muscle myofibrillar gene transcription and whether some of these pathways converge with those affecting mTORC1 activation for MPS. In this regard, recent findings suggest how mechanical stress may implicate integrin deformation and/or actin dynamics to signal through a Ras homolog gene family member A protein (RhoA)-striated muscle activator of Rho signaling (STARS) axis or implicate deformation of Notch to affect Bone Morphogenetic Protein (BMP) signaling through a small mother of decapentaplegic (Smad) axis.
Collapse
Affiliation(s)
- Emil Rindom
- Section of Sport Science, Department of Public Health, Aarhus UniversityAarhus, Denmark; Department of Biomedicine, Aarhus UniversityAarhus, Denmark
| | - Kristian Vissing
- Section of Sport Science, Department of Public Health, Aarhus University Aarhus, Denmark
| |
Collapse
|
20
|
Fernández-Verdejo R, Vanwynsberghe AM, Essaghir A, Demoulin JB, Hai T, Deldicque L, Francaux M. Activating transcription factor 3 attenuates chemokine and cytokine expression in mouse skeletal muscle after exercise and facilitates molecular adaptation to endurance training. FASEB J 2016; 31:840-851. [PMID: 27856557 DOI: 10.1096/fj.201600987r] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2016] [Accepted: 10/31/2016] [Indexed: 12/17/2022]
Abstract
Activating transcription factor (ATF)3 regulates the expression of inflammation-related genes in several tissues under pathological contexts. In skeletal muscle, atf3 expression increases after exercise, but its target genes remain unknown. We aimed to identify those genes and to determine the influence of ATF3 on muscle adaptation to training. Skeletal muscles of ATF3-knockout (ATF3-KO) and control mice were analyzed at rest, after exercise, and after training. In resting muscles, there was no difference between genotypes in enzymatic activities or fiber type. After exercise, a microarray analysis in quadriceps revealed ATF3 affects genes modulating chemotaxis and chemokine/cytokine activity. Quantitative PCR showed that the mRNA levels of chemokine C-C motif ligand (ccl)8 and chemokine C-X-C motif ligand (cxcl)13 were higher in quadriceps of ATF3-KO mice than in control mice. The same was observed for ccl9 and cxcl13 in soleus. Also in soleus, ccl2, interleukin (il)6, il1β, and cluster of differentiation (cd)68 mRNA levels increased after exercise only in ATF3-KO mice. Endurance training increased the basal mRNA level of hexokinase-2, hormone sensitive lipase, glutathione peroxidase-1, and myosin heavy chain IIa in quadriceps of control mice but not in ATF3-KO mice. In summary, ATF3 attenuates the expression of inflammation-related genes after exercise and thus facilitates molecular adaptation to training.-Fernández-Verdejo, R., Vanwynsberghe, A. M., Essaghir, A., Demoulin, J.-B., Hai, T., Deldicque, L., Francaux, M. Activating transcription factor 3 attenuates chemokine and cytokine expression in mouse skeletal muscle after exercise and facilitates molecular adaptation to endurance training.
Collapse
Affiliation(s)
| | - Aline M Vanwynsberghe
- Institute of Neuroscience, Université Catholique de Louvain, Louvain-la-Neuve, Belgium
| | - Ahmed Essaghir
- De Duve Institute, Université Catholique de Louvain, Brussels, Belgium; and
| | | | - Tsonwin Hai
- Department of Biological Chemistry and Pharmacology, Ohio State University, Columbus, Ohio, USA
| | - Louise Deldicque
- Institute of Neuroscience, Université Catholique de Louvain, Louvain-la-Neuve, Belgium
| | - Marc Francaux
- Institute of Neuroscience, Université Catholique de Louvain, Louvain-la-Neuve, Belgium;
| |
Collapse
|
21
|
Comparative Transcriptomic Analyses by RNA-seq to Elucidate Differentially Expressed Genes in the Muscle of Korean Thoroughbred Horses. Appl Biochem Biotechnol 2016; 180:588-608. [DOI: 10.1007/s12010-016-2118-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2016] [Accepted: 05/05/2016] [Indexed: 12/27/2022]
|
22
|
Leber Y, Ruparelia AA, Kirfel G, van der Ven PFM, Hoffmann B, Merkel R, Bryson-Richardson RJ, Fürst DO. Filamin C is a highly dynamic protein associated with fast repair of myofibrillar microdamage. Hum Mol Genet 2016; 25:2776-2788. [PMID: 27206985 DOI: 10.1093/hmg/ddw135] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2016] [Revised: 04/26/2016] [Accepted: 04/27/2016] [Indexed: 11/12/2022] Open
Abstract
Filamin c (FLNc) is a large dimeric actin-binding protein located at premyofibrils, myofibrillar Z-discs and myofibrillar attachment sites of striated muscle cells, where it is involved in mechanical stabilization, mechanosensation and intracellular signaling. Mutations in the gene encoding FLNc give rise to skeletal muscle diseases and cardiomyopathies. Here, we demonstrate by fluorescence recovery after photobleaching that a large fraction of FLNc is highly mobile in cultured neonatal mouse cardiomyocytes and in cardiac and skeletal muscles of live transgenic zebrafish embryos. Analysis of cardiomyocytes from Xirp1 and Xirp2 deficient animals indicates that both Xin actin-binding repeat-containing proteins stabilize FLNc selectively in premyofibrils. Using a novel assay to analyze myofibrillar microdamage and subsequent repair in cultured contracting cardiomyocytes by live cell imaging, we demonstrate that repair of damaged myofibrils is achieved within only 4 h, even in the absence of de novo protein synthesis. FLNc is immediately recruited to these sarcomeric lesions together with its binding partner aciculin and precedes detectable assembly of filamentous actin and recruitment of other myofibrillar proteins. These data disclose an unprecedented degree of flexibility of the almost crystalline contractile machinery and imply FLNc as a dynamic signaling hub, rather than a primarily structural protein. Our myofibrillar damage/repair model illustrates how (cardio)myocytes are kept functional in their mechanically and metabolically strained environment. Our results help to better understand the pathomechanisms and pathophysiology of early stages of FLNc-related myofibrillar myopathy and skeletal and cardiac diseases preceding pathological protein aggregation.
Collapse
Affiliation(s)
- Yvonne Leber
- Department of Molecular Cell Biology, Institute for Cell Biology, University of Bonn, D53121 Bonn, Germany
| | - Avnika A Ruparelia
- School of Biological Sciences, Monash University, Melbourne, Victoria 3800, Australia
| | - Gregor Kirfel
- Department of Molecular Cell Biology, Institute for Cell Biology, University of Bonn, D53121 Bonn, Germany
| | - Peter F M van der Ven
- Department of Molecular Cell Biology, Institute for Cell Biology, University of Bonn, D53121 Bonn, Germany
| | - Bernd Hoffmann
- Department of Biomechanics (ICS-7), Institute of Complex Systems, Forschungszentrum Jülich, D52428 Jülich, Germany and
| | - Rudolf Merkel
- Department of Biomechanics (ICS-7), Institute of Complex Systems, Forschungszentrum Jülich, D52428 Jülich, Germany and.,Department of Biomechanics, Institute for Physical and Theoretical Chemistry, University of Bonn, D53115 Bonn, Germany
| | | | - Dieter O Fürst
- Department of Molecular Cell Biology, Institute for Cell Biology, University of Bonn, D53121 Bonn, Germany
| |
Collapse
|
23
|
Wallace MA, Della Gatta PA, Ahmad Mir B, Kowalski GM, Kloehn J, McConville MJ, Russell AP, Lamon S. Overexpression of Striated Muscle Activator of Rho Signaling (STARS) Increases C2C12 Skeletal Muscle Cell Differentiation. Front Physiol 2016; 7:7. [PMID: 26903873 PMCID: PMC4745265 DOI: 10.3389/fphys.2016.00007] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2015] [Accepted: 01/11/2016] [Indexed: 01/10/2023] Open
Abstract
Background: Skeletal muscle growth and regeneration depend on the activation of satellite cells, which leads to myocyte proliferation, differentiation and fusion with existing muscle fibers. Skeletal muscle cell proliferation and differentiation are tightly coordinated by a continuum of molecular signaling pathways. The striated muscle activator of Rho signaling (STARS) is an actin binding protein that regulates the transcription of genes involved in muscle cell growth, structure and function via the stimulation of actin polymerization and activation of serum-response factor (SRF) signaling. STARS mediates cell proliferation in smooth and cardiac muscle models; however, whether STARS overexpression enhances cell proliferation and differentiation has not been investigated in skeletal muscle cells. Results: We demonstrate for the first time that STARS overexpression enhances differentiation but not proliferation in C2C12 mouse skeletal muscle cells. Increased differentiation was associated with an increase in the gene levels of the myogenic differentiation markers Ckm, Ckmt2 and Myh4, the differentiation factor Igf2 and the myogenic regulatory factors (MRFs) Myf5 and Myf6. Exposing C2C12 cells to CCG-1423, a pharmacological inhibitor of SRF preventing the nuclear translocation of its co-factor MRTF-A, had no effect on myotube differentiation rate, suggesting that STARS regulates differentiation via a MRTF-A independent mechanism. Conclusion: These findings position STARS as an important regulator of skeletal muscle growth and regeneration.
Collapse
Affiliation(s)
- Marita A Wallace
- Centre for Physical Activity and Nutrition Research, School of Exercise and Nutrition Sciences, Deakin University Burwood, VIC, Australia
| | - Paul A Della Gatta
- Centre for Physical Activity and Nutrition Research, School of Exercise and Nutrition Sciences, Deakin University Burwood, VIC, Australia
| | - Bilal Ahmad Mir
- Centre for Physical Activity and Nutrition Research, School of Exercise and Nutrition Sciences, Deakin University Burwood, VIC, Australia
| | - Greg M Kowalski
- Centre for Physical Activity and Nutrition Research, School of Exercise and Nutrition Sciences, Deakin University Burwood, VIC, Australia
| | - Joachim Kloehn
- Department of Biochemistry and Molecular Biology, Bio21 Institute of Molecular Science and Biotechnology, University of Melbourne Parkville, VIC, Australia
| | - Malcom J McConville
- Department of Biochemistry and Molecular Biology, Bio21 Institute of Molecular Science and Biotechnology, University of Melbourne Parkville, VIC, Australia
| | - Aaron P Russell
- Centre for Physical Activity and Nutrition Research, School of Exercise and Nutrition Sciences, Deakin University Burwood, VIC, Australia
| | - Séverine Lamon
- Centre for Physical Activity and Nutrition Research, School of Exercise and Nutrition Sciences, Deakin University Burwood, VIC, Australia
| |
Collapse
|
24
|
Abstract
Rnd3, also known as RhoE, belongs to the Rnd subclass of the Rho family of small guanosine triphosphate (GTP)-binding proteins. Rnd proteins are unique due to their inability to switch from a GTP-bound to GDP-bound conformation. Even though studies of the biological function of Rnd3 are far from being concluded, information is available regarding its expression pattern, cellular localization, and its activity, which can be altered depending on the conditions. The compiled data from these studies implies that Rnd3 may not be a traditional small GTPase. The basic role of Rnd3 is to report as an endogenous antagonist of RhoA signaling-mediated actin cytoskeleton dynamics, which specifically contributes to cell migration and neuron polarity. In addition, Rnd3 also plays a critical role in arresting cell cycle distribution, inhibiting cell growth, and inducing apoptosis and differentiation. Increasing data have shown that aberrant Rnd3 expression may be the leading cause of some systemic diseases; particularly highlighted in apoptotic cardiomyopathy, developmental arrhythmogenesis and heart failure, hydrocephalus, as well as tumor metastasis and chemotherapy resistance. Therefore, a better understanding of the function of Rnd3 under different physiological and pathological conditions, through the use of suitable models, would provide a novel insight into the origin and treatment of multiple human diseases.
Collapse
Affiliation(s)
- Wei Jie
- Department of Pathology, School of Basic Medicine Science, Guangdong Medical College, Zhanjiang, Guangdong Province, China
| | - Kelsey C Andrade
- Texas A&M University Health Science Center, Institute of Biosciences and Technology, Houston, Texas, USA
| | - Xi Lin
- Texas A&M University Health Science Center, Institute of Biosciences and Technology, Houston, Texas, USA
| | - Xiangsheng Yang
- Texas A&M University Health Science Center, Institute of Biosciences and Technology, Houston, Texas, USA
| | - Xiaojing Yue
- Texas A&M University Health Science Center, Institute of Biosciences and Technology, Houston, Texas, USA
| | - Jiang Chang
- Texas A&M University Health Science Center, Institute of Biosciences and Technology, Houston, Texas, USA
| |
Collapse
|
25
|
Repeated bouts of fast velocity eccentric contractions induce atrophy of gastrocnemius muscle in rats. J Muscle Res Cell Motil 2015; 36:317-27. [DOI: 10.1007/s10974-015-9426-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2015] [Accepted: 10/09/2015] [Indexed: 10/22/2022]
|
26
|
Dyar KA, Ciciliot S, Tagliazucchi GM, Pallafacchina G, Tothova J, Argentini C, Agatea L, Abraham R, Ahdesmäki M, Forcato M, Bicciato S, Schiaffino S, Blaauw B. The calcineurin-NFAT pathway controls activity-dependent circadian gene expression in slow skeletal muscle. Mol Metab 2015; 4:823-33. [PMID: 26629406 PMCID: PMC4632177 DOI: 10.1016/j.molmet.2015.09.004] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2015] [Accepted: 09/15/2015] [Indexed: 12/17/2022] Open
Abstract
Objective Physical activity and circadian rhythms are well-established determinants of human health and disease, but the relationship between muscle activity and the circadian regulation of muscle genes is a relatively new area of research. It is unknown whether muscle activity and muscle clock rhythms are coupled together, nor whether activity rhythms can drive circadian gene expression in skeletal muscle. Methods We compared the circadian transcriptomes of two mouse hindlimb muscles with vastly different circadian activity patterns, the continuously active slow soleus and the sporadically active fast tibialis anterior, in the presence or absence of a functional skeletal muscle clock (skeletal muscle-specific Bmal1 KO). In addition, we compared the effect of denervation on muscle circadian gene expression. Results We found that different skeletal muscles exhibit major differences in their circadian transcriptomes, yet core clock gene oscillations were essentially identical in fast and slow muscles. Furthermore, denervation caused relatively minor changes in circadian expression of most core clock genes, yet major differences in expression level, phase and amplitude of many muscle circadian genes. Conclusions We report that activity controls the oscillation of around 15% of skeletal muscle circadian genes independently of the core muscle clock, and we have identified the Ca2+-dependent calcineurin-NFAT pathway as an important mediator of activity-dependent circadian gene expression, showing that circadian locomotor activity rhythms drive circadian rhythms of NFAT nuclear translocation and target gene expression. Activity is a major extrinsic factor driving ∼15% of muscle circadian genes. Calcineurin-NFAT drives activity-dependent circadian gene expression in muscle. The majority of skeletal muscle circadian genes are muscle type-specific. A common set of skeletal muscle circadian genes are clock-dependent.
Collapse
Affiliation(s)
- Kenneth A Dyar
- Venetian Institute of Molecular Medicine (VIMM), Padova, Italy
| | | | - Guidantonio Malagoli Tagliazucchi
- Center for Genome Research, Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy ; Istituto Nazionale Genetica Molecolare 'Romeo ed Enrica Invernizzi', Via F. Sforza 35, 20122 Milan, Italy
| | - Giorgia Pallafacchina
- Venetian Institute of Molecular Medicine (VIMM), Padova, Italy ; Institute of Neurosciences, Consiglio Nazionale delle Ricerche (CNR), Padova, Italy
| | - Jana Tothova
- Venetian Institute of Molecular Medicine (VIMM), Padova, Italy
| | - Carla Argentini
- Venetian Institute of Molecular Medicine (VIMM), Padova, Italy
| | - Lisa Agatea
- Venetian Institute of Molecular Medicine (VIMM), Padova, Italy
| | - Reimar Abraham
- Venetian Institute of Molecular Medicine (VIMM), Padova, Italy
| | - Miika Ahdesmäki
- Institute for Medical Informatics, Statistics and Epidemiology, University of Leipzig, Germany
| | - Mattia Forcato
- Center for Genome Research, Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Silvio Bicciato
- Center for Genome Research, Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Stefano Schiaffino
- Venetian Institute of Molecular Medicine (VIMM), Padova, Italy ; Institute of Neurosciences, Consiglio Nazionale delle Ricerche (CNR), Padova, Italy
| | - Bert Blaauw
- Venetian Institute of Molecular Medicine (VIMM), Padova, Italy ; Department of Biomedical Sciences, University of Padova, Italy
| |
Collapse
|
27
|
Cho HW, Shin S, Song KD, Park JW, Choi JY, Lee HK, Cho BW. Molecular Characterization and Expression Analysis of Adrenergic Receptor Beta 2 (ADRB2) Gene before and after Exercise in the Horse. ASIAN-AUSTRALASIAN JOURNAL OF ANIMAL SCIENCES 2015; 28:686-90. [PMID: 25924960 PMCID: PMC4412999 DOI: 10.5713/ajas.14.0573] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/29/2014] [Revised: 08/28/2014] [Accepted: 09/23/2014] [Indexed: 02/05/2023]
Abstract
The adrenergic receptor beta 2 (ADRB2) plays a role in various physiological responses of the muscle to exercise, such as contraction and relaxation. Given its important role in muscle function, we investigated the structure of the horse ADRB2 gene and its expression pattern after exercise to determine if it can serve as a putative biomarker for recovery. Evolutionary analyses using synonymous and non-synonymous mutation ratios, were compared with other species (human, chimpanzee, mouse, rat, cow, pig, chicken, dog, and cat), and revealed the occurrence of positive selection in the horse ADRB2 gene. In addition, expression analyses by quantitative polymerase chain reaction exhibited ubiquitous distribution of horse ADRB2 in various tissues including lung, skeletal muscle, kidney, thyroid, appendix, colon, spinal cord and heart, with the highest expression observed in the lung. The expression of ADRB2 in skeletal muscle was significantly up-regulated about four folds 30 minutes post-exercise compared to pre-exercise. The expression level of ADRB2 in leukocytes, which could be collected with convenience compared with other tissues in horse, increased until 60 min after exercise but decreased afterward until 120 min, suggesting the ADRB2 expression levels in leukocytes could be a useful biomarker to check the early recovery status of horse after exercise. In conclusion, we identified horse ADRB2 gene and analyzed expression profiles in various tissues. Additionally, analysis of ADBR2 gene expression in leukocytes could be a useful biomarker useful for evaluation of early recovery status after exercise in racing horses.
Collapse
Affiliation(s)
- Hyun-Woo Cho
- Department of Animal Science, College of Natural Resources and Life Sciences, Pusan National University, Miryang 627-702, Korea
| | - Sangsu Shin
- Life and Industry Convergence Research Institute, College of Natural Resources and Life Sciences, Pusan National University, Miryang 627-702, Korea
| | - Ki-Duk Song
- Genomic informatics Center, Hankyong National University, Anseong 456-749, Korea
| | - Jeong-Woong Park
- Department of Animal Science, College of Natural Resources and Life Sciences, Pusan National University, Miryang 627-702, Korea
| | - Jae-Young Choi
- Department of Animal Science, College of Natural Resources and Life Sciences, Pusan National University, Miryang 627-702, Korea
| | - Hak-Kyo Lee
- Genomic informatics Center, Hankyong National University, Anseong 456-749, Korea
| | - Byung-Wook Cho
- Department of Animal Science, College of Natural Resources and Life Sciences, Pusan National University, Miryang 627-702, Korea
| |
Collapse
|
28
|
Simplified data access on human skeletal muscle transcriptome responses to differentiated exercise. Sci Data 2014; 1:140041. [PMID: 25984345 PMCID: PMC4432635 DOI: 10.1038/sdata.2014.41] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2014] [Accepted: 09/30/2014] [Indexed: 11/25/2022] Open
Abstract
Few studies have investigated exercise-induced global gene expression responses in human skeletal muscle and these have typically focused at one specific mode of exercise and not implemented non-exercise control models. However, interpretation on effects of differentiated exercise necessitate direct comparison between essentially different modes of exercise and the ability to identify true exercise effect, necessitate implementation of independent non-exercise control subjects. Furthermore, muscle transcriptome data made available through previous exercise studies can be difficult to extract and interpret by individuals that are inexperienced with bioinformatics procedures. In a comparative study, we therefore; (1) investigated the human skeletal muscle transcriptome responses to differentiated exercise and non-exercise control intervention, and; (2) set out to develop a straightforward search tool to allow for easy access and interpretation of our data. We provide a simple-to-use spread sheet containing transcriptome data allowing other investigators to easily see how mRNA of their gene(s) of interest behave in skeletal muscle following exercise, both endurance, resistance and non-exercise, to better aid hypothesis-driven question in this field of research.
Collapse
|
29
|
Chaillou T, Jackson JR, England JH, Kirby TJ, Richards-White J, Esser KA, Dupont-Versteegden EE, McCarthy JJ. Identification of a conserved set of upregulated genes in mouse skeletal muscle hypertrophy and regrowth. J Appl Physiol (1985) 2014; 118:86-97. [PMID: 25554798 DOI: 10.1152/japplphysiol.00351.2014] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
The purpose of this study was to compare the gene expression profile of mouse skeletal muscle undergoing two forms of growth (hypertrophy and regrowth) with the goal of identifying a conserved set of differentially expressed genes. Expression profiling by microarray was performed on the plantaris muscle subjected to 1, 3, 5, 7, 10, and 14 days of hypertrophy or regrowth following 2 wk of hind-limb suspension. We identified 97 differentially expressed genes (≥2-fold increase or ≥50% decrease compared with control muscle) that were conserved during the two forms of muscle growth. The vast majority (∼90%) of the differentially expressed genes was upregulated and occurred at a single time point (64 out of 86 genes), which most often was on the first day of the time course. Microarray analysis from the conserved upregulated genes showed a set of genes related to contractile apparatus and stress response at day 1, including three genes involved in mechanotransduction and four genes encoding heat shock proteins. Our analysis further identified three cell cycle-related genes at day and several genes associated with extracellular matrix (ECM) at both days 3 and 10. In conclusion, we have identified a core set of genes commonly upregulated in two forms of muscle growth that could play a role in the maintenance of sarcomere stability, ECM remodeling, cell proliferation, fast-to-slow fiber type transition, and the regulation of skeletal muscle growth. These findings suggest conserved regulatory mechanisms involved in the adaptation of skeletal muscle to increased mechanical loading.
Collapse
Affiliation(s)
- Thomas Chaillou
- Center for Muscle Biology, University of Kentucky, Lexington, Kentucky; Department of Physiology, College of Medicine, University of Kentucky, Lexington, Kentucky
| | - Janna R Jackson
- Center for Muscle Biology, University of Kentucky, Lexington, Kentucky; Department of Physiology, College of Medicine, University of Kentucky, Lexington, Kentucky; Department of Rehabilitation Sciences, College of Health Sciences, University of Kentucky, Lexington, Kentucky
| | - Jonathan H England
- Center for Muscle Biology, University of Kentucky, Lexington, Kentucky; Department of Physiology, College of Medicine, University of Kentucky, Lexington, Kentucky
| | - Tyler J Kirby
- Center for Muscle Biology, University of Kentucky, Lexington, Kentucky; Department of Physiology, College of Medicine, University of Kentucky, Lexington, Kentucky; Department of Rehabilitation Sciences, College of Health Sciences, University of Kentucky, Lexington, Kentucky
| | - Jena Richards-White
- Department of Rehabilitation Sciences, College of Health Sciences, University of Kentucky, Lexington, Kentucky
| | - Karyn A Esser
- Center for Muscle Biology, University of Kentucky, Lexington, Kentucky; Department of Physiology, College of Medicine, University of Kentucky, Lexington, Kentucky
| | - Esther E Dupont-Versteegden
- Center for Muscle Biology, University of Kentucky, Lexington, Kentucky; Department of Physiology, College of Medicine, University of Kentucky, Lexington, Kentucky; Department of Rehabilitation Sciences, College of Health Sciences, University of Kentucky, Lexington, Kentucky
| | - John J McCarthy
- Center for Muscle Biology, University of Kentucky, Lexington, Kentucky; Department of Physiology, College of Medicine, University of Kentucky, Lexington, Kentucky;
| |
Collapse
|
30
|
Lamon S, Wallace MA, Russell AP. The STARS signaling pathway: a key regulator of skeletal muscle function. Pflugers Arch 2014; 466:1659-71. [DOI: 10.1007/s00424-014-1475-5] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2013] [Revised: 02/04/2014] [Accepted: 02/05/2014] [Indexed: 01/08/2023]
|
31
|
Neubauer O, Sabapathy S, Ashton KJ, Desbrow B, Peake JM, Lazarus R, Wessner B, Cameron-Smith D, Wagner KH, Haseler LJ, Bulmer AC. Time course-dependent changes in the transcriptome of human skeletal muscle during recovery from endurance exercise: from inflammation to adaptive remodeling. J Appl Physiol (1985) 2013; 116:274-87. [PMID: 24311745 DOI: 10.1152/japplphysiol.00909.2013] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Reprogramming of gene expression is fundamental for skeletal muscle adaptations in response to endurance exercise. This study investigated the time course-dependent changes in the muscular transcriptome after an endurance exercise trial consisting of 1 h of intense cycling immediately followed by 1 h of intense running. Skeletal muscle samples were taken at baseline, 3 h, 48 h, and 96 h postexercise from eight healthy, endurance-trained men. RNA was extracted from muscle. Differential gene expression was evaluated using Illumina microarrays and validated with qPCR. Gene set enrichment analysis identified enriched molecular signatures chosen from the Molecular Signatures Database. Three hours postexercise, 102 gene sets were upregulated [family wise error rate (FWER), P < 0.05], including groups of genes related with leukocyte migration, immune and chaperone activation, and cyclic AMP responsive element binding protein (CREB) 1 signaling. Forty-eight hours postexercise, among 19 enriched gene sets (FWER, P < 0.05), two gene sets related to actin cytoskeleton remodeling were upregulated. Ninety-six hours postexercise, 83 gene sets were enriched (FWER, P < 0.05), 80 of which were upregulated, including gene groups related to chemokine signaling, cell stress management, and extracellular matrix remodeling. These data provide comprehensive insights into the molecular pathways involved in acute stress, recovery, and adaptive muscular responses to endurance exercise. The novel 96 h postexercise transcriptome indicates substantial transcriptional activity potentially associated with the prolonged presence of leukocytes in the muscles. This suggests that muscular recovery, from a transcriptional perspective, is incomplete 96 h after endurance exercise involving muscle damage.
Collapse
Affiliation(s)
- Oliver Neubauer
- Emerging Field Oxidative Stress and DNA Stability, Research Platform Active Aging, and Department of Nutritional Sciences, University of Vienna, Austria
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Scheler M, Irmler M, Lehr S, Hartwig S, Staiger H, Al-Hasani H, Beckers J, Hrabé de Angelis M, Häring HU, Weigert C. Cytokine response of primary human myotubes in an in vitro exercise model. Am J Physiol Cell Physiol 2013; 305:C877-86. [DOI: 10.1152/ajpcell.00043.2013] [Citation(s) in RCA: 94] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Muscle contraction during exercise is a major stimulus for the release of peptides and proteins (myokines) that are supposed to take part in the beneficial adaptation to exercise. We hypothesize that application of an in vitro exercise stimulus as electric pulse stimulation (EPS) to human myotubes enables the investigation of the molecular response to exercise in a clearly defined model. We applied EPS for 24 h to primary human myotubes and studied the whole genome-wide transcriptional response as well as the release of candidate myokines. We observed 183 differentially regulated transcripts with fold changes >1.3. The transcriptional response resembles several properties of the in vivo situation in the skeletal muscle after endurance exercise, namely significant enrichment of pathways associated with interleukin and chemokine signaling, lipid metabolism, and antioxidant defense. Multiplex immunoassays verified the translation of the transcriptional response of several cytokines into high-secretion levels (IL-6, IL-8, CXCL1, LIF, CSF3, IL-1B, and TNF) and the increased secretion of further myokines such as angiopoietin-like 4. Notably, EPS did not induce the release of creatine kinase. Inhibitor studies and immunoblotting revealed the participation of ERK1/2-, JNK-, and NF-κB-dependent pathways in the upregulation of myokines. To conclude, our data highlight the importance of skeletal muscle cells as endocrine cells. This in vitro exercise model is not only suitable to identify exercise-regulated myokines, but it might be applied to primary human myotubes obtained from different muscle biopsy donors to study the molecular mechanisms of the individual response to exercise.
Collapse
Affiliation(s)
- Mika Scheler
- Institute of Experimental Genetics, Helmholtz Zentrum Muenchen, German Research Center for Environmental Health, Neuherberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany; and
| | - Martin Irmler
- Institute of Experimental Genetics, Helmholtz Zentrum Muenchen, German Research Center for Environmental Health, Neuherberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany; and
| | - Stefan Lehr
- Institute of Clinical Biochemistry and Pathobiochemistry, German Diabetes Center, Duesseldorf, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany; and
| | - Sonja Hartwig
- Institute of Clinical Biochemistry and Pathobiochemistry, German Diabetes Center, Duesseldorf, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany; and
| | - Harald Staiger
- Division of Endocrinology, Diabetology, Angiology, Nephrology, Pathobiochemistry and Clinical Chemistry, Department of Internal Medicine, University Tuebingen, Tuebingen, Germany
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Zentrum Muenchen at the University of Tuebingen, Tuebingen, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany; and
| | - Hadi Al-Hasani
- Institute of Clinical Biochemistry and Pathobiochemistry, German Diabetes Center, Duesseldorf, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany; and
| | - Johannes Beckers
- Institute of Experimental Genetics, Helmholtz Zentrum Muenchen, German Research Center for Environmental Health, Neuherberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany; and
- Chair of Experimental Genetics, Center of Life and Food Sciences Weihenstephan, Technical University Muenchen, Freising-Weihenstephan, Germany
| | - Martin Hrabé de Angelis
- Institute of Experimental Genetics, Helmholtz Zentrum Muenchen, German Research Center for Environmental Health, Neuherberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany; and
- Chair of Experimental Genetics, Center of Life and Food Sciences Weihenstephan, Technical University Muenchen, Freising-Weihenstephan, Germany
| | - Hans-Ulrich Häring
- Division of Endocrinology, Diabetology, Angiology, Nephrology, Pathobiochemistry and Clinical Chemistry, Department of Internal Medicine, University Tuebingen, Tuebingen, Germany
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Zentrum Muenchen at the University of Tuebingen, Tuebingen, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany; and
| | - Cora Weigert
- Division of Endocrinology, Diabetology, Angiology, Nephrology, Pathobiochemistry and Clinical Chemistry, Department of Internal Medicine, University Tuebingen, Tuebingen, Germany
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Zentrum Muenchen at the University of Tuebingen, Tuebingen, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany; and
| |
Collapse
|
33
|
Chaillou T, Lee JD, England JH, Esser KA, McCarthy JJ. Time course of gene expression during mouse skeletal muscle hypertrophy. J Appl Physiol (1985) 2013; 115:1065-74. [PMID: 23869057 DOI: 10.1152/japplphysiol.00611.2013] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
The purpose of this study was to perform a comprehensive transcriptome analysis during skeletal muscle hypertrophy to identify signaling pathways that are operative throughout the hypertrophic response. Global gene expression patterns were determined from microarray results on days 1, 3, 5, 7, 10, and 14 during plantaris muscle hypertrophy induced by synergist ablation in adult mice. Principal component analysis and the number of differentially expressed genes (cutoffs ≥2-fold increase or ≥50% decrease compared with control muscle) revealed three gene expression patterns during overload-induced hypertrophy: early (1 day), intermediate (3, 5, and 7 days), and late (10 and 14 days) patterns. Based on the robust changes in total RNA content and in the number of differentially expressed genes, we focused our attention on the intermediate gene expression pattern. Ingenuity Pathway Analysis revealed a downregulation of genes encoding components of the branched-chain amino acid degradation pathway during hypertrophy. Among these genes, five were predicted by Ingenuity Pathway Analysis or previously shown to be regulated by the transcription factor Kruppel-like factor-15, which was also downregulated during hypertrophy. Moreover, the integrin-linked kinase signaling pathway was activated during hypertrophy, and the downregulation of muscle-specific micro-RNA-1 correlated with the upregulation of five predicted targets associated with the integrin-linked kinase pathway. In conclusion, we identified two novel pathways that may be involved in muscle hypertrophy, as well as two upstream regulators (Kruppel-like factor-15 and micro-RNA-1) that provide targets for future studies investigating the importance of these pathways in muscle hypertrophy.
Collapse
Affiliation(s)
- Thomas Chaillou
- Center for Muscle Biology, University of Kentucky, Lexington, Kentucky
| | | | | | | | | |
Collapse
|
34
|
Vissing K, Rahbek SK, Lamon S, Farup J, Stefanetti RJ, Wallace MA, Vendelbo MH, Russell A. Effect of resistance exercise contraction mode and protein supplementation on members of the STARS signalling pathway. J Physiol 2013; 591:3749-63. [PMID: 23753523 DOI: 10.1113/jphysiol.2012.249755] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
The striated muscle activator of Rho signalling (STARS) pathway is suggested to provide a link between external stress responses and transcriptional regulation in muscle. However, the sensitivity of STARS signalling to different mechanical stresses has not been investigated. In a comparative study, we examined the regulation of the STARS signalling pathway in response to unilateral resistance exercise performed as either eccentric (ECC) or concentric (CONC) contractions as well as prolonged training; with and without whey protein supplementation. Skeletal muscle STARS, myocardian-related transcription factor-A (MRTF-A) and serum response factor (SRF) mRNA and protein, as well as muscle cross-sectional area and maximal voluntary contraction, were measured. A single-bout of exercise produced increases in STARS and SRF mRNA and decreases in MRTF-A mRNA with both ECC and CONC exercise, but with an enhanced response occurring following ECC exercise. A 31% increase in STARS protein was observed exclusively after CONC exercise (P < 0.001), while pSRF protein levels increased similarly by 48% with both CONC and ECC exercise (P < 0.001). Prolonged ECC and CONC training equally stimulated muscle hypertrophy and produced increases in MRTF-A protein of 125% and 99%, respectively (P < 0.001). No changes occurred for total SRF protein. There was no effect of whey protein supplementation. These results show that resistance exercise provides an acute stimulation of the STARS pathway that is contraction mode dependent. The responses to acute exercise were more pronounced than responses to accumulated training, suggesting that STARS signalling is primarily involved in the initial phase of exercise-induced muscle adaptations.
Collapse
Affiliation(s)
- Kristian Vissing
- Section of Sport Science, Department of Public Health, Aarhus University, Dalgas Avenue 4, DK-8000 Aarhus C, Denmark.
| | | | | | | | | | | | | | | |
Collapse
|
35
|
Lamon S, Wallace MA, Stefanetti RJ, Rahbek SK, Vendelbo MH, Russell AP, Vissing K. Regulation of the STARS signaling pathway in response to endurance and resistance exercise and training. Pflugers Arch 2013; 465:1317-25. [DOI: 10.1007/s00424-013-1265-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2013] [Revised: 03/07/2013] [Accepted: 03/08/2013] [Indexed: 11/27/2022]
|
36
|
Wallace MA, Lamon S, Russell AP. The regulation and function of the striated muscle activator of rho signaling (STARS) protein. Front Physiol 2012; 3:469. [PMID: 23248604 PMCID: PMC3520124 DOI: 10.3389/fphys.2012.00469] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2012] [Accepted: 11/28/2012] [Indexed: 11/15/2022] Open
Abstract
Healthy living throughout the lifespan requires continual growth and repair of cardiac, smooth, and skeletal muscle. To effectively maintain these processes muscle cells detect extracellular stress signals and efficiently transmit them to activate appropriate intracellular transcriptional programs. The striated muscle activator of Rho signaling (STARS) protein, also known as Myocyte Stress-1 (MS1) protein and Actin-binding Rho-activating protein (ABRA) is highly enriched in cardiac, skeletal, and smooth muscle. STARS binds actin, co-localizes to the sarcomere and is able to stabilize the actin cytoskeleton. By regulating actin polymerization, STARS also controls an intracellular signaling cascade that stimulates the serum response factor (SRF) transcriptional pathway; a pathway controlling genes involved in muscle cell proliferation, differentiation, and growth. Understanding the activation, transcriptional control and biological roles of STARS in cardiac, smooth, and skeletal muscle, will improve our understanding of physiological and pathophysiological muscle development and function.
Collapse
Affiliation(s)
- Marita A Wallace
- Centre for Physical Activity and Nutrition Research, School of Exercise and Nutrition Sciences, Deakin University Burwood, VIC, Australia
| | | | | |
Collapse
|
37
|
Schoenfeld BJ. Does Exercise-Induced Muscle Damage Play a Role in Skeletal Muscle Hypertrophy? J Strength Cond Res 2012; 26:1441-53. [DOI: 10.1519/jsc.0b013e31824f207e] [Citation(s) in RCA: 124] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
38
|
Ho TJ, Huang CC, Huang CY, Lin WT. Fasudil, a Rho-kinase inhibitor, protects against excessive endurance exercise training-induced cardiac hypertrophy, apoptosis and fibrosis in rats. Eur J Appl Physiol 2011; 112:2943-55. [PMID: 22160250 DOI: 10.1007/s00421-011-2270-z] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2011] [Accepted: 11/28/2011] [Indexed: 01/08/2023]
Abstract
Excessive endurance exercise training (EEET) is accompanied by cardiac remodeling, changes in ventricular function and increased heart failure risk. Fasudil, a potent Rho-kinase inhibitor, has been demonstrated to blunt cardiomyocyte hypertrophy, cardiac remodeling, and heart failure progression in pre-clinical trials and has been approved for clinical use in Japan. We examined the in vivo bioefficacy of fasudil against EEET-induced cardiac remodeling and the underlying molecular mechanisms. Male Sprague-Dawley rats were randomly divided into three groups: sedentary control (SC), EEET, and EEET with fasudil treatment (EEET-F). Rats in EEET and EEET-F groups ran on a motorized treadmill for 12 weeks. The results revealed that EEET increased myocardial hypertrophy (LV weight/tibial length), myocyte cross-sectional area, hypertrophy-related pathways (IL6/STAT3-MEK5-ERK5, calcineurin-NFATc3, p38 and JNK MAPK), hypertrophic markers (ANP/BNP), pro-apoptotic molecules (cytochrome C, cleaved caspase-3 and PARP), and fibrosis-related pathways (FGF-2-ERK1/2) and fibrosis markers (uPA, MMP-9 and -2). These pathways were then expressed lower in the EEET-F group when compared with the EEET group. The cardiac hypertrophic level, apoptotic pathway and fibrosis signaling were further inhibited in the fasudil-treated group. We systematically investigated the possible signaling pathways leading to EEET-induced cardiac hypertrophy, apoptosis and fibrosis. We also provide evidence for the novel function of fasudil in suppressing EEET-induced cardiac remodeling and impairment by multiple mechanisms, which suggests that the RhoA signaling pathway contributes to EEET-induced cardiac remodeling and dysfunction.
Collapse
Affiliation(s)
- Tsung-Jung Ho
- School of Chinese Medicine, College of Chinese Medicine, China Medical University, Taichung, 40402, Taiwan, ROC
| | | | | | | |
Collapse
|
39
|
Tarnopolsky MA, Pearce E, Smith K, Lach B. Suction-modified Bergström muscle biopsy technique: experience with 13,500 procedures. Muscle Nerve 2011; 43:717-25. [PMID: 21462204 DOI: 10.1002/mus.21945] [Citation(s) in RCA: 165] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/13/2010] [Indexed: 11/05/2022]
Abstract
INTRODUCTION Bergström needle muscle biopsies have been used by exercise physiologists for over 35 years but have been less accepted by neuromuscular clinicians due to size concerns. METHODS We retrospectively reviewed over 13,500 muscle Bergström needle biopsies done over a 21-year period to determine sampling success, patient/subject experience, and complications. We compared sample yield between two different needles (Bergström vs. UCH), with and without suction modifications. RESULTS Needle biopsies adequate for histology and enzymology were obtainable from the vastus lateralis, deltoid, biceps brachii, soleus, and medial gastrocnemius muscles, with a success rate of >99.9% and a minor complication rate of 0.15%. Approximately 450 muscle fibers were submitted for histologic assessment; suction modification and use of the Bergström vs. UCH needle were associated with larger sample size (P < 0.05). CONCLUSIONS The suction-modified Bergström needle muscle biopsy technique is safe and provides an adequate sample size for histologic, ultrastructural, DNA, and enzyme analysis.
Collapse
Affiliation(s)
- Mark A Tarnopolsky
- Department of Pediatrics, McMaster University, 1200 Main Street W, HSC-2H26, Hamilton, Ontario L8N 3Z5, Canada.
| | | | | | | |
Collapse
|
40
|
|