1
|
Bastianello G, Kidiyoor GR, Lowndes C, Li Q, Bonnal R, Godwin J, Iannelli F, Drufuca L, Bason R, Orsenigo F, Parazzoli D, Pavani M, Cancila V, Piccolo S, Scita G, Ciliberto A, Tripodo C, Pagani M, Foiani M. Mechanical stress during confined migration causes aberrant mitoses and c-MYC amplification. Proc Natl Acad Sci U S A 2024; 121:e2404551121. [PMID: 38990945 PMCID: PMC11260125 DOI: 10.1073/pnas.2404551121] [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: 03/04/2024] [Accepted: 06/07/2024] [Indexed: 07/13/2024] Open
Abstract
Confined cell migration hampers genome integrity and activates the ATR and ATM mechano-transduction pathways. We investigated whether the mechanical stress generated by metastatic interstitial migration contributes to the enhanced chromosomal instability observed in metastatic tumor cells. We employed live cell imaging, micro-fluidic approaches, and scRNA-seq to follow the fate of tumor cells experiencing confined migration. We found that, despite functional ATR, ATM, and spindle assembly checkpoint (SAC) pathways, tumor cells dividing across constriction frequently exhibited altered spindle pole organization, chromosome mis-segregations, micronuclei formation, chromosome fragility, high gene copy number variation, and transcriptional de-regulation and up-regulation of c-MYC oncogenic transcriptional signature via c-MYC locus amplifications. In vivo tumor settings showed that malignant cells populating metastatic foci or infiltrating the interstitial stroma gave rise to cells expressing high levels of c-MYC. Altogether, our data suggest that mechanical stress during metastatic migration contributes to override the checkpoint controls and boosts genotoxic and oncogenic events. Our findings may explain why cancer aneuploidy often does not correlate with mutations in SAC genes and why c-MYC amplification is strongly linked to metastatic tumors.
Collapse
Affiliation(s)
- Giulia Bastianello
- Istituto Fondazione Italiana per la Ricerca sul Cancro di Oncologia molecolare—the Associazione Italiana per la Ricerca sul Cancro Institute of Molecular Oncology, Milano20139, Italy
- Università degli Studi di Milano, Milan20122, Italy
| | - Gururaj Rao Kidiyoor
- Istituto Fondazione Italiana per la Ricerca sul Cancro di Oncologia molecolare—the Associazione Italiana per la Ricerca sul Cancro Institute of Molecular Oncology, Milano20139, Italy
| | - Conor Lowndes
- Istituto Fondazione Italiana per la Ricerca sul Cancro di Oncologia molecolare—the Associazione Italiana per la Ricerca sul Cancro Institute of Molecular Oncology, Milano20139, Italy
| | - Qingsen Li
- Istituto Fondazione Italiana per la Ricerca sul Cancro di Oncologia molecolare—the Associazione Italiana per la Ricerca sul Cancro Institute of Molecular Oncology, Milano20139, Italy
| | - Raoul Bonnal
- Istituto Fondazione Italiana per la Ricerca sul Cancro di Oncologia molecolare—the Associazione Italiana per la Ricerca sul Cancro Institute of Molecular Oncology, Milano20139, Italy
| | - Jeffrey Godwin
- Istituto Fondazione Italiana per la Ricerca sul Cancro di Oncologia molecolare—the Associazione Italiana per la Ricerca sul Cancro Institute of Molecular Oncology, Milano20139, Italy
| | - Fabio Iannelli
- Istituto Fondazione Italiana per la Ricerca sul Cancro di Oncologia molecolare—the Associazione Italiana per la Ricerca sul Cancro Institute of Molecular Oncology, Milano20139, Italy
| | | | - Ramona Bason
- Istituto Fondazione Italiana per la Ricerca sul Cancro di Oncologia molecolare—the Associazione Italiana per la Ricerca sul Cancro Institute of Molecular Oncology, Milano20139, Italy
| | - Fabrizio Orsenigo
- Istituto Fondazione Italiana per la Ricerca sul Cancro di Oncologia molecolare—the Associazione Italiana per la Ricerca sul Cancro Institute of Molecular Oncology, Milano20139, Italy
| | - Dario Parazzoli
- Istituto Fondazione Italiana per la Ricerca sul Cancro di Oncologia molecolare—the Associazione Italiana per la Ricerca sul Cancro Institute of Molecular Oncology, Milano20139, Italy
| | - Mattia Pavani
- Istituto Fondazione Italiana per la Ricerca sul Cancro di Oncologia molecolare—the Associazione Italiana per la Ricerca sul Cancro Institute of Molecular Oncology, Milano20139, Italy
| | - Valeria Cancila
- Tumor Immunology Unit, Department of Health Science, University of Palermo School of Medicine, Palermo90133, Italy
| | - Stefano Piccolo
- Istituto Fondazione Italiana per la Ricerca sul Cancro di Oncologia molecolare—the Associazione Italiana per la Ricerca sul Cancro Institute of Molecular Oncology, Milano20139, Italy
- Department of Molecular Medicine, University of Padua, Padua35123, Italy
| | - Giorgio Scita
- Istituto Fondazione Italiana per la Ricerca sul Cancro di Oncologia molecolare—the Associazione Italiana per la Ricerca sul Cancro Institute of Molecular Oncology, Milano20139, Italy
- Università degli Studi di Milano, Milan20122, Italy
| | - Andrea Ciliberto
- Istituto Fondazione Italiana per la Ricerca sul Cancro di Oncologia molecolare—the Associazione Italiana per la Ricerca sul Cancro Institute of Molecular Oncology, Milano20139, Italy
| | - Claudio Tripodo
- Istituto Fondazione Italiana per la Ricerca sul Cancro di Oncologia molecolare—the Associazione Italiana per la Ricerca sul Cancro Institute of Molecular Oncology, Milano20139, Italy
- Tumor Immunology Unit, Department of Health Science, University of Palermo School of Medicine, Palermo90133, Italy
| | - Massimiliano Pagani
- Istituto Fondazione Italiana per la Ricerca sul Cancro di Oncologia molecolare—the Associazione Italiana per la Ricerca sul Cancro Institute of Molecular Oncology, Milano20139, Italy
- Università degli Studi di Milano, Milan20122, Italy
| | - Marco Foiani
- Istituto Fondazione Italiana per la Ricerca sul Cancro di Oncologia molecolare—the Associazione Italiana per la Ricerca sul Cancro Institute of Molecular Oncology, Milano20139, Italy
- Istituto di Genetica Molecolare, Centro Nazionale Ricerca, Pavia27100, Italy
- Cancer Science Institute of Singapore, National University of Singapore, Singapore117599, Singapore
| |
Collapse
|
2
|
Kanzaki K, Wada M. Effects of Leucine Ingestion and Contraction on the Sestrin/GATOR2 Pathway and mTORC1 Activation in Rat Fast-Twitch muscle. J Nutr 2023; 153:2228-2236. [PMID: 37328110 DOI: 10.1016/j.tjnut.2023.06.011] [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: 04/02/2023] [Revised: 06/05/2023] [Accepted: 06/13/2023] [Indexed: 06/18/2023] Open
Abstract
BACKGROUND Leucine activates the mechanistic/mammalian target of rapamycin complex 1 (mTORC1) in mammalian skeletal muscle. Recent studies have shown that Sestrin, a leucine sensor, might play a role in this process. However, it remains unknown whether Sestrin dissociates from GATOR2 in a dose- and time-dependent manner and whether an acute bout of muscle contraction augments this dissociation. OBJECTIVE This study aimed to examine the effects of leucine ingestion and muscle contraction on the interaction between Sestrin1/2 and GATOR2 and on mTORC1 activation. METHODS Male Wistar rats were randomly assigned to control (C), leucine 3 (L3), or leucine 10 (L10) groups. Intact gastrocnemius muscles were subjected to 30 repetitive unilateral contractions. The L3 and L10 groups were then orally administered 3 and 10 mmol/kg body weight of L-leucine 2 h after the end of the contractions, respectively. Blood and muscle samples were collected 30, 60, or 120 min after the administration. RESULTS The blood and muscle leucine concentrations increased in a dose-dependent manner. The ratio of phosphorylated ribosomal protein S6 kinase (S6K) to total S6K (which indicates mTORC1 signaling activation) was markedly increased by muscle contraction and increased in a dose-dependent manner only in rested muscle. Leucine ingestion but not muscle contraction increased Sestrin1 dissociation from GATOR2 and Sestrin2 association with GATOR2. A negative relationship was observed between the blood and muscle leucine concentrations and the Sestrin1 association with GATOR2. CONCLUSIONS The results suggest that Sestrin1, but not Sestrin2, regulates leucine-related mTORC1 activation via its dissociation from GATOR2 and that acute exercise-induced mTORC1 activation involves pathways other than the leucine-related Sestrin1/GATOR2 pathway.
Collapse
Affiliation(s)
- Keita Kanzaki
- Department of Clinical Nutrition, Faculty of Health Science and Technology, Kawasaki University of Medical Welfare, Okayama, Japan.
| | - Masanobu Wada
- Graduate School of Humanities and Social Sciences, Hiroshima University, Hiroshima, Japan
| |
Collapse
|
3
|
Bersiner K, Park SY, Schaaf K, Yang WH, Theis C, Jacko D, Gehlert S. Resistance exercise: a mighty tool that adapts, destroys, rebuilds and modulates the molecular and structural environment of skeletal muscle. Phys Act Nutr 2023; 27:78-95. [PMID: 37583075 PMCID: PMC10440184 DOI: 10.20463/pan.2023.0021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 06/29/2023] [Accepted: 06/30/2023] [Indexed: 08/17/2023] Open
Abstract
PURPOSE Skeletal muscle regulates health and performance by maintaining or increasing strength and muscle mass. Although the molecular mechanisms in response to resistance exercise (RE) significantly target the activation of protein synthesis, a plethora of other mechanisms and structures must be involved in orchestrating the communication, repair, and restoration of homeostasis after RE stimulation. In practice, RE can be modulated by variations in intensity, continuity and volume, which affect molecular responses and skeletal muscle adaptation. Knowledge of these aspects is important with respect to planning of training programs and assessing the impact of RE training on skeletal muscle. METHODS In this narrative review, we introduce general aspects of skeletal muscle substructures that adapt in response to RE. We further highlighted the molecular mechanisms that control human skeletal muscle anabolism, degradation, repair and memory in response to acute and repeated RE and linked these aspects to major training variables. RESULTS Although RE is a key stimulus for the activation of skeletal muscle anabolism, it also induces myofibrillar damage. Nevertheless, to increase muscle mass accompanied by a corresponding adaptation of the essential substructures of the sarcomeric environment, RE must be continuously repeated. This requires the permanent engagement of molecular mechanisms that re-establish skeletal muscle integrity after each RE-induced muscle damage. CONCLUSION Various molecular regulators coordinately control the adaptation of skeletal muscle after acute and repeated RE and expand their actions far beyond muscle growth. Variations of key resistance training variables likely affect these mechanisms without affecting muscle growth.
Collapse
Affiliation(s)
- Käthe Bersiner
- Department for Biosciences of Sports, Institute of Sports Science, University of Hildesheim, Hildesheim, Germany
| | - So-Young Park
- Graduate School of Sports Medicine, CHA University, Pocheon, Republic of Korea
| | - Kirill Schaaf
- Department of Molecular and Cellular Sports Medicine, Institute of Cardiovascular Research and Sports Medicine, German Sport University Cologne, Cologne, Germany
| | - Woo-Hwi Yang
- Graduate School of Sports Medicine, CHA University, Pocheon, Republic of Korea
- Department of Medicine, General Graduate School, CHA University, Pocheon, Republic of Korea
| | - Christian Theis
- Center for Anaesthesiology, Helios University Hospital Wuppertal, Wuppertal, Germany
| | - Daniel Jacko
- Department of Molecular and Cellular Sports Medicine, Institute of Cardiovascular Research and Sports Medicine, German Sport University Cologne, Cologne, Germany
| | - Sebastian Gehlert
- Department for Biosciences of Sports, Institute of Sports Science, University of Hildesheim, Hildesheim, Germany
| |
Collapse
|
4
|
Alcohol, Resistance Exercise, and mTOR Pathway Signaling: An Evidence-Based Narrative Review. Biomolecules 2022; 13:biom13010002. [PMID: 36671386 PMCID: PMC9855961 DOI: 10.3390/biom13010002] [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: 11/17/2022] [Revised: 12/14/2022] [Accepted: 12/17/2022] [Indexed: 12/24/2022] Open
Abstract
Skeletal muscle mass is determined by the balance between muscle protein synthesis (MPS) and degradation. Several intracellular signaling pathways control this balance, including mammalian/mechanistic target of rapamycin (mTOR) complex 1 (C1). Activation of this pathway in skeletal muscle is controlled, in part, by nutrition (e.g., amino acids and alcohol) and exercise (e.g., resistance exercise (RE)). Acute and chronic alcohol use can result in myopathy, and evidence points to altered mTORC1 signaling as a contributing factor. Moreover, individuals who regularly perform RE or vigorous aerobic exercise are more likely to use alcohol frequently and in larger quantities. Therefore, alcohol may antagonize beneficial exercise-induced increases in mTORC1 pathway signaling. The purpose of this review is to synthesize up-to-date evidence regarding mTORC1 pathway signaling and the independent and combined effects of acute alcohol and RE on activation of the mTORC1 pathway. Overall, acute alcohol impairs and RE activates mTORC1 pathway signaling; however, effects vary by model, sex, feeding, training status, quantity, etc., such that anabolic stimuli may partially rescue the alcohol-mediated pathway inhibition. Likewise, the impact of alcohol on RE-induced mTORC1 pathway signaling appears dependent on several factors including nutrition and sex, although many questions remain unanswered. Accordingly, we identify gaps in the literature that remain to be elucidated to fully understand the independent and combined impacts of alcohol and RE on mTORC1 pathway signaling.
Collapse
|
5
|
Jacko D, Schaaf K, Masur L, Windoffer H, Aussieker T, Schiffer T, Zacher J, Bloch W, Gehlert S. Repeated and Interrupted Resistance Exercise Induces the Desensitization and Re-Sensitization of mTOR-Related Signaling in Human Skeletal Muscle Fibers. Int J Mol Sci 2022; 23:ijms23105431. [PMID: 35628242 PMCID: PMC9141560 DOI: 10.3390/ijms23105431] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 05/07/2022] [Accepted: 05/09/2022] [Indexed: 02/06/2023] Open
Abstract
The acute resistance exercise (RE)-induced phosphorylation of mTOR-related signaling proteins in skeletal muscle can be blunted after repeated RE. The time frame in which the phosphorylation (p) of mTORS2448, p70S6kT421/S424, and rpS6S235/236 will be reduced during an RE training period in humans and whether progressive (PR) loading can counteract such a decline has not been described. (1) To enclose the time frame in which pmTORS2448, prpS6S235/236, and pp70S6kT421/S424 are acutely reduced after RE occurs during repeated RE. (2) To test whether PR will prevent that reduction compared to constant loading (CO) and (3) whether 10 days without RE may re-increase blunted signaling. Fourteen healthy males (24 ± 2.8 yrs.; 1.83 ± 0.1 cm; 79.3 ± 8.5 kg) were subjected to RE with either PR (n = 8) or CO (n = 6) loading. Subjects performed RE thrice per week, conducting three sets with 10−12 repetitions on a leg press and leg extension machine. Muscle biopsies were collected at rest (T0), 45 min after the first (T1), seventh (T7), 13th (T13), and 14th (X-T14) RE session. No differences were found between PR and CO for any parameter. Thus, the groups were combined, and the results show the merged values. prpS6S235/236 and pp70s6kT421/S424 were increased at T1, but were already reduced at T7 and up to T13 compared to T1. Ten days without RE re-increased prpS6S235/236 and pp70S6kT421/S424 at X-T14 to a level comparable to that of T1. pmTORS2448 was increased from T1 to X-T14 and did not decline over the training period. Single-fiber immunohistochemistry revealed a reduction in prpS6S235/236 in type I fibers from T1 to T13 and a re-increase at X-T14, which was more augmented in type II fibers at T13 (p < 0.05). The entity of myofibers revealed a high heterogeneity in the level of prpS6S235/236, possibly reflecting individual contraction-induced stress during RE. The type I and II myofiber diameter increased from T0 and T1 to T13 and X-T14 (p < 0.05) prpS6S235/236 and pp70s6kT421/S424 reflect RE-induced states of desensitization and re-sensitization in dependency on frequent loading by RE, but also by its cessation.
Collapse
Affiliation(s)
- Daniel Jacko
- Department of Molecular and Cellular Sports Medicine, Institute of Cardiovascular Research and Sports Medicine, German Sport University Cologne, 50933 Cologne, Germany; (D.J.); (K.S.); (L.M.); (H.W.); (T.A.); (W.B.)
- Olympic Base Center NRW/Rhineland, 50933 Cologne, Germany
| | - Kirill Schaaf
- Department of Molecular and Cellular Sports Medicine, Institute of Cardiovascular Research and Sports Medicine, German Sport University Cologne, 50933 Cologne, Germany; (D.J.); (K.S.); (L.M.); (H.W.); (T.A.); (W.B.)
| | - Lukas Masur
- Department of Molecular and Cellular Sports Medicine, Institute of Cardiovascular Research and Sports Medicine, German Sport University Cologne, 50933 Cologne, Germany; (D.J.); (K.S.); (L.M.); (H.W.); (T.A.); (W.B.)
| | - Hannes Windoffer
- Department of Molecular and Cellular Sports Medicine, Institute of Cardiovascular Research and Sports Medicine, German Sport University Cologne, 50933 Cologne, Germany; (D.J.); (K.S.); (L.M.); (H.W.); (T.A.); (W.B.)
| | - Thorben Aussieker
- Department of Molecular and Cellular Sports Medicine, Institute of Cardiovascular Research and Sports Medicine, German Sport University Cologne, 50933 Cologne, Germany; (D.J.); (K.S.); (L.M.); (H.W.); (T.A.); (W.B.)
| | - Thorsten Schiffer
- Outpatient Clinic for Sports Traumatology and Public Health Consultation, German Sport University Cologne, 50933 Cologne, Germany;
| | - Jonas Zacher
- Department ofPreventative and Rehabilitative Sports and Performance Medicine, Institute of Cardiology and Sports Medicine, German Sports University Cologne, 50933 Cologne, Germany;
| | - Wilhelm Bloch
- Department of Molecular and Cellular Sports Medicine, Institute of Cardiovascular Research and Sports Medicine, German Sport University Cologne, 50933 Cologne, Germany; (D.J.); (K.S.); (L.M.); (H.W.); (T.A.); (W.B.)
- German Research Centre of Elite Sport (Momentum), German Sport University Cologne, 50933 Cologne, Germany
| | - Sebastian Gehlert
- Department of Molecular and Cellular Sports Medicine, Institute of Cardiovascular Research and Sports Medicine, German Sport University Cologne, 50933 Cologne, Germany; (D.J.); (K.S.); (L.M.); (H.W.); (T.A.); (W.B.)
- Institute of Sport Science, Biosciences of Sports, University of Hildesheim, 31141 Hildesheim, Germany
- Correspondence: ; Tel.: +49-(0)-5121-883-580; Fax: +49-(0)-5121-883-591
| |
Collapse
|
6
|
Gargalionis AN, Papavassiliou KA, Basdra EK, Papavassiliou AG. mTOR Signaling Components in Tumor Mechanobiology. Int J Mol Sci 2022; 23:1825. [PMID: 35163745 PMCID: PMC8837098 DOI: 10.3390/ijms23031825] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 02/02/2022] [Accepted: 02/03/2022] [Indexed: 11/17/2022] Open
Abstract
Mechanistic target of rapamycin (mTOR) is a central signaling hub that integrates networks of nutrient availability, cellular metabolism, and autophagy in eukaryotic cells. mTOR kinase, along with its upstream regulators and downstream substrates, is upregulated in most human malignancies. At the same time, mechanical forces from the tumor microenvironment and mechanotransduction promote cancer cells' proliferation, motility, and invasion. mTOR signaling pathway has been recently found on the crossroads of mechanoresponsive-induced signaling cascades to regulate cell growth, invasion, and metastasis in cancer cells. In this review, we examine the emerging association of mTOR signaling components with certain protein tools of tumor mechanobiology. Thereby, we highlight novel mechanisms of mechanotransduction, which regulate tumor progression and invasion, as well as mechanisms related to the therapeutic efficacy of antitumor drugs.
Collapse
Affiliation(s)
- Antonios N. Gargalionis
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (K.A.P.); (E.K.B.)
- Department of Biopathology, Aeginition Hospital, Medical School, National and Kapodistrian University of Athens, 11528 Athens, Greece
| | - Kostas A. Papavassiliou
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (K.A.P.); (E.K.B.)
| | - Efthimia K. Basdra
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (K.A.P.); (E.K.B.)
| | - Athanasios G. Papavassiliou
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (K.A.P.); (E.K.B.)
| |
Collapse
|
7
|
Miyazaki M, Moriya N, Takemasa T. Transient activation of mTORC1 signaling in skeletal muscle is independent of Akt1 regulation. Physiol Rep 2021; 8:e14599. [PMID: 33038070 PMCID: PMC7547586 DOI: 10.14814/phy2.14599] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2020] [Revised: 09/08/2020] [Accepted: 09/09/2020] [Indexed: 12/17/2022] Open
Abstract
The regulation of cellular protein synthesis is a critical determinant of skeletal muscle growth and hypertrophy in response to an increased workload such as resistance exercise. The mechanistic target of rapamycin complex 1 (mTORC1) and its upstream protein kinase Akt1 have been implicated as a central signaling pathway that regulates protein synthesis in the skeletal muscle; however, the precise molecular regulation of mTORC1 activity is largely unknown. This study employed germline Akt1 knockout (KO) mice to examine whether upstream Akt1 regulation is necessary for the acute activation of mTORC1 signaling in the plantaris muscle following mechanical overload. The phosphorylation states of S6 kinase 1, ribosomal protein S6, and eukaryotic translation initiation factor 4E‐binding protein 1 which show the functional activity of mTORC1 signaling, were significantly increased in the skeletal muscle of both wildtype and Akt1 KO mice following an acute bout (3 and 12 hr) of mechanical overload. Akt1 deficiency did not affect load‐induced alteration of insulin‐like growth factor‐1 (IGF‐1)/IGF receptor mRNA expression. Also, no effect of Akt1 deficiency was observed on the overload‐induced increase in the gene expressions of pax7 and myogenic regulatory factor of myogenin. These observations show that the upstream IGF‐1/Akt1 regulation is dispensable for the acute activation of mTORC1 signaling and regulation of satellite cells in response to mechanical overload.
Collapse
Affiliation(s)
- Mitsunori Miyazaki
- Department of Physical Therapy, School of Rehabilitation Sciences, Health Sciences University of Hokkaido, Hokkaido, Japan
| | - Nobuki Moriya
- Department of Physical Therapy, School of Rehabilitation Sciences, Health Sciences University of Hokkaido, Hokkaido, Japan.,Department of Rehabilitation, Faculty of Medical Science and Welfare, Tohoku Bunka Gakuen University, Miyagi, Japan
| | - Tohru Takemasa
- Graduate School of Comprehensive Human Sciences, University of Tsukuba, Ibaraki, Japan
| |
Collapse
|
8
|
Höhfeld J, Benzing T, Bloch W, Fürst DO, Gehlert S, Hesse M, Hoffmann B, Hoppe T, Huesgen PF, Köhn M, Kolanus W, Merkel R, Niessen CM, Pokrzywa W, Rinschen MM, Wachten D, Warscheid B. Maintaining proteostasis under mechanical stress. EMBO Rep 2021; 22:e52507. [PMID: 34309183 PMCID: PMC8339670 DOI: 10.15252/embr.202152507] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Revised: 06/28/2021] [Accepted: 07/01/2021] [Indexed: 12/11/2022] Open
Abstract
Cell survival, tissue integrity and organismal health depend on the ability to maintain functional protein networks even under conditions that threaten protein integrity. Protection against such stress conditions involves the adaptation of folding and degradation machineries, which help to preserve the protein network by facilitating the refolding or disposal of damaged proteins. In multicellular organisms, cells are permanently exposed to stress resulting from mechanical forces. Yet, for long time mechanical stress was not recognized as a primary stressor that perturbs protein structure and threatens proteome integrity. The identification and characterization of protein folding and degradation systems, which handle force-unfolded proteins, marks a turning point in this regard. It has become apparent that mechanical stress protection operates during cell differentiation, adhesion and migration and is essential for maintaining tissues such as skeletal muscle, heart and kidney as well as the immune system. Here, we provide an overview of recent advances in our understanding of mechanical stress protection.
Collapse
Affiliation(s)
- Jörg Höhfeld
- Institute for Cell BiologyRheinische Friedrich‐Wilhelms University BonnBonnGermany
| | - Thomas Benzing
- Department II of Internal Medicine and Center for Molecular Medicine Cologne (CMMC)University of CologneCologneGermany
| | - Wilhelm Bloch
- Institute of Cardiovascular Research and Sports MedicineGerman Sport UniversityCologneGermany
| | - Dieter O Fürst
- Institute for Cell BiologyRheinische Friedrich‐Wilhelms University BonnBonnGermany
| | - Sebastian Gehlert
- Institute of Cardiovascular Research and Sports MedicineGerman Sport UniversityCologneGermany
- Department for the Biosciences of SportsInstitute of Sports ScienceUniversity of HildesheimHildesheimGermany
| | - Michael Hesse
- Institute of Physiology I, Life & Brain CenterMedical FacultyRheinische Friedrich‐Wilhelms UniversityBonnGermany
| | - Bernd Hoffmann
- Institute of Biological Information Processing, IBI‐2: MechanobiologyForschungszentrum JülichJülichGermany
| | - Thorsten Hoppe
- Institute for GeneticsCologne Excellence Cluster on Cellular Stress Responses in Aging‐Associated Diseases (CECAD) and CMMCUniversity of CologneCologneGermany
| | - Pitter F Huesgen
- Central Institute for Engineering, Electronics and Analytics, ZEA3Forschungszentrum JülichJülichGermany
- CECADUniversity of CologneCologneGermany
| | - Maja Köhn
- Institute of Biology IIIFaculty of Biology, and Signalling Research Centres BIOSS and CIBSSAlbert‐Ludwigs‐University FreiburgFreiburgGermany
| | - Waldemar Kolanus
- LIMES‐InstituteRheinische Friedrich‐Wilhelms University BonnBonnGermany
| | - Rudolf Merkel
- Institute of Biological Information Processing, IBI‐2: MechanobiologyForschungszentrum JülichJülichGermany
| | - Carien M Niessen
- Department of Dermatology and CECADUniversity of CologneCologneGermany
| | | | - Markus M Rinschen
- Department of Biomedicine and Aarhus Institute of Advanced StudiesAarhus UniversityAarhusDenmark
- Department of MedicineUniversity Medical Center Hamburg‐EppendorfHamburgGermany
| | - Dagmar Wachten
- Institute of Innate ImmunityUniversity Hospital BonnBonnGermany
| | - Bettina Warscheid
- Institute of Biology IIFaculty of Biology, and Signalling Research Centres BIOSS and CIBSSAlbert‐Ludwigs‐University FreiburgFreiburgGermany
| |
Collapse
|
9
|
Nissinen TA, Hentilä J, Fachada V, Lautaoja JH, Pasternack A, Ritvos O, Kivelä R, Hulmi JJ. Muscle follistatin gene delivery increases muscle protein synthesis independent of periodical physical inactivity and fasting. FASEB J 2021; 35:e21387. [PMID: 33559263 DOI: 10.1096/fj.202002008r] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Revised: 11/27/2020] [Accepted: 01/07/2021] [Indexed: 12/21/2022]
Abstract
Blocking of myostatin and activins effectively counteracts muscle atrophy. However, the potential interaction with physical inactivity and fasting in the regulation of muscle protein synthesis is poorly understood. We used blockade of myostatin and activins by recombinant adeno-associated virus (rAAV)-mediated follistatin (FS288) overexpression in mouse tibialis anterior muscle. To investigate the effects on muscle protein synthesis, muscles were collected 7 days after rAAV-injection in the nighttime or in the daytime representing high and low levels of activity and feeding, respectively, or after overnight fasting, refeeding, or ad libitum feeding. Muscle protein synthesis was increased by FS288 independent of the time of the day or the feeding status. However, the activation of mTORC1 signaling by FS288 was attenuated in the daytime and by overnight fasting. FS288 also increased the amount of mTOR colocalized with lysosomes, but did not alter their localization toward the sarcolemma. This study shows that FS288 gene delivery increases muscle protein synthesis largely independent of diurnal fluctuations in physical activity and food intake or feeding status, overriding the physiological signals. This is important for eg cachectic and sarcopenic patients with reduced physical activity and appetite. The FS288-induced increase in mTORC1 signaling and protein synthesis may be in part driven by increased amount of mTOR colocalized with lysosomes, but not by their localization toward sarcolemma.
Collapse
Affiliation(s)
- Tuuli A Nissinen
- Faculty of Sport and Health Sciences, NeuroMuscular Research Center, University of Jyväskylä, Jyväskylä, Finland
| | - Jaakko Hentilä
- Faculty of Sport and Health Sciences, NeuroMuscular Research Center, University of Jyväskylä, Jyväskylä, Finland
| | - Vasco Fachada
- Faculty of Sport and Health Sciences, NeuroMuscular Research Center, University of Jyväskylä, Jyväskylä, Finland
| | - Juulia H Lautaoja
- Faculty of Sport and Health Sciences, NeuroMuscular Research Center, University of Jyväskylä, Jyväskylä, Finland
| | - Arja Pasternack
- Department of Physiology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Olli Ritvos
- Department of Physiology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Riikka Kivelä
- Stem Cells and Metabolism Research Program, Research Programs Unit, Faculty of Medicine, University of Helsinki, Helsinki, Finland.,Wihuri Research Institute, Helsinki, Finland
| | - Juha J Hulmi
- Faculty of Sport and Health Sciences, NeuroMuscular Research Center, University of Jyväskylä, Jyväskylä, Finland
| |
Collapse
|
10
|
Solsona R, Pavlin L, Bernardi H, Sanchez AMJ. Molecular Regulation of Skeletal Muscle Growth and Organelle Biosynthesis: Practical Recommendations for Exercise Training. Int J Mol Sci 2021; 22:2741. [PMID: 33800501 PMCID: PMC7962973 DOI: 10.3390/ijms22052741] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 03/04/2021] [Accepted: 03/04/2021] [Indexed: 12/18/2022] Open
Abstract
The regulation of skeletal muscle mass and organelle homeostasis is dependent on the capacity of cells to produce proteins and to recycle cytosolic portions. In this investigation, the mechanisms involved in skeletal muscle mass regulation-especially those associated with proteosynthesis and with the production of new organelles-are presented. Thus, the critical roles of mammalian/mechanistic target of rapamycin complex 1 (mTORC1) pathway and its regulators are reviewed. In addition, the importance of ribosome biogenesis, satellite cells involvement, myonuclear accretion, and some major epigenetic modifications related to protein synthesis are discussed. Furthermore, several studies conducted on the topic of exercise training have recognized the central role of both endurance and resistance exercise to reorganize sarcomeric proteins and to improve the capacity of cells to build efficient organelles. The molecular mechanisms underlying these adaptations to exercise training are presented throughout this review and practical recommendations for exercise prescription are provided. A better understanding of the aforementioned cellular pathways is essential for both healthy and sick people to avoid inefficient prescriptions and to improve muscle function with emergent strategies (e.g., hypoxic training). Finally, current limitations in the literature and further perspectives, notably on epigenetic mechanisms, are provided to encourage additional investigations on this topic.
Collapse
Affiliation(s)
- Robert Solsona
- Laboratoire Interdisciplinaire Performance Santé Environnement de Montagne (LIPSEM), Faculty of Sports Sciences, University of Perpignan Via Domitia, UR 4640, 7 Avenue Pierre de Coubertin, 66120 Font-Romeu, France;
| | - Laura Pavlin
- DMEM, University of Montpellier, INRAE UMR866, 2 Place Pierre Viala, 34060 Montpellier, France; (L.P.); (H.B.)
| | - Henri Bernardi
- DMEM, University of Montpellier, INRAE UMR866, 2 Place Pierre Viala, 34060 Montpellier, France; (L.P.); (H.B.)
| | - Anthony MJ Sanchez
- Laboratoire Interdisciplinaire Performance Santé Environnement de Montagne (LIPSEM), Faculty of Sports Sciences, University of Perpignan Via Domitia, UR 4640, 7 Avenue Pierre de Coubertin, 66120 Font-Romeu, France;
| |
Collapse
|
11
|
Liegnell R, Apró W, Danielsson S, Ekblom B, van Hall G, Holmberg HC, Moberg M. Elevated plasma lactate levels via exogenous lactate infusion do not alter resistance exercise-induced signaling or protein synthesis in human skeletal muscle. Am J Physiol Endocrinol Metab 2020; 319:E792-E804. [PMID: 32830552 DOI: 10.1152/ajpendo.00291.2020] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Lactate has been implicated as a potential signaling molecule. In myotubes, lactate incubation increases mechanistic target of rapamycin complex 1 (mTORC1)- and ERK-signaling and induces hypertrophy, indicating that lactate could be a mediator of muscle adaptations to resistance exercise. However, the potential signaling properties of lactate, at rest or with exercise, have not been explored in human tissue. In a crossover design study, 8 men and 8 women performed one-legged resistance exercise while receiving venous infusion of saline or sodium lactate. Blood was sampled repeatedly, and muscle biopsies were collected at rest and at 0, 90, and 180 min and 24 h after exercise. The primary outcomes examined were intracellular signaling, fractional protein synthesis rate (FSR), and blood/muscle levels of lactate and pH. Postexercise blood lactate concentrations were 130% higher in the Lactate trial (3.0 vs. 7.0 mmol/L, P < 0.001), whereas muscle levels were only marginally higher (27 vs. 32 mmol/kg dry wt, P = 0.003) compared with the Saline trial. Postexercise blood pH was higher in the Lactate trial (7.34 vs. 7.44, P < 0.001), with no differences in intramuscular pH. Exercise increased the phosphorylation of mTORS2448 (∼40%), S6K1T389 (∼3-fold), and p44T202/T204 (∼80%) during recovery, without any differences between trials. FSR over the 24-h recovery period did not differ between the Saline (0.067%/h) and Lactate (0.062%/h) trials. This study does not support the hypothesis that blood lactate levels can modulate anabolic signaling in contracted human muscle. Further in vivo research investigating the impact of exercised versus rested muscle and the role of intramuscular lactate is needed to elucidate its potential signaling properties.
Collapse
Affiliation(s)
- Rasmus Liegnell
- Department of Physiology, Nutrition and Biomechaniscs, Swedish School of Sport and Health Sciences, Stockholm, Sweden
| | - William Apró
- Department of Physiology, Nutrition and Biomechaniscs, Swedish School of Sport and Health Sciences, Stockholm, Sweden
| | - Sebastian Danielsson
- Department of Physiology, Nutrition and Biomechaniscs, Swedish School of Sport and Health Sciences, Stockholm, Sweden
| | - Björn Ekblom
- Department of Physiology, Nutrition and Biomechaniscs, Swedish School of Sport and Health Sciences, Stockholm, Sweden
| | - Gerrit van Hall
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
- Clinical Metabolomics Core Facility, Clinical Biochemistry, Rigshospitalet, Copenhagen, Denmark
| | - Hans-Christer Holmberg
- Swedish Winter Sports Research Centre, Department of Health Sciences, Mid Sweden University, Östersund, Sweden
- Department of Physiology and Pharmacology, Biomedicum C5, Karolinska Institute, Stockholm, Sweden
| | - Marcus Moberg
- Department of Physiology, Nutrition and Biomechaniscs, Swedish School of Sport and Health Sciences, Stockholm, Sweden
| |
Collapse
|
12
|
Rindom E, Kristensen AM, Overgaard K, Vissing K, Paoli FV. Activation of mTORC1 signalling in rat skeletal muscle is independent of the EC-coupling sequence but dependent on tension per se in a dose-response relationship. Acta Physiol (Oxf) 2019; 227:e13336. [PMID: 31231946 DOI: 10.1111/apha.13336] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2019] [Revised: 06/04/2019] [Accepted: 06/20/2019] [Indexed: 12/29/2022]
Abstract
AIM mTORC1 is regarded as an important key regulator of protein synthesis and hypertrophy following mechanical stimuli in skeletal muscle. However, as excitation and tension development is tightly coupled in most experimental models, very little and largely indirect evidence exist for such a mechanosensitive pathway. Here, we sought to examine whether activation of mTORC1 signalling is dependent on tension per se in rat skeletal muscle. METHODS To examine the mechanosensitivity of mTORC1, rat EDL muscles were exposed to either excitation-induced eccentric contractions (ECC), passive stretching (PAS) with identical peak tension (Tpeak ) and Tension-Time-Integral (TTI), or ECC with addition of inhibitors of the myosin ATPases (IMA ). To further explore the relationship between tension and mTORC1 signalling, rat EDL muscles were subjected to PAS of different magnitudes of Tpeak while standardizing TTI and vice versa. RESULTS PAS and ECC with equal Tpeak and TTI produced similar responses in mTORC1 signalling despite different modes of tension development. When active tension during ECC was nearly abolished by addition of IMA , mTORC1 signalling was reduced to a level comparable to non-stimulated controls. In addition, when muscles were exposed to PAS of varying levels of Tpeak with standardized TTI, activation of mTORC1 signalling displayed a positive relationship with peak tension. CONCLUSIONS The current study directly links tension per se to activation of mTORC1 signalling, which is independent of an active EC-coupling sequence. Moreover, activation of mTORC1 signalling displays a positive dose-response relationship with peak tension.
Collapse
Affiliation(s)
- Emil Rindom
- Department of Biomedicine Aarhus University Aarhus Denmark
| | - Anders M. Kristensen
- Section for Sport Science, Department of Public Health Aarhus University Aarhus Denmark
| | - Kristian Overgaard
- Section for Sport Science, Department of Public Health Aarhus University Aarhus Denmark
| | - Kristian Vissing
- Section for Sport Science, Department of Public Health Aarhus University Aarhus Denmark
| | | |
Collapse
|
13
|
Filipovic A, DeMarees M, Grau M, Hollinger A, Seeger B, Schiffer T, Bloch W, Gehlert S. Superimposed Whole-Body Electrostimulation Augments Strength Adaptations and Type II Myofiber Growth in Soccer Players During a Competitive Season. Front Physiol 2019; 10:1187. [PMID: 31607944 PMCID: PMC6768094 DOI: 10.3389/fphys.2019.01187] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2019] [Accepted: 09/02/2019] [Indexed: 12/25/2022] Open
Abstract
Background The improvement of strength and athletic performance during a competitive season in elite soccer players is a demanding task for the coach. Aims As whole-body electrostimulation (WB-EMS) training provides a time efficient stimulation potentially capable in exerting skeletal muscle adaptations we aimed to test this approach over 7 weeks in trained male soccer players during a competitive season. Hypothesis We hypothesized that a superimposed WB-EMS will increase maximal strength and type I and type II myofiber hypertrophy. Methods Twenty-eight male field soccer players were assigned in either a WB-EMS group (EG, n = 10), a training group (TG, n = 10), or a control group (CG, n = 8). The regular soccer training consists of two to four sessions and one match per week. In concurrent, the EG performed 3 × 10 squat jumps superimposed with WB-EMS twice per week, TG performed 3 × 10 squat jumps without EMS twice per week, and the CG only performed the regular soccer training. Muscle biopsies were collected and strength tests were performed under resting conditions before (Baseline) and after the intervention period (Posttest). Muscle biopsies were analyzed via western blotting and immunohistochemistry for skeletal muscle adaptive responses. To determine the effect of the training interventions a 2 × 3 (time ∗ group) mixed ANOVA with repeated measures was conducted. Results Maximal strength in leg press (p = 0.009) and leg curl (p = 0.026) was significantly increased in EG along with a small but significant increase in type II myofiber diameter (p = 0.023). All of these adaptations were not observed in TG and CG. Conclusion WB-EMS can serve as a time efficient training method to augment strength capacities and type II fiber myofiber growth in soccer players when combined with specific resistance training. This combination may therefore be a promising training modification compared to traditional strength training for performance enhancement.
Collapse
Affiliation(s)
- Andre Filipovic
- Section of Molecular and Cellular Sport Medicine, Institute of Cardiology and Sports Medicine, German Sport University Cologne, Cologne, Germany
| | - Markus DeMarees
- Section of Sports Medicine and Sports Nutrition, Faculty of Sports Science, Ruhr-University Bochum, Bochum, Germany
| | - Marijke Grau
- Section of Molecular and Cellular Sport Medicine, Institute of Cardiology and Sports Medicine, German Sport University Cologne, Cologne, Germany
| | - Anna Hollinger
- Section of Molecular and Cellular Sport Medicine, Institute of Cardiology and Sports Medicine, German Sport University Cologne, Cologne, Germany
| | - Benedikt Seeger
- Section of Molecular and Cellular Sport Medicine, Institute of Cardiology and Sports Medicine, German Sport University Cologne, Cologne, Germany
| | - Thorsten Schiffer
- Outpatient Clinic for Sports Traumatology and Public Health Consultation, German Sport University Cologne, Cologne, Germany
| | - Wilhelm Bloch
- Section of Molecular and Cellular Sport Medicine, Institute of Cardiology and Sports Medicine, German Sport University Cologne, Cologne, Germany
| | - Sebastian Gehlert
- Section of Molecular and Cellular Sport Medicine, Institute of Cardiology and Sports Medicine, German Sport University Cologne, Cologne, Germany.,Institute of Sport Science, Biosciences of Sports, University of Hildesheim, Hildesheim, Germany
| |
Collapse
|
14
|
Kirby TJ. Mechanosensitive pathways controlling translation regulatory processes in skeletal muscle and implications for adaptation. J Appl Physiol (1985) 2019; 127:608-618. [PMID: 31295035 DOI: 10.1152/japplphysiol.01031.2018] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The ability of myofibers to sense and respond appropriately to mechanical signals is one of the primary determinants of the skeletal muscle phenotype. In response to a change in mechanical load, muscle cells alter their protein metabolism, primarily through the regulation of protein synthesis rate. Protein synthesis rates are determined by both translation efficiency and translational capacity within the muscle. Translational capacity is strongly determined by the ribosome content of the muscle; thus the regulation of ribosomal biogenesis by mechanical inputs has been an area of recent interest. Despite the clear association between mechanical signals and changes in protein metabolism, the molecular pathways that link these events are still not fully elucidated. This review focuses on recent studies looking at how mechanosignaling impacts translational events. The role of impaired mechanotransduction in aging is discussed, as is the connection between age-dependent signaling defects and compromised ribosomal biogenesis during mechanical overload. Finally, emerging evidence suggests that the nucleus can act as a mechanosensitive element and that this mode of mechanotransduction may have an important role in skeletal muscle physiology and adaptation.
Collapse
Affiliation(s)
- Tyler J Kirby
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York.,Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, New York
| |
Collapse
|
15
|
Shirvani H, Rahmati-Ahmadabad S, Broom DR, Mirnejad R. Eccentric resistance training and β-hydroxy-β-methylbutyrate free acid affects muscle PGC-1α expression and serum irisin, nesfatin-1 and resistin in rats. ACTA ACUST UNITED AC 2019; 222:jeb.198424. [PMID: 31085594 DOI: 10.1242/jeb.198424] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Accepted: 05/05/2019] [Indexed: 01/13/2023]
Abstract
The hypothalamus controls metabolism and feeding behaviour via several signals with other tissues. Exercise and supplements can change hypothalamic signalling pathways, so the present study investigated the influence of eccentric resistance training and β-hydroxy-β-methylbutyrate free acid supplementation on PGC-1α expression, serum irisin, nesfatin-1 and resistin concentrations. Thirty-two male rats (8 weeks old, 200±17 g body mass) were randomly allocated to control, β-hydroxy-β-methylbutyrate free acid supplementation (HMB), eccentric resistance training (ERT), and β-hydroxy-β-methylbutyrate free acid supplementation plus eccentric resistance training (HMB+ERT) groups. Training groups undertook eccentric resistance training (6 weeks, 3 times a week) and supplement groups consumed β-hydroxy-β-methylbutyrate free acid (HMB-FA) orally (76 mg kg-1 day-1). Twenty-four hours after the last training session, serum and triceps brachii muscle samples were collected and sent to the laboratory for analysis. Two-way ANOVA and Pearson correlation were employed (significance level: P<0.05). The results showed that eccentric resistance training increases skeletal muscle PGC-1α gene expression, as well as serum levels of irisin and nesfatin-1 (P=0.001). Eccentric resistance training decreased the serum concentration of resistin (P=0.001). HMB-FA supplementation increased skeletal muscle PGC-1α gene expression (P=0.002), as well as the serum concentration of irisin and nesfatin-1 (P=0.001), but decreased the serum concentration of resistin (P=0.001). Significant correlations were observed between PGC-1α gene expression and serum concentrations of irisin, nesfatin-1 and resistin. HMB-FA supplementation with eccentric resistance training may induce crosstalk between peptide release from other tissues and increases maximal muscle strength. The combination of the two interventions had a more substantial effect than each in isolation.
Collapse
Affiliation(s)
- Hossein Shirvani
- Exercise Physiology Research Center, Life Style Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | | | - David Robert Broom
- Academy of Sport and Physical Activity, Faculty of Health and Wellbeing, Sheffield Hallam University, Sheffield S10 2BP, UK
| | - Reza Mirnejad
- Molecular Biology Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| |
Collapse
|
16
|
Abstract
Some amino acids (AA) act through several signalling pathways and mechanisms to mediate the control of gene expression at the translation level, and the regulation occurs, specifically, on the initiation and the signalling pathways for translation. The translation of mRNA to protein synthesis proceeds through the steps of initiation and elongation, and AA act as important feed-forward activators that are involved in many pathways, such as the sensing and the transportation of AA by cells, in these steps in many tissues of mammals. For the translation, phosphorylation of eukaryotic translation initiation factor 2α (eIF2α) is a critical molecule that controls the translation initiation and its functions can be regulated by some AA. Another control point in the mRNA binding step in the translation initiation is at the regulation by mammalian target of rapamycin, which requires a change of phosphorylation status of ribosomal protein S6. In fact, the change of phosphorylation status of ribosomal protein S6 might be involved in global protein synthesis. The present review summarises recent work on the molecular mechanisms of the regulation of protein synthesis by AA and highlights new findings.
Collapse
|
17
|
You JS, McNally RM, Jacobs BL, Privett RE, Gundermann DM, Lin KH, Steinert ND, Goodman CA, Hornberger TA. The role of raptor in the mechanical load-induced regulation of mTOR signaling, protein synthesis, and skeletal muscle hypertrophy. FASEB J 2019; 33:4021-4034. [PMID: 30509128 PMCID: PMC6404572 DOI: 10.1096/fj.201801653rr] [Citation(s) in RCA: 104] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Accepted: 11/05/2018] [Indexed: 12/21/2022]
Abstract
It is well known that an increase in mechanical loading can induce skeletal muscle hypertrophy, and a long standing model in the field indicates that mechanical loads induce hypertrophy via a mechanism that requires signaling through the mechanistic target of rapamycin complex 1 (mTORC1). Specifically, it has been widely proposed that mechanical loads activate signaling through mTORC1 and that this, in turn, promotes an increase in the rate of protein synthesis and the subsequent hypertrophic response. However, this model is based on a number of important assumptions that have not been rigorously tested. In this study, we created skeletal muscle specific and inducible raptor knockout mice to eliminate signaling by mTORC1, and with these mice we were able to directly demonstrate that mechanical stimuli can activate signaling by mTORC1, and that mTORC1 is necessary for mechanical load-induced hypertrophy. Surprisingly, however, we also obtained multiple lines of evidence that indicate that mTORC1 is not required for a mechanical load-induced increase in the rate of protein synthesis. This observation highlights an important shortcoming in our understanding of how mechanical loads induce hypertrophy and illustrates that additional mTORC1-independent mechanisms play a critical role in this process.-You, J.-S., McNally, R. M., Jacobs, B. L., Privett, R. E., Gundermann, D. M., Lin, K.-H., Steinert, N. D., Goodman, C. A., Hornberger, T. A. The role of raptor in the mechanical load-induced regulation of mTOR signaling, protein synthesis, and skeletal muscle hypertrophy.
Collapse
Affiliation(s)
- Jae-Sung You
- Department of Comparative Biosciences, University of Wisconsin–Madison, Madison, Wisconsin, USA
- School of Veterinary Medicine, University of Wisconsin–Madison, Madison, Wisconsin, USA
| | - Rachel M. McNally
- Department of Comparative Biosciences, University of Wisconsin–Madison, Madison, Wisconsin, USA
- School of Veterinary Medicine, University of Wisconsin–Madison, Madison, Wisconsin, USA
| | - Brittany L. Jacobs
- Department of Comparative Biosciences, University of Wisconsin–Madison, Madison, Wisconsin, USA
- School of Veterinary Medicine, University of Wisconsin–Madison, Madison, Wisconsin, USA
| | - Rachel E. Privett
- Department of Comparative Biosciences, University of Wisconsin–Madison, Madison, Wisconsin, USA
- School of Veterinary Medicine, University of Wisconsin–Madison, Madison, Wisconsin, USA
| | - David M. Gundermann
- Department of Comparative Biosciences, University of Wisconsin–Madison, Madison, Wisconsin, USA
- School of Veterinary Medicine, University of Wisconsin–Madison, Madison, Wisconsin, USA
| | - Kuan-Hung Lin
- Department of Comparative Biosciences, University of Wisconsin–Madison, Madison, Wisconsin, USA
- School of Veterinary Medicine, University of Wisconsin–Madison, Madison, Wisconsin, USA
| | - Nate D. Steinert
- Department of Comparative Biosciences, University of Wisconsin–Madison, Madison, Wisconsin, USA
- School of Veterinary Medicine, University of Wisconsin–Madison, Madison, Wisconsin, USA
| | - Craig A. Goodman
- Department of Comparative Biosciences, University of Wisconsin–Madison, Madison, Wisconsin, USA
- School of Veterinary Medicine, University of Wisconsin–Madison, Madison, Wisconsin, USA
- Institute of Health and Sport, Victoria University, Melbourne, Victoria, Australia
- Australian Institute for Musculoskeletal Science, Victoria University, St. Albans, Victoria, Australia
| | - Troy A. Hornberger
- Department of Comparative Biosciences, University of Wisconsin–Madison, Madison, Wisconsin, USA
- School of Veterinary Medicine, University of Wisconsin–Madison, Madison, Wisconsin, USA
| |
Collapse
|
18
|
Goodman CA. Role of mTORC1 in mechanically induced increases in translation and skeletal muscle mass. J Appl Physiol (1985) 2019; 127:581-590. [PMID: 30676865 DOI: 10.1152/japplphysiol.01011.2018] [Citation(s) in RCA: 82] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Skeletal muscle mass is, in part, regulated by the rate of mRNA translation (i.e., protein synthesis). The conserved serine/threonine kinase, mTOR (the mammalian/mechanistic target of rapamycin), found in the multiprotein complex, mTOR complex 1 (mTORC1), is a major positive regulator of protein synthesis. The purpose of this review is to describe some of the critical steps in translation initiation, mTORC1 and its potential direct and indirect roles in regulating translation, and evidence that mTORC1 regulates protein synthesis and muscle mass, with a particular focus on basal conditions and the response to mechanical stimuli. Current evidence suggests that for acute contraction models of mechanical stimuli, there is an emerging pattern suggesting that there is an early increase in protein synthesis governed by a rapamycin-sensitive mTORC1-dependent mechanism, while at later poststimulation time points, the mechanism may change to a rapamycin-insensitive mTORC1-dependent or even an mTORC1-independent mechanism. Furthermore, evidence suggests that mTORC1 appears to be absolutely necessary for muscle fiber hypertrophy induced by chronic mechanical loading but may only play a partial role in the hypertrophy induced by more intermittent types of acute resistance exercise, with the possibility of mTORC1-independent mechanisms also playing a role. Despite the progress that has been made, many questions about the activation of mTORC1, and its downstream targets, remain to be answered. Further research will hopefully provide novel insights into the regulation of skeletal muscle mTORC1 that may eventually be translated into novel exercise programing and/or targeted pharmacological therapies aimed at preventing muscle wasting and/or increasing muscle mass.
Collapse
Affiliation(s)
- Craig A Goodman
- Institute of Health and Sport; Victoria University, Melbourne, Australia.,Australian Institute for Musculoskeletal Science (AIMSS), Victoria University, St. Albans, Victoria, Australia
| |
Collapse
|
19
|
Hodson N, Philp A. The Importance of mTOR Trafficking for Human Skeletal Muscle Translational Control. Exerc Sport Sci Rev 2019; 47:46-53. [PMID: 30334852 PMCID: PMC6310455 DOI: 10.1249/jes.0000000000000173] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
This review will critique cell, rodent, and human models of mTOR regulation to discuss why mTOR trafficking may represent a novel and physiologically relevant model of regulation in skeletal muscle. The mechanistic target of rapamycin (mTOR) is a central regulator of muscle protein synthesis, and its activation has long been attributed to its translocation to the lysosome. Here, we present a novel model of mTOR activation in skeletal muscle where the translocation of mTOR and the lysosome toward the cell membrane is a key process in mTOR activation.
Collapse
Affiliation(s)
- Nathan Hodson
- School of Sport, Exercise and Rehabilitation Sciences, University of Birmingham, Birmingham, UK
| | - Andrew Philp
- School of Sport, Exercise and Rehabilitation Sciences, University of Birmingham, Birmingham, UK.,Garvan Institute of Medical Research, Darlinghurst, Sydney, New South Wales, Australia
| |
Collapse
|
20
|
Exercise and the control of muscle mass in human. Pflugers Arch 2018; 471:397-411. [DOI: 10.1007/s00424-018-2217-x] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Revised: 10/02/2018] [Accepted: 10/04/2018] [Indexed: 12/19/2022]
|
21
|
Bamman MM, Roberts BM, Adams GR. Molecular Regulation of Exercise-Induced Muscle Fiber Hypertrophy. Cold Spring Harb Perspect Med 2018; 8:cshperspect.a029751. [PMID: 28490543 DOI: 10.1101/cshperspect.a029751] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Skeletal muscle hypertrophy is a widely sought exercise adaptation to counteract the muscle atrophy of aging and disease, or to improve athletic performance. While this desired muscle enlargement is a well-known adaptation to resistance exercise training (RT), the mechanistic underpinnings are not fully understood. The purpose of this review is thus to provide the reader with a summary of recent advances in molecular mechanisms-based on the most current literature-that are thought to promote RT-induced muscle hypertrophy. We have therefore focused this discussion on the following areas of fertile investigation: ribosomal function and biogenesis, muscle stem (satellite) cell activity, transcriptional regulation, mechanotransduction, and myokine signaling.
Collapse
Affiliation(s)
- Marcas M Bamman
- Department of Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, Birmingham, Alabama 35294.,UAB Center for Exercise Medicine, University of Alabama at Birmingham, Birmingham, Alabama 35205.,Geriatric Research, Education, and Clinical Center, Veterans' Affairs Medical Center, Birmingham, Alabama 35233
| | - Brandon M Roberts
- Department of Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, Birmingham, Alabama 35294.,UAB Center for Exercise Medicine, University of Alabama at Birmingham, Birmingham, Alabama 35205
| | - Gregory R Adams
- Department of Physiology and Biophysics, University of California Irvine, Irvine, California 92617
| |
Collapse
|
22
|
Nissinen TA, Hentilä J, Penna F, Lampinen A, Lautaoja JH, Fachada V, Holopainen T, Ritvos O, Kivelä R, Hulmi JJ. Treating cachexia using soluble ACVR2B improves survival, alters mTOR localization, and attenuates liver and spleen responses. J Cachexia Sarcopenia Muscle 2018; 9:514-529. [PMID: 29722201 PMCID: PMC5989872 DOI: 10.1002/jcsm.12310] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Revised: 03/20/2018] [Accepted: 03/27/2018] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Cancer cachexia increases morbidity and mortality, and blocking of activin receptor ligands has improved survival in experimental cancer. However, the underlying mechanisms have not yet been fully uncovered. METHODS The effects of blocking activin receptor type 2 (ACVR2) ligands on both muscle and non-muscle tissues were investigated in a preclinical model of cancer cachexia using a recombinant soluble ACVR2B (sACVR2B-Fc). Treatment with sACVR2B-Fc was applied either only before the tumour formation or with continued treatment both before and after tumour formation. The potential roles of muscle and non-muscle tissues in cancer cachexia were investigated in order to understand the possible mechanisms of improved survival mediated by ACVR2 ligand blocking. RESULTS Blocking of ACVR2 ligands improved survival in tumour-bearing mice only when the mice were treated both before and after the tumour formation. This occurred without effects on tumour growth, production of pro-inflammatory cytokines or the level of physical activity. ACVR2 ligand blocking was associated with increased muscle (limb and diaphragm) mass and attenuation of both hepatic protein synthesis and splenomegaly. Especially, the effects on the liver and the spleen were observed independent of the treatment protocol. The prevention of splenomegaly by sACVR2B-Fc was not explained by decreased markers of myeloid-derived suppressor cells. Decreased tibialis anterior, diaphragm, and heart protein synthesis were observed in cachectic mice. This was associated with decreased mechanistic target of rapamycin (mTOR) colocalization with late-endosomes/lysosomes, which correlated with cachexia and reduced muscle protein synthesis. CONCLUSIONS The prolonged survival with continued ACVR2 ligand blocking could potentially be attributed in part to the maintenance of limb and respiratory muscle mass, but many observed non-muscle effects suggest that the effect may be more complex than previously thought. Our novel finding showing decreased mTOR localization in skeletal muscle with lysosomes/late-endosomes in cancer opens up new research questions and possible treatment options for cachexia.
Collapse
Affiliation(s)
- Tuuli A Nissinen
- Neuromuscular Research Center, Biology of Physical Activity, Faculty of Sport and Health Sciences, University of Jyväskylä, Rautpohjankatu 8, Jyväskylä, 40014, Finland
| | - Jaakko Hentilä
- Neuromuscular Research Center, Biology of Physical Activity, Faculty of Sport and Health Sciences, University of Jyväskylä, Rautpohjankatu 8, Jyväskylä, 40014, Finland
| | - Fabio Penna
- Department of Clinical and Biological Sciences, University of Turin, Corso Raffaello, Turin, 10125, Italy
| | - Anita Lampinen
- Neuromuscular Research Center, Biology of Physical Activity, Faculty of Sport and Health Sciences, University of Jyväskylä, Rautpohjankatu 8, Jyväskylä, 40014, Finland
| | - Juulia H Lautaoja
- Neuromuscular Research Center, Biology of Physical Activity, Faculty of Sport and Health Sciences, University of Jyväskylä, Rautpohjankatu 8, Jyväskylä, 40014, Finland
| | - Vasco Fachada
- Neuromuscular Research Center, Biology of Physical Activity, Faculty of Sport and Health Sciences, University of Jyväskylä, Rautpohjankatu 8, Jyväskylä, 40014, Finland
| | - Tanja Holopainen
- Translational Cancer Biology Program, Research Programs Unit, Faculty of Medicine, University of Helsinki, and Wihuri Research Institute, Haartmaninkatu 8, Helsinki, 00290, Finland
| | - Olli Ritvos
- Department of Physiology, Faculty of Medicine, University of Helsinki, Haartmaninkatu 8, Helsinki, 00290, Finland
| | - Riikka Kivelä
- Translational Cancer Biology Program, Research Programs Unit, Faculty of Medicine, University of Helsinki, and Wihuri Research Institute, Haartmaninkatu 8, Helsinki, 00290, Finland
| | - Juha J Hulmi
- Neuromuscular Research Center, Biology of Physical Activity, Faculty of Sport and Health Sciences, University of Jyväskylä, Rautpohjankatu 8, Jyväskylä, 40014, Finland.,Department of Physiology, Faculty of Medicine, University of Helsinki, Haartmaninkatu 8, Helsinki, 00290, Finland
| |
Collapse
|
23
|
Kim J, Bilder D, Neufeld TP. Mechanical stress regulates insulin sensitivity through integrin-dependent control of insulin receptor localization. Genes Dev 2018; 32:156-164. [PMID: 29440263 PMCID: PMC5830928 DOI: 10.1101/gad.305870.117] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Accepted: 01/02/2018] [Indexed: 12/22/2022]
Abstract
Kim et al. show that insulin signaling in Drosophila adipocytes is abolished in the absence of physical activity and mechanical stress. The insulin receptor and downstream components are recruited to the plasma membrane upon stress sensing mediated by integrins. Insulin resistance, the failure to activate insulin signaling in the presence of ligand, leads to metabolic diseases, including type 2 diabetes. Physical activity and mechanical stress have been shown to protect against insulin resistance, but the molecular mechanisms remain unclear. Here, we address this relationship in the Drosophila larval fat body, an insulin-sensitive organ analogous to vertebrate adipose tissue and livers. We found that insulin signaling in Drosophila fat body cells is abolished in the absence of physical activity and mechanical stress even when excess insulin is present. Physical movement is required for insulin sensitivity in both intact larvae and fat bodies cultured ex vivo. Interestingly, the insulin receptor and other downstream components are recruited to the plasma membrane in response to mechanical stress, and this membrane localization is rapidly lost upon disruption of larval or tissue movement. Sensing of mechanical stimuli is mediated in part by integrins, whose activation is necessary and sufficient for mechanical stress-dependent insulin signaling. Insulin resistance develops naturally during the transition from the active larval stage to the immotile pupal stage, suggesting that regulation of insulin sensitivity by mechanical stress may help coordinate developmental programming with metabolism.
Collapse
Affiliation(s)
- Jung Kim
- Department of Genetics, Cell Biology, and Development, University of Minnesota, Minneapolis, Minnesota 55455, USA.,Department of Molecular and Cell Biology, University of California at Berkeley, Berkeley, California 94720, USA
| | - David Bilder
- Department of Molecular and Cell Biology, University of California at Berkeley, Berkeley, California 94720, USA
| | - Thomas P Neufeld
- Department of Genetics, Cell Biology, and Development, University of Minnesota, Minneapolis, Minnesota 55455, USA
| |
Collapse
|
24
|
Resistance exercise initiates mechanistic target of rapamycin (mTOR) translocation and protein complex co-localisation in human skeletal muscle. Sci Rep 2017; 7:5028. [PMID: 28694500 PMCID: PMC5504043 DOI: 10.1038/s41598-017-05483-x] [Citation(s) in RCA: 78] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2015] [Accepted: 06/19/2017] [Indexed: 12/18/2022] Open
Abstract
The mechanistic target of rapamycin (mTOR) is a central mediator of protein synthesis in skeletal muscle. We utilized immunofluorescence approaches to study mTOR cellular distribution and protein-protein co-localisation in human skeletal muscle in the basal state as well as immediately, 1 and 3 h after an acute bout of resistance exercise in a fed (FED; 20 g Protein/40 g carbohydrate/1 g fat) or energy-free control (CON) state. mTOR and the lysosomal protein LAMP2 were highly co-localised in basal samples. Resistance exercise resulted in rapid translocation of mTOR/LAMP2 towards the cell membrane. Concurrently, resistance exercise led to the dissociation of TSC2 from Rheb and increased in the co-localisation of mTOR and Rheb post exercise in both FED and CON. In addition, mTOR co-localised with Eukaryotic translation initiation factor 3 subunit F (eIF3F) at the cell membrane post-exercise in both groups, with the response significantly greater at 1 h of recovery in the FED compared to CON. Collectively our data demonstrate that cellular trafficking of mTOR occurs in human muscle in response to an anabolic stimulus, events that appear to be primarily influenced by muscle contraction. The translocation and association of mTOR with positive regulators (i.e. Rheb and eIF3F) is consistent with an enhanced mRNA translational capacity after resistance exercise.
Collapse
|
25
|
Zhikrivetskaya SO, Snezhkina AV, Zaretsky AR, Alekseev BY, Pokrovsky AV, Golovyuk AL, Melnikova NV, Stepanov OA, Kalinin DV, Moskalev AA, Krasnov GS, Dmitriev AA, Kudryavtseva AV. Molecular markers of paragangliomas/pheochromocytomas. Oncotarget 2017; 8:25756-25782. [PMID: 28187001 PMCID: PMC5421967 DOI: 10.18632/oncotarget.15201] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Accepted: 01/23/2017] [Indexed: 12/14/2022] Open
Abstract
Paragangliomas/pheochromocytomas comprise rare tumors that arise from the extra-adrenal paraganglia, with an incidence of about 2 to 8 per million people each year. Approximately 40% of cases are due to genetic mutations in at least one out of more than 30 causative genes. About 25-30% of pheochromocytomas/paragangliomas develop under the conditions of a hereditary tumor syndrome a third of which are caused by mutations in the VHL gene. Together, the gene mutations in this disorder have implicated multiple processes including signaling pathways, translation initiation, hypoxia regulation, protein synthesis, differentiation, survival, proliferation, and cell growth. The present review contemplates the mutations associated with the development of pheochromocytomas/paragangliomas and their potential to serve as specific markers of these tumors and their progression. These data will improve our understanding of the pathogenesis of these tumors and likely reveal certain features that may be useful for early diagnostics, malignancy prognostics, and the determination of new targets for disease therapeutics.
Collapse
Affiliation(s)
| | | | - Andrew R Zaretsky
- M.M. Shemyakin - Yu.A. Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
| | - Boris Y Alekseev
- National Medical Research Radiological Center, Ministry of Health of the Russian Federation, Moscow, Russia
| | | | | | - Nataliya V Melnikova
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
| | - Oleg A Stepanov
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
- National Medical Research Radiological Center, Ministry of Health of the Russian Federation, Moscow, Russia
| | | | - Alexey A Moskalev
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
| | - George S Krasnov
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
| | - Alexey A Dmitriev
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
| | - Anna V Kudryavtseva
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
- National Medical Research Radiological Center, Ministry of Health of the Russian Federation, Moscow, Russia
| |
Collapse
|
26
|
Jacobs BL, McNally RM, Kim KJ, Blanco R, Privett RE, You JS, Hornberger TA. Identification of mechanically regulated phosphorylation sites on tuberin (TSC2) that control mechanistic target of rapamycin (mTOR) signaling. J Biol Chem 2017; 292:6987-6997. [PMID: 28289099 PMCID: PMC5409467 DOI: 10.1074/jbc.m117.777805] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2017] [Revised: 03/08/2017] [Indexed: 12/31/2022] Open
Abstract
Mechanistic target of rapamycin (mTOR) signaling is necessary to generate a mechanically induced increase in skeletal muscle mass, but the mechanism(s) through which mechanical stimuli regulate mTOR signaling remain poorly defined. Recent studies have suggested that Ras homologue enriched in brain (Rheb), a direct activator of mTOR, and its inhibitor, the GTPase-activating protein tuberin (TSC2), may play a role in this pathway. To address this possibility, we generated inducible and skeletal muscle-specific knock-out mice for Rheb (iRhebKO) and TSC2 (iTSC2KO) and mechanically stimulated muscles from these mice with eccentric contractions (EC). As expected, the knock-out of TSC2 led to an elevation in the basal level of mTOR signaling. Moreover, we found that the magnitude of the EC-induced activation of mTOR signaling was significantly blunted in muscles from both inducible and skeletal muscle-specific knock-out mice for Rheb and iTSC2KO mice. Using mass spectrometry, we identified six sites on TSC2 whose phosphorylation was significantly altered by the EC treatment. Employing a transient transfection-based approach to rescue TSC2 function in muscles of the iTSC2KO mice, we demonstrated that these phosphorylation sites are required for the role that TSC2 plays in the EC-induced activation of mTOR signaling. Importantly, however, these phosphorylation sites were not required for an insulin-induced activation of mTOR signaling. As such, our results not only establish a critical role for Rheb and TSC2 in the mechanical activation of mTOR signaling, but they also expose the existence of a previously unknown branch of signaling events that can regulate the TSC2/mTOR pathway.
Collapse
Affiliation(s)
- Brittany L Jacobs
- From the Department of Comparative Biosciences and.,the School of Veterinary Medicine, University of Wisconsin-Madison, Madison, Wisconsin, 53706
| | - Rachel M McNally
- From the Department of Comparative Biosciences and.,the School of Veterinary Medicine, University of Wisconsin-Madison, Madison, Wisconsin, 53706
| | - Kook-Joo Kim
- From the Department of Comparative Biosciences and.,the School of Veterinary Medicine, University of Wisconsin-Madison, Madison, Wisconsin, 53706
| | - Rocky Blanco
- From the Department of Comparative Biosciences and.,the School of Veterinary Medicine, University of Wisconsin-Madison, Madison, Wisconsin, 53706
| | - Rachel E Privett
- From the Department of Comparative Biosciences and.,the School of Veterinary Medicine, University of Wisconsin-Madison, Madison, Wisconsin, 53706
| | - Jae-Sung You
- From the Department of Comparative Biosciences and.,the School of Veterinary Medicine, University of Wisconsin-Madison, Madison, Wisconsin, 53706
| | - Troy A Hornberger
- From the Department of Comparative Biosciences and .,the School of Veterinary Medicine, University of Wisconsin-Madison, Madison, Wisconsin, 53706
| |
Collapse
|
27
|
Kathage B, Gehlert S, Ulbricht A, Lüdecke L, Tapia VE, Orfanos Z, Wenzel D, Bloch W, Volkmer R, Fleischmann BK, Fürst DO, Höhfeld J. The cochaperone BAG3 coordinates protein synthesis and autophagy under mechanical strain through spatial regulation of mTORC1. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2016; 1864:62-75. [PMID: 27756573 DOI: 10.1016/j.bbamcr.2016.10.007] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/24/2016] [Revised: 10/04/2016] [Accepted: 10/11/2016] [Indexed: 12/20/2022]
Abstract
The cochaperone BAG3 is a central protein homeostasis factor in mechanically strained mammalian cells. It mediates the degradation of unfolded and damaged forms of the actin-crosslinker filamin through chaperone-assisted selective autophagy (CASA). In addition, BAG3 stimulates filamin transcription in order to compensate autophagic disposal and to maintain the actin cytoskeleton under strain. Here we demonstrate that BAG3 coordinates protein synthesis and autophagy through spatial regulation of the mammalian target of rapamycin complex 1 (mTORC1). The cochaperone utilizes its WW domain to contact a proline-rich motif in the tuberous sclerosis protein TSC1 that functions as an mTORC1 inhibitor in association with TSC2. Interaction with BAG3 results in a recruitment of TSC complexes to actin stress fibers, where the complexes act on a subpopulation of mTOR-positive vesicles associated with the cytoskeleton. Local inhibition of mTORC1 is essential to initiate autophagy at sites of filamin unfolding and damage. At the same time, BAG3-mediated sequestration of TSC1/TSC2 relieves mTORC1 inhibition in the remaining cytoplasm, which stimulates protein translation. In human muscle, an exercise-induced association of TSC1 with the cytoskeleton coincides with mTORC1 activation in the cytoplasm. The spatial regulation of mTORC1 exerted by BAG3 apparently provides the basis for a simultaneous induction of autophagy and protein synthesis to maintain the proteome under mechanical strain.
Collapse
Affiliation(s)
- Barbara Kathage
- Institute for Cell Biology, University of Bonn, Ulrich-Haberland-Str. 61a, 53121 Bonn, Germany
| | - Sebastian Gehlert
- German Sport University Cologne, Department of Molecular and Cellular Sport Medicine, Am Sportpark Müngersdorf 6, 50933 Cologne, Germany
| | - Anna Ulbricht
- Institute for Cell Biology, University of Bonn, Ulrich-Haberland-Str. 61a, 53121 Bonn, Germany
| | - Laura Lüdecke
- Institute for Cell Biology, University of Bonn, Ulrich-Haberland-Str. 61a, 53121 Bonn, Germany
| | - Victor E Tapia
- Department of Medicinal Immunology, Charité - University Medicine Berlin, Augustenburger Platz 1, 13353, Berlin, Germany
| | - Zacharias Orfanos
- Institute for Cell Biology, University of Bonn, Ulrich-Haberland-Str. 61a, 53121 Bonn, Germany
| | - Daniela Wenzel
- Institute of Physiology I, Life & Brain Center, University Clinic Bonn, Sigmund Freud Str. 25, 53105 Bonn, Germany
| | - Wilhelm Bloch
- German Sport University Cologne, Department of Molecular and Cellular Sport Medicine, Am Sportpark Müngersdorf 6, 50933 Cologne, Germany
| | - Rudolf Volkmer
- Department of Medicinal Immunology, Charité - University Medicine Berlin, Augustenburger Platz 1, 13353, Berlin, Germany
| | - Bernd K Fleischmann
- Institute of Physiology I, Life & Brain Center, University Clinic Bonn, Sigmund Freud Str. 25, 53105 Bonn, Germany
| | - Dieter O Fürst
- Institute for Cell Biology, University of Bonn, Ulrich-Haberland-Str. 61a, 53121 Bonn, Germany
| | - Jörg Höhfeld
- Institute for Cell Biology, University of Bonn, Ulrich-Haberland-Str. 61a, 53121 Bonn, Germany.
| |
Collapse
|
28
|
Camera DM, Smiles WJ, Hawley JA. Exercise-induced skeletal muscle signaling pathways and human athletic performance. Free Radic Biol Med 2016; 98:131-143. [PMID: 26876650 DOI: 10.1016/j.freeradbiomed.2016.02.007] [Citation(s) in RCA: 82] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2015] [Revised: 01/28/2016] [Accepted: 02/03/2016] [Indexed: 12/18/2022]
Abstract
Skeletal muscle is a highly malleable tissue capable of altering its phenotype in response to external stimuli including exercise. This response is determined by the mode, (endurance- versus resistance-based), volume, intensity and frequency of exercise performed with the magnitude of this response-adaptation the basis for enhanced physical work capacity. However, training-induced adaptations in skeletal muscle are variable and unpredictable between individuals. With the recent application of molecular techniques to exercise biology, there has been a greater understanding of the multiplicity and complexity of cellular networks involved in exercise responses. This review summarizes the molecular and cellular events mediating adaptation processes in skeletal muscle in response to exercise. We discuss established and novel cell signaling proteins mediating key physiological responses associated with enhanced exercise performance and the capacity for reactive oxygen and nitrogen species to modulate training adaptation responses. We also examine the molecular bases underpinning heterogeneous responses to resistance and endurance exercise and the dissociation between molecular 'markers' of training adaptation and subsequent exercise performance.
Collapse
Affiliation(s)
- Donny M Camera
- Centre for Exercise and Nutrition, Mary MacKillop Institute for Health Research, Australian Catholic University, Melbourne, Vic. 3065, Australia
| | - William J Smiles
- Centre for Exercise and Nutrition, Mary MacKillop Institute for Health Research, Australian Catholic University, Melbourne, Vic. 3065, Australia
| | - John A Hawley
- Centre for Exercise and Nutrition, Mary MacKillop Institute for Health Research, Australian Catholic University, Melbourne, Vic. 3065, Australia; Research Institute for Sport and Exercise Sciences, Liverpool John Moores University, Liverpool L3 3AF, United Kingdom.
| |
Collapse
|
29
|
Teixeira GR, Gobbo LA, Santos NJD, Araújo RGD, Santos CCD, Malheiro OCDM, Castoldi RC, Camargo-Filho JCS, Papoti M. The effect of β-hydroxy-β-methylbutyrate (HMB) on the morphology of skeletal muscle after concurrent training. MOTRIZ: REVISTA DE EDUCACAO FISICA 2016. [DOI: 10.1590/s1980-6574201600030010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
|
30
|
Abstract
Skeletal muscle homeostasis is regulated by a constant influx of chemicals and exposure to mechanical stimuli. A number of key signaling pathways that translate these stimuli into changes in muscle physiology have been established. The GPCR family known as adhesion GPCRs (aGPCRs) has largely elusive roles in skeletal muscle biology; however, their unique capacity to activate adhesion and G protein signaling pathways makes them an attractive point of investigation. The skeletal muscle myofiber contains a highly organized cytoarchitecture to ensure contractile function. This requires intricate interactions with components of the extracellular matrix (ECM) surrounding each fiber. aGPCRs possess extended N-termini known to interact with ECM proteins and complexes suggesting a compatible role in skeletal muscle biology. Furthermore, recent work demonstrated the involvement of certain aGPCRs in whole muscle hypertrophy and differentiation of muscle progenitor cells. Signaling pathways downstream of aGPCRs are still incompletely understood; however, initial findings show involvement of the Gα12/13 subunit signaling to the pro-anabolic Akt/mTOR pathway. Together, this chapter will review the emerging role of aGPCRs in skeletal muscle biology and putative mechanism(s) employed to regulate skeletal muscle growth.
Collapse
Affiliation(s)
- James P White
- Duke University Medical Center, 300 North Duke Street, Durham, NC, 27701, USA.
| |
Collapse
|
31
|
Nagano K. Alteration of cathepsin-D expression in atrophied muscles and apoptotic myofibers by hindlimb unloading in a low-temperature environment. J Phys Ther Sci 2015; 27:3585-91. [PMID: 26696743 PMCID: PMC4681950 DOI: 10.1589/jpts.27.3585] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2015] [Accepted: 08/24/2015] [Indexed: 01/20/2023] Open
Abstract
[Purpose] The purpose of this study was to elucidate the cathepsin-D involvement in
signaling pathways for the survival and apoptosis of myofibers in rats with
hindlimb-unloading in a low-temperature environment. [Subjects and Methods] Wistar rats
were divided into two groups: a control group and a group that underwent hindlimb
unloading in a low-temperature environment to induce muscle apoptosis. Cathepsin-D
localization in the soleus and extensor digitorum longus muscles, along with the
expression of cathepsin-D in apoptotic myofibers, was examined. Expression of the active
and inactive forms of cathepsin-D was also analyzed. [Results] Cathepsin-D was mainly
expressed in type I myofibers and was observed to have punctate patterns in the control
group. In the hindlimb unloading in a low-temperature environment group, the type I
myofiber composition ratio decreased, and caspase-3 activation and TUNEL-positive
apoptotic myofibers were observed. In caspase-3-activated myofibers, cathepsin-D
overexpression and leakage of it into the cytoplasm were observed. In the hindlimb
unloading in a low-temperature environment group, the amount of inactive cathepsin-D
decreased, whereas that of the active form increased. [Conclusion] Cathepsin-D was deduced
to be indicative of a myofiber-type classification and a factor related to myofiber type
maintenance. In addition, cathepsin-D leakage into the cytoplasm was appeared to be
involved in caspase-3 activation in the hindlimb unloading in a low-temperature
environment group.
Collapse
Affiliation(s)
- Katsuhito Nagano
- Department of Physical Therapy, Faculty of Rehabilitation, Fukui College of Health Sciences, Japan
| |
Collapse
|
32
|
Bakker AD, Gakes T, Hogervorst JMA, de Wit GMJ, Klein-Nulend J, Jaspers RT. Mechanical Stimulation and IGF-1 Enhance mRNA Translation Rate in Osteoblasts Via Activation of the AKT-mTOR Pathway. J Cell Physiol 2015; 231:1283-90. [PMID: 26505782 DOI: 10.1002/jcp.25228] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2015] [Accepted: 10/26/2015] [Indexed: 01/31/2023]
Abstract
Insulin-like growth factor-1 (IGF-1) is anabolic for muscle by enhancing the rate of mRNA translation via activation of AKT and subsequent activation of the mammalian target of rapamycin complex 1 (mTOR), thereby increasing cellular protein production. IGF-1 is also anabolic for bone, but whether the mTOR pathway plays a role in the rate of bone matrix protein production by osteoblasts is unknown. We hypothesized that anabolic stimuli such as mechanical loading and IGF-1 stimulate protein synthesis in osteoblasts via activation of the AKT-mTOR pathway. MC3T3-E1 osteoblasts were either or not subjected for 1 h to mechanical loading by pulsating fluid flow (PFF) or treated with or without human recombinant IGF-1 (1-100 ng/ml) for 0.5-6 h, to determine phosphorylation of AKT and p70S6K (downstream of mTOR) by Western blot. After 4 days of culture with or without the mTOR inhibitor rapamycin, total protein, DNA, and gene expression were quantified. IGF-1 (100 ng/ml) reduced IGF-1 gene expression, although PFF enhanced IGF-1 expression. IGF-1 did not affect collagen-I gene expression. IGF-1 dose-dependently enhanced AKT and p70S6K phosphorylation at 2 and 6 h. PFF enhanced phosphorylation of AKT and p70S6K already within 1 h. Both IGF-1 and PFF enhanced total protein per cell by ∼30%, but not in the presence of rapamycin. Our results show that IGF-1 and PFF activate mTOR, thereby stimulating the rate of mRNA translation in osteoblasts. The known anabolic effect of mechanical loading and IGF-1 on bone may thus be partly explained by mTOR-mediated enhanced protein synthesis in osteoblasts.
Collapse
Affiliation(s)
- Astrid D Bakker
- Department of Oral Cell Biology, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and VU University Amsterdam, MOVE Research Institute Amsterdam, Amsterdam, The Netherlands
| | - Tom Gakes
- Department of Oral Cell Biology, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and VU University Amsterdam, MOVE Research Institute Amsterdam, Amsterdam, The Netherlands
| | - Jolanda M A Hogervorst
- Department of Oral Cell Biology, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and VU University Amsterdam, MOVE Research Institute Amsterdam, Amsterdam, The Netherlands
| | - Gerard M J de Wit
- Laboratory for Myology, Research Institute MOVE Amsterdam, Faculty of Behavioural and Movement Sciences, Vrije Universiteit Amsterdam, The Netherlands
| | - Jenneke Klein-Nulend
- Department of Oral Cell Biology, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and VU University Amsterdam, MOVE Research Institute Amsterdam, Amsterdam, The Netherlands
| | - Richard T Jaspers
- Laboratory for Myology, Research Institute MOVE Amsterdam, Faculty of Behavioural and Movement Sciences, Vrije Universiteit Amsterdam, The Netherlands
| |
Collapse
|
33
|
Goodman CA, Hornberger TA, Robling AG. Bone and skeletal muscle: Key players in mechanotransduction and potential overlapping mechanisms. Bone 2015; 80:24-36. [PMID: 26453495 PMCID: PMC4600534 DOI: 10.1016/j.bone.2015.04.014] [Citation(s) in RCA: 107] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2014] [Revised: 03/18/2015] [Accepted: 04/07/2015] [Indexed: 12/16/2022]
Abstract
The development and maintenance of skeletal muscle and bone mass is critical for movement, health and issues associated with the quality of life. Skeletal muscle and bone mass are regulated by a variety of factors that include changes in mechanical loading. Moreover, bone mass is, in large part, regulated by muscle-derived mechanical forces and thus by changes in muscle mass/strength. A thorough understanding of the cellular mechanism(s) responsible for mechanotransduction in bone and skeletal muscle is essential for the development of effective exercise and pharmaceutical strategies aimed at increasing, and/or preventing the loss of, mass in these tissues. Thus, in this review we will attempt to summarize the current evidence for the major molecular mechanisms involved in mechanotransduction in skeletal muscle and bone. By examining the differences and similarities in mechanotransduction between these two tissues, it is hoped that this review will stimulate new insights and ideas for future research and promote collaboration between bone and muscle biologists.(1).
Collapse
Affiliation(s)
- Craig A Goodman
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI 53706, USA; Centre for Chronic Disease Prevention and Management, College of Health and Biomedicine, Victoria University, Melbourne, Australia; Institute of Sport, Exercise and Active Living (ISEAL), Victoria University, Melbourne, VIC, Australia.
| | - Troy A Hornberger
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Alexander G Robling
- Department of Anatomy and Cell Biology, Indiana University School of Medicine, Indianapolis, IN 46202, USA; Roudebush Veterans Affairs Medical Center, Indianapolis, IN 46202, USA; Department of Biomedical Engineering, Indiana University-Purdue University at Indianapolis, Indianapolis, IN 46202, USA
| |
Collapse
|
34
|
McGlory C, Phillips SM. Exercise and the Regulation of Skeletal Muscle Hypertrophy. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2015; 135:153-73. [PMID: 26477914 DOI: 10.1016/bs.pmbts.2015.06.018] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Skeletal muscle is a critical organ serving as the primary site for postprandial glucose disposal and the generation of contractile force. The size of human skeletal muscle mass is dependent upon the temporal relationship between changes in muscle protein synthesis (MPS) and muscle protein breakdown. The aim of this chapter is to review our current understanding of how resistance exercise influences protein turnover with a specific emphasis on the molecular factors regulating MPS. We also will discuss recent data relating to the prescription of resistance exercise to maximize skeletal muscle hypertrophy. Finally, we evaluate the impact of age and periods of disuse on the loss of muscle mass and the controversy surround the etiology of muscle disuse atrophy.
Collapse
Affiliation(s)
- Chris McGlory
- Department of Kinesiology, McMaster University, Hamilton, Ontario, Canada
| | - Stuart M Phillips
- Department of Kinesiology, McMaster University, Hamilton, Ontario, Canada.
| |
Collapse
|
35
|
McGlory C, Phillips SM. Assessing the regulation of skeletal muscle plasticity in response to protein ingestion and resistance exercise: recent developments. Curr Opin Clin Nutr Metab Care 2014; 17:412-7. [PMID: 25105927 DOI: 10.1097/mco.0000000000000083] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
PURPOSE OF REVIEW The main purpose of this review is to discuss novel methodological advances in the assessment of muscle protein synthesis (MPS) in response to protein feeding and resistance exercise. RECENT FINDINGS In the past 20 years, there has been a shift from application of the nitrogen balance methods toward the infusion of stable isotopic tracers to assess rates of MPS in response to a range of perturbations. Although this approach has enabled MPS to be assessed with a greater temporal resolution and precision, the method limits the capture of MPS to relatively short-duration infusions of approximately 3-12 h. Recent refinement of analytical methods to assess long-term MPS responses have now provided a platform for studying the impact of exercise and nutrition on muscle anabolism with an extended temporal resolution from hours to days or even weeks. Finally, novel insights into cellular signaling processes may help delineate the molecular mechanisms that govern skeletal muscle plasticity in response to exercise and feeding. SUMMARY Future work should focus on the impact of novel exercise and nutritional interventions on MPS in an extended postexercise adaptive period, that is, days. The findings of such investigations will help test the long-term efficacy of interventions to enhance skeletal muscle protein reconditioning and hypertrophy.
Collapse
Affiliation(s)
- Chris McGlory
- Department of Kinesiology, Exercise Metabolism Research Group, McMaster University, Hamilton, Ontario, USA
| | | |
Collapse
|
36
|
Goodman CA, Hornberger TA. New roles for Smad signaling and phosphatidic acid in the regulation of skeletal muscle mass. F1000PRIME REPORTS 2014; 6:20. [PMID: 24765525 PMCID: PMC3974568 DOI: 10.12703/p6-20] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Skeletal muscle is essential for normal bodily function and the loss of skeletal muscle (i.e. muscle atrophy/wasting) can have a major impact on mobility, whole-body metabolism, disease resistance, and quality of life. Thus, there is a clear need for the development of therapies that can prevent the loss, or increase, of skeletal muscle mass. However, in order to develop such therapies, we will first have to develop a thorough understanding of the molecular mechanisms that regulate muscle mass. Fortunately, our knowledge is rapidly advancing, and in this review, we will summarize recent studies that have expanded our understanding of the roles that Smad signaling and the synthesis of phosphatidic acid play in the regulation of skeletal muscle mass.
Collapse
|
37
|
Ottenheijm CAC, Jaspers RT, Wüst RCI, van der Velden J. Muscle physiology: move to translation. J Muscle Res Cell Motil 2014; 35:1-2. [PMID: 24639084 DOI: 10.1007/s10974-014-9381-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Coen A C Ottenheijm
- Department of Physiology, Institute for Cardiovascular Research (ICaR-VU), VU University Medical Center, van der Boechorststraat 7, 1081 BT, Amsterdam, The Netherlands
| | | | | | | |
Collapse
|
38
|
You JS, Lincoln HC, Kim CR, Frey JW, Goodman CA, Zhong XP, Hornberger TA. The role of diacylglycerol kinase ζ and phosphatidic acid in the mechanical activation of mammalian target of rapamycin (mTOR) signaling and skeletal muscle hypertrophy. J Biol Chem 2013; 289:1551-63. [PMID: 24302719 DOI: 10.1074/jbc.m113.531392] [Citation(s) in RCA: 112] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
The activation of mTOR signaling is essential for mechanically induced changes in skeletal muscle mass, and previous studies have suggested that mechanical stimuli activate mTOR (mammalian target of rapamycin) signaling through a phospholipase D (PLD)-dependent increase in the concentration of phosphatidic acid (PA). Consistent with this conclusion, we obtained evidence which further suggests that mechanical stimuli utilize PA as a direct upstream activator of mTOR signaling. Unexpectedly though, we found that the activation of PLD is not necessary for the mechanically induced increases in PA or mTOR signaling. Motivated by this observation, we performed experiments that were aimed at identifying the enzyme(s) that promotes the increase in PA. These experiments revealed that mechanical stimulation increases the concentration of diacylglycerol (DAG) and the activity of DAG kinases (DGKs) in membranous structures. Furthermore, using knock-out mice, we determined that the ζ isoform of DGK (DGKζ) is necessary for the mechanically induced increase in PA. We also determined that DGKζ significantly contributes to the mechanical activation of mTOR signaling, and this is likely driven by an enhanced binding of PA to mTOR. Last, we found that the overexpression of DGKζ is sufficient to induce muscle fiber hypertrophy through an mTOR-dependent mechanism, and this event requires DGKζ kinase activity (i.e. the synthesis of PA). Combined, these results indicate that DGKζ, but not PLD, plays an important role in mechanically induced increases in PA and mTOR signaling. Furthermore, this study suggests that DGKζ could be a fundamental component of the mechanism(s) through which mechanical stimuli regulate skeletal muscle mass.
Collapse
Affiliation(s)
- Jae-Sung You
- From the Program in Cellular and Molecular Biology and
| | | | | | | | | | | | | |
Collapse
|
39
|
The role of mTORC1 in regulating protein synthesis and skeletal muscle mass in response to various mechanical stimuli. Rev Physiol Biochem Pharmacol 2013; 166:43-95. [PMID: 24442322 DOI: 10.1007/112_2013_17] [Citation(s) in RCA: 93] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Skeletal muscle plays a fundamental role in mobility, disease prevention, and quality of life. Skeletal muscle mass is, in part, determined by the rates of protein synthesis, and mechanical loading is a major regulator of protein synthesis and skeletal muscle mass. The mammalian/mechanistic target of rapamycin (mTOR), found in the multi-protein complex, mTORC1, is proposed to play an essential role in the regulation of protein synthesis and skeletal muscle mass. The purpose of this review is to examine the function of mTORC1 in relation to protein synthesis and cell growth, the current evidence from rodent and human studies for the activation of mTORC1 signaling by different types of mechanical stimuli, whether mTORC1 signaling is necessary for changes in protein synthesis and skeletal muscle mass that occur in response to different types of mechanical stimuli, and the proposed molecular signaling mechanisms that may be responsible for the mechanical activation of mTORC1 signaling.
Collapse
|