1
|
Thomas ACQ, Stead CA, Burniston JG, Phillips SM. Exercise-specific adaptations in human skeletal muscle: Molecular mechanisms of making muscles fit and mighty. Free Radic Biol Med 2024; 223:341-356. [PMID: 39147070 DOI: 10.1016/j.freeradbiomed.2024.08.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 07/30/2024] [Accepted: 08/09/2024] [Indexed: 08/17/2024]
Abstract
The mechanisms leading to a predominantly hypertrophied phenotype versus a predominantly oxidative phenotype, the hallmarks of resistance training (RT) or aerobic training (AT), respectively, are being unraveled. In humans, exposure of naïve persons to either AT or RT results in their skeletal muscle exhibiting generic 'exercise stress-related' signaling, transcription, and translation responses. However, with increasing engagement in AT or RT, the responses become refined, and the phenotype typically associated with each form of exercise emerges. Here, we review some of the mechanisms underpinning the adaptations of how muscles become, through AT, 'fit' and RT, 'mighty.' Much of our understanding of molecular exercise physiology has arisen from targeted analysis of post-translational modifications and measures of protein synthesis. Phosphorylation of specific residue sites has been a dominant focus, with canonical signaling pathways (AMPK and mTOR) studied extensively in the context of AT and RT, respectively. These alone, along with protein synthesis, have only begun to elucidate key differences in AT and RT signaling. Still, key yet uncharacterized differences exist in signaling and regulation of protein synthesis that drive unique adaptation to AT and RT. Omic studies are required to better understand the divergent relationship between exercise and phenotypic outcomes of training.
Collapse
Affiliation(s)
- Aaron C Q Thomas
- Protein Metabolism Research Lab, Department of Kinesiology, McMaster University, Hamilton, ON, Canada; Research Institute for Sport & Exercise Sciences, Liverpool John Moores University, Liverpool, United Kingdom
| | - Connor A Stead
- Research Institute for Sport & Exercise Sciences, Liverpool John Moores University, Liverpool, United Kingdom
| | - Jatin G Burniston
- Research Institute for Sport & Exercise Sciences, Liverpool John Moores University, Liverpool, United Kingdom
| | - Stuart M Phillips
- Protein Metabolism Research Lab, Department of Kinesiology, McMaster University, Hamilton, ON, Canada.
| |
Collapse
|
2
|
Hannaian SJ, Lov J, Cheng-Boivin Z, Abou Sawan S, Hodson N, Gentil BJ, Morais JA, Churchward-Venne TA. Acute effects of a ketone monoester, whey protein, or their coingestion on mTOR trafficking and protein-protein colocalization in human skeletal muscle. Am J Physiol Cell Physiol 2024; 326:C1769-C1775. [PMID: 38682238 PMCID: PMC11371313 DOI: 10.1152/ajpcell.00207.2024] [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: 03/28/2024] [Revised: 04/22/2024] [Accepted: 04/22/2024] [Indexed: 05/01/2024]
Abstract
We recently demonstrated that acute oral ketone monoester intake induces a stimulation of postprandial myofibrillar protein synthesis rates comparable to that elicited following the ingestion of 10 g whey protein or their coingestion. The present investigation aimed to determine the acute effects of ingesting a ketone monoester, whey protein, or their coingestion on mechanistic target of rapamycin (mTOR)-related protein-protein colocalization and intracellular trafficking in human skeletal muscle. In a randomized, double-blind, parallel group design, 36 healthy recreationally active young males (age: 24.2 ± 4.1 yr) ingested either: 1) 0.36 g·kg-1 bodyweight of the ketone monoester (R)-3-hydroxybutyl (R)-3-hydroxybutyrate (KET), 2) 10 g whey protein (PRO), or 3) the combination of both (KET + PRO). Muscle biopsies were obtained in the overnight postabsorptive state (basal conditions), and at 120 and 300 min in the postprandial period for immunofluorescence assessment of protein translocation and colocalization of mTOR-related signaling molecules. All treatments resulted in a significant (Interaction: P < 0.0001) decrease in tuberous sclerosis complex 2 (TSC2)-Ras homolog enriched in brain (Rheb) colocalization at 120 min versus basal; however, the decrease was sustained at 300 min versus basal (P < 0.0001) only in KET + PRO. PRO and KET + PRO increased (Interaction: P < 0.0001) mTOR-Rheb colocalization at 120 min versus basal; however, KET + PRO resulted in a sustained increase in mTOR-Rheb colocalization at 300 min that was greater than KET and PRO. Treatment intake increased mTOR-wheat germ agglutinin (WGA) colocalization at 120 and 300 min (Time: P = 0.0031), suggesting translocation toward the fiber periphery. These findings demonstrate that ketone monoester intake can influence the spatial mechanisms involved in the regulation of mTORC1 in human skeletal muscle.NEW & NOTEWORTHY We explored the effects of a ketone monoester (KET), whey protein (PRO), or their coingestion (KET + PRO) on mTOR-related protein-protein colocalization and intracellular trafficking in human muscle. All treatments decreased TSC2-Rheb colocalization at 120 minutes; however, KET + PRO sustained the decrease at 300 min. Only PRO and KET + PRO increased mTOR-Rheb colocalization; however, the increase at 300 min was greater in KET + PRO. Treatment intake increased mTOR-WGA colocalization, suggesting translocation to the fiber periphery. Ketone bodies influence the spatial regulation of mTOR.
Collapse
Affiliation(s)
- Sarkis J Hannaian
- Department of Kinesiology and Physical Education, McGill University, Montreal, Quebec, Canada
- Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
| | - Jamie Lov
- Department of Kinesiology and Physical Education, McGill University, Montreal, Quebec, Canada
| | - Zacharie Cheng-Boivin
- Department of Kinesiology and Physical Education, McGill University, Montreal, Quebec, Canada
| | | | - Nathan Hodson
- Department of Sport and Exercise Science, Institute of Sport, Manchester Metropolitan University, Manchester, United Kingdom
| | - Benoit J Gentil
- Department of Kinesiology and Physical Education, McGill University, Montreal, Quebec, Canada
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada
| | - José A Morais
- Department of Kinesiology and Physical Education, McGill University, Montreal, Quebec, Canada
- Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
- Department of Medicine, Division of Geriatric Medicine, McGill University, Montreal, Quebec, Canada
| | - Tyler A Churchward-Venne
- Department of Kinesiology and Physical Education, McGill University, Montreal, Quebec, Canada
- Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
- Department of Medicine, Division of Geriatric Medicine, McGill University, Montreal, Quebec, Canada
| |
Collapse
|
3
|
Pizzol MSD, Ibelli AMG, Cantão ME, Campos FG, de Oliveira HC, de Oliveira Peixoto J, Fernandes LT, de Castro Tavernari F, Morés MAZ, Bastos APA, Ledur MC. Differential expression of miRNAs associated with pectoral myopathies in young broilers: insights from a comparative transcriptome analysis. BMC Genomics 2024; 25:104. [PMID: 38262955 PMCID: PMC10807067 DOI: 10.1186/s12864-024-09983-9] [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: 08/07/2023] [Accepted: 01/06/2024] [Indexed: 01/25/2024] Open
Abstract
INTRODUCTION White Striping (WS) and Wooden Breast (WB) pectoral myopathies are relevant disorders for contemporary broiler production worldwide. Several studies aimed to elucidate the genetic components associated with the occurrence of these myopathies. However, epigenetic factors that trigger or differentiate these two conditions are still unclear. The aim of this study was to identify miRNAs differentially expressed (DE) between normal and WS and WB-affected broilers, and to verify the possible role of these miRNAs in metabolic pathways related to the manifestation of these pectoral myopathies in 28-day-old broilers. RESULTS Five miRNAs were DE in the WS vs control (gga-miR-375, gga-miR-200b-3p, gga-miR-429-3p, gga-miR-1769-5p, gga-miR-200a-3p), 82 between WB vs control and 62 between WB vs WS. Several known miRNAs were associated with WB, such as gga-miR-155, gga-miR-146b, gga-miR-222, gga-miR-146-5p, gga-miR- 29, gga-miR-21-5p, gga-miR-133a-3p and gga-miR-133b. Most of them had not previously been associated with the development of this myopathy in broilers. We also have predicted 17 new miRNAs expressed in the broilers pectoral muscle. DE miRNA target gene ontology analysis enriched 6 common pathways for WS and WB compared to control: autophagy, insulin signaling, FoxO signaling, endocytosis, and metabolic pathways. The WS vs control contrast had two unique pathways, ERBB signaling and the mTOR signaling, while WB vs control had 14 unique pathways, with ubiquitin-mediated proteolysis and endoplasmic reticulum protein processing being the most significant. CONCLUSIONS We found miRNAs DE between normal broilers and those affected with breast myopathies at 28 days of age. Our results also provide novel evidence of the miRNAs role on the regulation of WS and in the differentiation of both WS and WB myopathies. Overall, our study provides insights into miRNA-mediated and pathways involved in the occurrence of WS and WB helping to better understand these chicken growth disorders in an early age. These findings can help developing new approaches to reduce these complex issues in poultry production possibly by adjustments in nutrition and management conditions. Moreover, the miRNAs and target genes associated with the initial stages of WS and WB development could be potential biomarkers to be used in selection to reduce the occurrence of these myopathies in broiler production.
Collapse
Affiliation(s)
- Mariane Spudeit Dal Pizzol
- Programa de Pós-Graduação em Zootecnia, Universidade do Estado de Santa Catarina, UDESC-Oeste, Chapecó, Santa Catarina, Brazil
| | - Adriana Mércia Guaratini Ibelli
- Embrapa Suínos e Aves, Concórdia, Santa Catarina, Brazil
- Programa de Pós-Graduação em Ciências Veterinárias, Universidade Estadual do Centro Oeste, Guarapuava, Paraná, Brazil
- Present Address: Embrapa Pecuária Sudeste, São Carlos, São Paulo, Brazil
| | | | - Francelly Geralda Campos
- Departamento de Zootecnia, Programa de Pós- Graduação em Zootecnia, Universidade Federal de Viçosa, Viçosa, Minas Gerais, Brazil
| | - Haniel Cedraz de Oliveira
- Departamento de Zootecnia, Programa de Pós- Graduação em Zootecnia, Universidade Federal de Viçosa, Viçosa, Minas Gerais, Brazil
| | - Jane de Oliveira Peixoto
- Embrapa Suínos e Aves, Concórdia, Santa Catarina, Brazil
- Programa de Pós-Graduação em Ciências Veterinárias, Universidade Estadual do Centro Oeste, Guarapuava, Paraná, Brazil
| | | | - Fernando de Castro Tavernari
- Programa de Pós-Graduação em Zootecnia, Universidade do Estado de Santa Catarina, UDESC-Oeste, Chapecó, Santa Catarina, Brazil
- Embrapa Suínos e Aves, Concórdia, Santa Catarina, Brazil
| | | | - Ana Paula Almeida Bastos
- Embrapa Suínos e Aves, Concórdia, Santa Catarina, Brazil
- Programa de Pós-Graduação em Ciências Veterinárias, Universidade Estadual do Centro Oeste, Guarapuava, Paraná, Brazil
| | - Mônica Corrêa Ledur
- Programa de Pós-Graduação em Zootecnia, Universidade do Estado de Santa Catarina, UDESC-Oeste, Chapecó, Santa Catarina, Brazil.
- Embrapa Suínos e Aves, Concórdia, Santa Catarina, Brazil.
| |
Collapse
|
4
|
Kataoka R, Hammert WB, Yamada Y, Song JS, Seffrin A, Kang A, Spitz RW, Wong V, Loenneke JP. The Plateau in Muscle Growth with Resistance Training: An Exploration of Possible Mechanisms. Sports Med 2024; 54:31-48. [PMID: 37787845 DOI: 10.1007/s40279-023-01932-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/01/2023] [Indexed: 10/04/2023]
Abstract
It is hypothesized that there is likely a finite ability for muscular adaptation. While it is difficult to distinguish between a true plateau following a long-term training period and short-term stalling in muscle growth, a plateau in muscle growth has been attributed to reaching a genetic potential, with limited discussion on what might physiologically contribute to this muscle growth plateau. The present paper explores potential physiological factors that may drive the decline in muscle growth after prolonged resistance training. Overall, with chronic training, the anabolic signaling pathways may become more refractory to loading. While measures of anabolic markers may have some predictive capabilities regarding muscle growth adaptation, they do not always demonstrate a clear connection. Catabolic processes may also constrain the ability to achieve further muscle growth, which is influenced by energy balance. Although speculative, muscle cells may also possess cell scaling mechanisms that sense and regulate their own size, along with molecular brakes that hinder growth rate over time. When considering muscle growth over the lifespan, there comes a point when the anabolic response is attenuated by aging, regardless of whether or not individuals approach their muscle growth potential. Our goal is that the current review opens avenues for future experimental studies to further elucidate potential mechanisms to explain why muscle growth may plateau.
Collapse
Affiliation(s)
- Ryo Kataoka
- Department of Health, Exercise Science, and Recreation Management, Kevser Ermin Applied Physiology Laboratory, The University of Mississippi, P.O. Box 1848, University, MS, 38677, USA
| | - William B Hammert
- Department of Health, Exercise Science, and Recreation Management, Kevser Ermin Applied Physiology Laboratory, The University of Mississippi, P.O. Box 1848, University, MS, 38677, USA
| | - Yujiro Yamada
- Department of Health, Exercise Science, and Recreation Management, Kevser Ermin Applied Physiology Laboratory, The University of Mississippi, P.O. Box 1848, University, MS, 38677, USA
| | - Jun Seob Song
- Department of Health, Exercise Science, and Recreation Management, Kevser Ermin Applied Physiology Laboratory, The University of Mississippi, P.O. Box 1848, University, MS, 38677, USA
| | - Aldo Seffrin
- Department of Health, Exercise Science, and Recreation Management, Kevser Ermin Applied Physiology Laboratory, The University of Mississippi, P.O. Box 1848, University, MS, 38677, USA
| | - Anna Kang
- Department of Health, Exercise Science, and Recreation Management, Kevser Ermin Applied Physiology Laboratory, The University of Mississippi, P.O. Box 1848, University, MS, 38677, USA
| | - Robert W Spitz
- Department of Health, Exercise Science, and Recreation Management, Kevser Ermin Applied Physiology Laboratory, The University of Mississippi, P.O. Box 1848, University, MS, 38677, USA
| | - Vickie Wong
- Department of Health, Exercise Science, and Recreation Management, Kevser Ermin Applied Physiology Laboratory, The University of Mississippi, P.O. Box 1848, University, MS, 38677, USA
| | - Jeremy P Loenneke
- Department of Health, Exercise Science, and Recreation Management, Kevser Ermin Applied Physiology Laboratory, The University of Mississippi, P.O. Box 1848, University, MS, 38677, USA.
| |
Collapse
|
5
|
Roberts MD, McCarthy JJ, Hornberger TA, Phillips SM, Mackey AL, Nader GA, Boppart MD, Kavazis AN, Reidy PT, Ogasawara R, Libardi CA, Ugrinowitsch C, Booth FW, Esser KA. Mechanisms of mechanical overload-induced skeletal muscle hypertrophy: current understanding and future directions. Physiol Rev 2023; 103:2679-2757. [PMID: 37382939 PMCID: PMC10625844 DOI: 10.1152/physrev.00039.2022] [Citation(s) in RCA: 39] [Impact Index Per Article: 39.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 06/12/2023] [Accepted: 06/21/2023] [Indexed: 06/30/2023] Open
Abstract
Mechanisms underlying mechanical overload-induced skeletal muscle hypertrophy have been extensively researched since the landmark report by Morpurgo (1897) of "work-induced hypertrophy" in dogs that were treadmill trained. Much of the preclinical rodent and human resistance training research to date supports that involved mechanisms include enhanced mammalian/mechanistic target of rapamycin complex 1 (mTORC1) signaling, an expansion in translational capacity through ribosome biogenesis, increased satellite cell abundance and myonuclear accretion, and postexercise elevations in muscle protein synthesis rates. However, several lines of past and emerging evidence suggest that additional mechanisms that feed into or are independent of these processes are also involved. This review first provides a historical account of how mechanistic research into skeletal muscle hypertrophy has progressed. A comprehensive list of mechanisms associated with skeletal muscle hypertrophy is then outlined, and areas of disagreement involving these mechanisms are presented. Finally, future research directions involving many of the discussed mechanisms are proposed.
Collapse
Affiliation(s)
- Michael D Roberts
- School of Kinesiology, Auburn University, Auburn, Alabama, United States
| | - John J McCarthy
- Department of Physiology, College of Medicine, University of Kentucky, Lexington, Kentucky, United States
| | - Troy A Hornberger
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, Wisconsin, United States
| | - Stuart M Phillips
- Department of Kinesiology, McMaster University, Hamilton, Ontario, Canada
| | - Abigail L Mackey
- Institute of Sports Medicine Copenhagen, Department of Orthopedic Surgery, Copenhagen University Hospital-Bispebjerg and Frederiksberg, and Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Gustavo A Nader
- Department of Kinesiology and Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, Pennsylvania, United States
| | - Marni D Boppart
- Department of Kinesiology and Community Health, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States
| | - Andreas N Kavazis
- School of Kinesiology, Auburn University, Auburn, Alabama, United States
| | - Paul T Reidy
- Department of Kinesiology, Nutrition and Health, Miami University, Oxford, Ohio, United States
| | - Riki Ogasawara
- Healthy Food Science Research Group, Cellular and Molecular Biotechnology Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba, Japan
| | - Cleiton A Libardi
- MUSCULAB-Laboratory of Neuromuscular Adaptations to Resistance Training, Department of Physical Education, Federal University of São Carlos, São Carlos, Brazil
| | - Carlos Ugrinowitsch
- School of Physical Education and Sport, University of São Paulo, São Paulo, Brazil
| | - Frank W Booth
- Department of Biomedical Sciences, University of Missouri, Columbia, Missouri, United States
| | - Karyn A Esser
- Department of Physiology and Aging, College of Medicine, University of Florida, Gainesville, Florida, United States
| |
Collapse
|
6
|
Leucine ingestion promotes mTOR translocation to the periphery and enhances total and peripheral RPS6 phosphorylation in human skeletal muscle. Amino Acids 2023; 55:253-261. [PMID: 36474017 DOI: 10.1007/s00726-022-03221-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Accepted: 11/24/2022] [Indexed: 12/12/2022]
Abstract
The activation of the mechanistic target of rapamycin complex 1 (mTORC1), a master regulator of protein synthesis, by anabolic stimuli (such as muscle contraction or essential amino acids) involves its translocation to the cell periphery. Leucine is generally considered the most anabolic of amino acids for its ability to independently modulate muscle protein synthesis. However, it is currently unknown if free leucine impacts region-specific mTORC1-mediated phosphorylation events and protein-protein interactions. In this clinical trial (NCT03952884; registered May 16, 2019), we used immunofluorescence methods to investigate the role of dietary leucine on the postprandial regulation of mTORC1 and ribosomal protein S6 (RPS6), an important downstream readout of mTORC1 activity. Eight young, healthy, recreationally active males (n = 8; 23 ± 3 yrs) ingested 2 g of leucine with vastus lateralis biopsies collected at baseline, 30, 60, and 180 min postprandial. Leucine promoted mTOR translocation to the periphery (~ 18-29%; p ≤ 0.012) and enhanced mTOR localization with the lysosome (~ 16%; both p = 0.049) at 30 and 60 min post-feeding. p-RPS6Ser240/244 staining intensity, a readout of mTORC1 activity, was significantly elevated at all postprandial timepoints in both the total fiber (~ 14-30%; p ≤ 0.032) and peripheral regions (~ 16-33%; p ≤ 0.014). Additionally, total and peripheral p-RPS6Ser240/244 staining intensity at 60 min was positively correlated (r = 0.74, p = 0.036; r = 0.80, p = 0.016, respectively) with rates of myofibrillar protein synthesis over 180 min. The ability of leucine to activate mTORC1 in peripheral regions favors an enhanced rate of MPS, as this is the intracellular space thought to be replete with the cellular machinery that facilitates this anabolic process.
Collapse
|
7
|
Tinline-Goodfellow CT, Lees MJ, Hodson N. The skeletal muscle fiber periphery: A nexus of mTOR-related anabolism. SPORTS MEDICINE AND HEALTH SCIENCE 2022; 5:10-19. [PMID: 36994172 PMCID: PMC10040390 DOI: 10.1016/j.smhs.2022.11.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 11/16/2022] [Accepted: 11/23/2022] [Indexed: 12/02/2022] Open
Abstract
Skeletal muscle anabolism is driven by numerous stimuli such as growth factors, nutrients (i.e., amino acids, glucose), and mechanical stress. These stimuli are integrated by the mechanistic target of rapamycin (mTOR) complex 1 (mTORC1) signal transduction cascade. In recent years, work from our laboratory and elsewhere has sought to unravel the molecular mechanisms underpinning the mTOR-related activation of muscle protein synthesis (MPS), as well as the spatial regulation of these mechanisms within the skeletal muscle cell. These studies have suggested that the skeletal muscle fiber periphery is a region of central importance in anabolism (i.e., growth/MPS). Indeed, the fiber periphery is replete with the substrates, molecular machinery, and translational apparatus necessary to facilitate MPS. This review provides a summary of the mechanisms underpinning the mTOR-associated activation of MPS from cell, rodent, and human studies. It also presents an overview of the spatial regulation of mTORC1 in response to anabolic stimuli and outlines the factors that distinguish the periphery of the cell as a highly notable region of skeletal muscle for the induction of MPS. Future research should seek to further explore the nutrient-induced activation of mTORC1 at the periphery of skeletal muscle fibers.
Collapse
Affiliation(s)
| | - Matthew J. Lees
- Faculty of Kinesiology and Physical Education, University of Toronto, Canada
| | - Nathan Hodson
- Faculty of Kinesiology and Physical Education, University of Toronto, Canada
- Institute of Sport, Manchester Metropolitan University, Manchester, United Kingdom
- Department of Sport and Exercise Sciences, Manchester Metropolitan University, Manchester, United Kingdom
- Corresponding author. Faculty of Kinesiology and Physical Education, University of Toronto, Canada.
| |
Collapse
|
8
|
LIM CHANGHYUN, NUNES EVERSONA, CURRIER BRADS, MCLEOD JONATHANC, THOMAS AARONCQ, PHILLIPS STUARTM. An Evidence-Based Narrative Review of Mechanisms of Resistance Exercise-Induced Human Skeletal Muscle Hypertrophy. Med Sci Sports Exerc 2022; 54:1546-1559. [PMID: 35389932 PMCID: PMC9390238 DOI: 10.1249/mss.0000000000002929] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Skeletal muscle plays a critical role in physical function and metabolic health. Muscle is a highly adaptable tissue that responds to resistance exercise (RE; loading) by hypertrophying, or during muscle disuse, RE mitigates muscle loss. Resistance exercise training (RET)-induced skeletal muscle hypertrophy is a product of external (e.g., RE programming, diet, some supplements) and internal variables (e.g., mechanotransduction, ribosomes, gene expression, satellite cells activity). RE is undeniably the most potent nonpharmacological external variable to stimulate the activation/suppression of internal variables linked to muscular hypertrophy or countering disuse-induced muscle loss. Here, we posit that despite considerable research on the impact of external variables on RET and hypertrophy, internal variables (i.e., inherent skeletal muscle biology) are dominant in regulating the extent of hypertrophy in response to external stimuli. Thus, identifying the key internal skeletal muscle-derived variables that mediate the translation of external RE variables will be pivotal to determining the most effective strategies for skeletal muscle hypertrophy in healthy persons. Such work will aid in enhancing function in clinical populations, slowing functional decline, and promoting physical mobility. We provide up-to-date, evidence-based perspectives of the mechanisms regulating RET-induced skeletal muscle hypertrophy.
Collapse
Affiliation(s)
- CHANGHYUN LIM
- Department of Kinesiology, McMaster University, Hamilton, Ontario, CANADA
| | - EVERSON A. NUNES
- Department of Kinesiology, McMaster University, Hamilton, Ontario, CANADA
- Department of Physiological Science, Federal University of Santa Catarina, Florianópolis, Santa-Catarina, BRAZIL
| | - BRAD S. CURRIER
- Department of Kinesiology, McMaster University, Hamilton, Ontario, CANADA
| | - JONATHAN C. MCLEOD
- Department of Kinesiology, McMaster University, Hamilton, Ontario, CANADA
| | - AARON C. Q. THOMAS
- Department of Kinesiology, McMaster University, Hamilton, Ontario, CANADA
| | - STUART M. PHILLIPS
- Department of Kinesiology, McMaster University, Hamilton, Ontario, CANADA
| |
Collapse
|
9
|
Abou Sawan S, Hodson N, Malowany JM, West DWD, Tinline-Goodfellow C, Brook MS, Smith K, Atherton PJ, Kumbhare D, Moore DR. Trained Integrated Postexercise Myofibrillar Protein Synthesis Rates Correlate with Hypertrophy in Young Males and Females. Med Sci Sports Exerc 2022; 54:953-964. [PMID: 35081094 DOI: 10.1249/mss.0000000000002878] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
PURPOSE Resistance training induces skeletal muscle hypertrophy via the summated effects of postexercise elevations in myofibrillar protein synthesis (MyoPS) that persist for up to 48 h, although research in females is currently lacking. MyoPS is regulated by mTOR translocation and colocalization; however, the effects of resistance training on these intracellular processes are unknown. We hypothesized that MyoPS would correlate with hypertrophy only after training in both sexes and would be associated with intracellular redistribution of mTOR. METHODS Recreationally active males and females (n = 10 each) underwent 8 wk of whole-body resistance exercise three times a week. Fasted muscle biopsies were obtained immediately before (REST) and 24 and 48 h after acute resistance exercise in the untrained (UT) and trained (T) states to determine integrated MyoPS over 48 h (D2O ingestion) and intracellular mTOR colocalization (immunofluorescence microscopy). RESULTS Training increased (P < 0.01) muscle strength (~20%-126%), muscle thickness (~8%-11%), and average fiber cross-sectional area (~15%-20%). MyoPS increased above REST in UT (P = 0.032) and T (P < 0.01), but to a greater extent in males (~23%; P = 0.023), and was positively (P < 0.01) associated with muscle thickness and fiber cross-sectional area at T only in both males and females. mTOR colocalization with the cell periphery increased (P < 0.01) in T, irrespective of sex or acute exercise. Training increased (P ≤ 0.043) total mTOR, LAMP2 (lysosomal marker), and their colocalization (P < 0.01), although their colocalization was greater in males at 24 and 48 h independent of training status (P < 0.01). CONCLUSIONS MyoPS during prolonged recovery from exercise is greater in males but related to muscle hypertrophy regardless of sex only in the trained state, which may be underpinned by altered mTOR localization.
Collapse
Affiliation(s)
- Sidney Abou Sawan
- Faculty of Kinesiology and Physical Education, University of Toronto, Toronto, CANADA
| | - Nathan Hodson
- Faculty of Kinesiology and Physical Education, University of Toronto, Toronto, CANADA
| | - Julia M Malowany
- Faculty of Kinesiology and Physical Education, University of Toronto, Toronto, CANADA
| | | | | | - Matthew S Brook
- School of Life Sciences, University of Nottingham, Nottingham, UNITED KINGDOM
| | - Kenneth Smith
- School of Medicine, University of Nottingham, Derby Medical School, Derby, UNITED KINGDOM
| | - Philip J Atherton
- School of Medicine, University of Nottingham, Derby Medical School, Derby, UNITED KINGDOM
| | | | - Daniel R Moore
- Faculty of Kinesiology and Physical Education, University of Toronto, Toronto, CANADA
| |
Collapse
|
10
|
Mangano GD, Fouani M, D’Amico D, Di Felice V, Barone R. Cancer-Related Cachexia: The Vicious Circle between Inflammatory Cytokines, Skeletal Muscle, Lipid Metabolism and the Possible Role of Physical Training. Int J Mol Sci 2022; 23:ijms23063004. [PMID: 35328423 PMCID: PMC8949960 DOI: 10.3390/ijms23063004] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 02/25/2022] [Accepted: 03/09/2022] [Indexed: 12/20/2022] Open
Abstract
Cachexia is a multifactorial and multi-organ syndrome that is a major cause of morbidity and mortality in late-stage chronic diseases. The main clinical features of cancer-related cachexia are chronic inflammation, wasting of skeletal muscle and adipose tissue, insulin resistance, anorexia, and impaired myogenesis. A multimodal treatment has been suggested to approach the multifactorial genesis of cachexia. In this context, physical exercise has been found to have a general effect on maintaining homeostasis in a healthy life, involving multiple organs and their metabolism. The purpose of this review is to present the evidence for the relationship between inflammatory cytokines, skeletal muscle, and fat metabolism and the potential role of exercise training in breaking the vicious circle of this impaired tissue cross-talk. Due to the wide-ranging effects of exercise training, from the body to the behavior and cognition of the individual, it seems to be able to improve the quality of life in this syndrome. Therefore, studying the molecular effects of physical exercise could provide important information about the interactions between organs and the systemic mediators involved in the overall homeostasis of the body.
Collapse
Affiliation(s)
- Giuseppe Donato Mangano
- Correspondence: (G.D.M.); (R.B.); Tel.: +39-09-1238-65823 (G.D.M.); +39-09-1238-65823 (R.B.)
| | | | | | | | - Rosario Barone
- Correspondence: (G.D.M.); (R.B.); Tel.: +39-09-1238-65823 (G.D.M.); +39-09-1238-65823 (R.B.)
| |
Collapse
|
11
|
Mavropalias G, Wu YF, Boppart MD, Blazevich AJ, Nosaka K. Increases in Integrin-ILK-RICTOR-Akt Proteins, Muscle Mass, and Strength after Eccentric Cycling Training. Med Sci Sports Exerc 2022; 54:89-97. [PMID: 34468415 PMCID: PMC8921492 DOI: 10.1249/mss.0000000000002778] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
PURPOSE Recently, it has been suggested that a cellular pathway composed of integrin, integrin-linked kinase (ILK), rapamycin-insensitive companion of mTOR (RICTOR), and Akt may facilitate long-term structural and functional adaptations associated with exercise, independent of the mTORC1 pathway. Therefore, we examined changes in integrin-ILK-RICTOR-Akt protein in vastus lateralis (VL) before and after 8 wk of eccentric cycling training (ECC), which was expected to increase muscle function and VL cross-sectional area (CSA). METHODS Eleven men (23 ± 4 yr) completed 24 sessions of ECC with progressive increases in intensity and duration, resulting in a twofold increase in work from the first three (75.4 ± 14.1 kJ) to the last three sessions (150.7 ± 28.4 kJ). Outcome measures included lower limb lean mass, VL CSA, static strength, and peak and average cycling power output. These measures and VL samples were taken before and 4-5 d after the last training session. RESULTS Significant (P < 0.05) increases in integrin-β1 (1.64-fold) and RICTOR (2.99-fold) protein as well as the phosphorylated-to-total ILK ratio (1.70-fold) were found, but integrin-α7 and Akt did not change. Increases in lower limb, thigh, and trunk lean mass (2.8%-5.3%, P < 0.05) and CSA (13.3% ± 9.0%, P < 0.001) were observed. Static strength (18.1% ± 10.8%) and both peak (8.6% ± 10.5%) and average power output (7.4% ± 8.3%) also increased (P < 0.05). However, no significant correlations were found between the magnitude of increases in protein and the magnitude of increases in CSA, static strength, or power output. CONCLUSIONS In addition to increased muscle mass, strength, and power, we demonstrate that ECC increases integrin-β1 and RICTOR total protein and p-ILK/t-ILK, which may play a role in protection against muscle damage as well as anabolic signaling to induce muscle adaptations.
Collapse
Affiliation(s)
- Georgios Mavropalias
- Exercise Medicine Research Institute, Edith Cowan University, Australia
- School of Medical and Health Sciences, Edith Cowan University, Australia
| | - Yu-Fu Wu
- Department of Kinesiology & Community Health, University of Illinois at Urbana-Champaign, IL
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, IL
| | - Marni D. Boppart
- Department of Kinesiology & Community Health, University of Illinois at Urbana-Champaign, IL
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, IL
| | | | - Kazunori Nosaka
- Exercise Medicine Research Institute, Edith Cowan University, Australia
- School of Medical and Health Sciences, Edith Cowan University, Australia
| |
Collapse
|
12
|
Hodson N, Mazzulla M, Holowaty MNH, Kumbhare D, Moore DR. RPS6 phosphorylation occurs to a greater extent in the periphery of human skeletal muscle fibers, near focal adhesions, after anabolic stimuli. Am J Physiol Cell Physiol 2021; 322:C94-C110. [PMID: 34852208 DOI: 10.1152/ajpcell.00357.2021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Following anabolic stimuli (mechanical loading and/or amino acid provision) the mechanistic target of rapamycin complex 1 (mTORC1), a master regulator of protein synthesis, translocates toward the cell periphery. However, it is unknown if mTORC1-mediated phosphorylation events occur in these peripheral regions or prior to translocation (i.e. in central regions). We therefore aimed to determine the cellular location of a mTORC1-mediated phosphorylation event, RPS6Ser240/244, in human skeletal muscle following anabolic stimuli. Fourteen young, healthy males either ingested a protein-carbohydrate beverage (0.25g/kg protein, 0.75g/kg carbohydrate) alone (n=7;23±5yrs;76.8±3.6kg;13.6±3.8%BF, FED) or following a whole-body resistance exercise bout (n=7;22±2yrs;78.1±3.6kg;12.2±4.9%BF, EXFED). Vastus lateralis muscle biopsies were obtained at rest (PRE) and 120 and 300min following anabolic stimuli. RPS6Ser240/244 phosphorylation measured by immunofluorescent staining or immunoblot was positively correlated (r=0.76, p<0.001). Peripheral staining intensity of p-RPS6Ser240/244 increased above PRE in both FED and EXFED at 120min (~54% and ~138% respectively, p<0.05) but was greater in EXFED at both post-stimuli time points (p<0.05). The peripheral-central ratio of p-RPS6240/244 staining displayed a similar pattern, even when corrected for total RPS6 distribution, suggesting RPS6 phosphorylation occurs to a greater extent in the periphery of fibers. Moreover, p-RPS6Ser240/244 intensity within paxillin-positive regions, a marker of focal adhesion complexes, was elevated at 120min irrespective of stimulus (p=0.006) before returning to PRE at 300min. These data confirm that RPS6Ser240/244 phosphorylation occurs in the region of human muscle fibers to which mTOR translocates following anabolic stimuli and identifies focal adhesion complexes as a potential site of mTORC1 regulation in vivo.
Collapse
Affiliation(s)
- Nathan Hodson
- Faculty of Kinesiology and Physical Education, University of Toronto, Toronto, ON, Canada
| | - Michael Mazzulla
- Faculty of Kinesiology and Physical Education, University of Toronto, Toronto, ON, Canada
| | - Maksym N H Holowaty
- Faculty of Kinesiology and Physical Education, University of Toronto, Toronto, ON, Canada
| | | | - Daniel R Moore
- Faculty of Kinesiology and Physical Education, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
13
|
Abou Sawan S, Hodson N, Tinline-Goodfellow C, West DWD, Malowany JM, Kumbhare D, Moore DR. Incorporation of Dietary Amino Acids Into Myofibrillar and Sarcoplasmic Proteins in Free-Living Adults Is Influenced by Sex, Resistance Exercise, and Training Status. J Nutr 2021; 151:3350-3360. [PMID: 34486662 DOI: 10.1093/jn/nxab261] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 06/25/2021] [Accepted: 07/15/2021] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Acute exercise increases the incorporation of dietary amino acids into de novo myofibrillar proteins after a single meal in controlled laboratory studies in males. It is unclear whether this extends to free-living settings or is influenced by training or sex. OBJECTIVES We determined the effects of exercise, training status, and sex on 24-hour free-living dietary phenylalanine incorporation into skeletal muscle proteins. METHODS In a parallel group design, recreationally active males (mean ± SD age, 23 ± 3 years; BMI. 23.4 ± 2.9 kg/m2; n = 10) and females (age 24 ± 5 years; BMI, 23.1 ± 3.9 kg/m2; n = 9) underwent 8 weeks of whole-body resistance exercise 3 times a week. Controlled diets containing 1.6 g/kg-1/d-1 (amino acids modelled after egg), enriched to 10% with [13C6] or [2H5]phenylalanine, were consumed before and after an acute bout of resistance exercise. Fasted muscle biopsies were obtained before [untrained, pre-exercise condition (REST ] and 24 hours after an acute bout of resistance exercise in untrained (UT) and trained (T) states to determine dietary phenylalanine incorporation into myofibrillar (ΔMyo) and sarcoplasmic (ΔSarc) proteins, intracellular mechanistic target of rapamycin (mTOR) colocalization with ulex europaeus agglutinin-1 (UEA-1; capillary marker; immunofluorescence), and amino acid transporter expression (Western blotting). RESULTS The ΔMyo values were ∼62% greater (P < 0.01) in females than males at REST. The ΔMyo values increased above REST by ∼51% during UT and ∼30% in T (both P < 0.01) in males, remained unchanged in females during UT, and were ∼33% lower at T when compared to UT (P = 0.013). Irrespective of sex, ΔMyo and ΔSarc were decreased at T compared to UT (P ≤ 0.026). Resistance training increased mTOR colocalization with UEA-1 (P = 0.004), while L amino acid transporter 1, which was greater in males (P < 0.01), and sodium-coupled neutral amino acid transporter 2 protein expression were not affected by acute exercise (P ≥ 0.33) or training (P ≥ 0.45). CONCLUSIONS The exercise-induced incorporation of dietary phenylalanine into myofibrillar and sarcoplasmic proteins is attenuated after training regardless of sex, suggesting a reduced reliance on dietary amino acids for postexercise skeletal muscle remodeling in the T state.
Collapse
Affiliation(s)
- Sidney Abou Sawan
- Faculty of Kinesiology and Physical Education, University of Toronto, Toronto, Ontario, Canada
| | - Nathan Hodson
- Faculty of Kinesiology and Physical Education, University of Toronto, Toronto, Ontario, Canada
| | | | - Daniel W D West
- Faculty of Kinesiology and Physical Education, University of Toronto, Toronto, Ontario, Canada.,Toronto Rehabilitation Institute, Toronto, Canada
| | - Julia M Malowany
- Faculty of Kinesiology and Physical Education, University of Toronto, Toronto, Ontario, Canada
| | | | - Daniel R Moore
- Faculty of Kinesiology and Physical Education, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
14
|
Mesquita PHC, Vann CG, Phillips SM, McKendry J, Young KC, Kavazis AN, Roberts MD. Skeletal Muscle Ribosome and Mitochondrial Biogenesis in Response to Different Exercise Training Modalities. Front Physiol 2021; 12:725866. [PMID: 34646153 PMCID: PMC8504538 DOI: 10.3389/fphys.2021.725866] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Accepted: 08/13/2021] [Indexed: 11/20/2022] Open
Abstract
Skeletal muscle adaptations to resistance and endurance training include increased ribosome and mitochondrial biogenesis, respectively. Such adaptations are believed to contribute to the notable increases in hypertrophy and aerobic capacity observed with each exercise mode. Data from multiple studies suggest the existence of a competition between ribosome and mitochondrial biogenesis, in which the first adaptation is prioritized with resistance training while the latter is prioritized with endurance training. In addition, reports have shown an interference effect when both exercise modes are performed concurrently. This prioritization/interference may be due to the interplay between the 5’ AMP-activated protein kinase (AMPK) and mechanistic target of rapamycin complex 1 (mTORC1) signaling cascades and/or the high skeletal muscle energy requirements for the synthesis and maintenance of cellular organelles. Negative associations between ribosomal DNA and mitochondrial DNA copy number in human blood cells also provide evidence of potential competition in skeletal muscle. However, several lines of evidence suggest that ribosome and mitochondrial biogenesis can occur simultaneously in response to different types of exercise and that the AMPK-mTORC1 interaction is more complex than initially thought. The purpose of this review is to provide in-depth discussions of these topics. We discuss whether a curious competition between mitochondrial and ribosome biogenesis exists and show the available evidence both in favor and against it. Finally, we provide future research avenues in this area of exercise physiology.
Collapse
Affiliation(s)
| | | | - Stuart M Phillips
- Department of Kinesiology, McMaster University, Hamilton, ON, Canada
| | - James McKendry
- Department of Kinesiology, McMaster University, Hamilton, ON, Canada
| | - Kaelin C Young
- School of Kinesiology, Auburn University, Auburn, AL, United States.,Department of Cell Biology and Physiology, Edward Via College of Osteopathic Medicine, Auburn, AL, United States
| | | | - Michael D Roberts
- School of Kinesiology, Auburn University, Auburn, AL, United States.,Department of Cell Biology and Physiology, Edward Via College of Osteopathic Medicine, Auburn, AL, United States
| |
Collapse
|
15
|
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: 2.7] [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
|
16
|
The effects of glucagon and the target of rapamycin (TOR) on skeletal muscle protein synthesis and age-dependent sarcopenia in humans. Clin Nutr ESPEN 2021; 44:15-25. [PMID: 34330459 DOI: 10.1016/j.clnesp.2021.06.025] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Accepted: 06/21/2021] [Indexed: 01/07/2023]
Abstract
BACKGROUND AND AIMS Human target of rapamycin (TOR) is a kinase that stimulates protein synthesis in the skeletal muscle in response to amino acids and physical activity. METHODS A comprehensive literature search was conducted on the PubMed database from its inception up to May 2021 to retrieve information on the effects of TOR and glucagon on muscle function. Articles written in English regarding human subjects were included. RESULTS l-leucine activates TOR to initiate protein synthesis in the skeletal muscle. Glucagon has a crucial role suppressing skeletal muscle protein synthesis by increasing l-leucine oxidation and the irreversible loss of this amino acid. Glucagon-induced l-leucine oxidation suppresses TOR and attenuates the ability of skeletal muscle to synthesize proteins. Conditions associated with increased glucagon secretion typically feature reduced ability to synthesize proteins in the skeletal muscle that may evolve into sarcopenia. Animal protein ingestion, unlike vegetable protein, stimulates glucagon secretion. High intake of animal protein increases l-leucine oxidation and promotes the use of amino acids as fuel. Sarcopenia and arterial stiffness characteristically occur together in conditions featuring insulin resistance, such as aging. Insulin resistance mediates the relationship between aging and sarcopenia and arterial stiffness. The loss of skeletal muscle fibers that characterizes sarcopenia is followed by collagen and lipid accumulation. Likewise, insulin resistance is associated with arterial stiffness and intima-media thickening due to adaptive accretion of collagen and lipids in the arterial wall. CONCLUSIONS Human TOR participates in the pathogenesis of sarcopenia and arterial stiffness, although its effects remain to be fully elucidated.
Collapse
|
17
|
Zakaria NF, Hamid M, Khayat ME. Amino Acid-Induced Impairment of Insulin Signaling and Involvement of G-Protein Coupling Receptor. Nutrients 2021; 13:nu13072229. [PMID: 34209599 PMCID: PMC8308393 DOI: 10.3390/nu13072229] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 06/18/2021] [Accepted: 06/24/2021] [Indexed: 12/12/2022] Open
Abstract
Amino acids are needed for general bodily function and well-being. Despite their importance, augmentation in their serum concentration is closely related to metabolic disorder, insulin resistance (IR), or worse, diabetes mellitus. Essential amino acids such as the branched-chain amino acids (BCAAs) have been heavily studied as a plausible biomarker or even a cause of IR. Although there is a long list of benefits, in subjects with abnormal amino acids profiles, some amino acids are correlated with a higher risk of IR. Metabolic dysfunction, upregulation of the mammalian target of the rapamycin (mTOR) pathway, the gut microbiome, 3-hydroxyisobutyrate, inflammation, and the collusion of G-protein coupled receptors (GPCRs) are among the indicators and causes of metabolic disorders generating from amino acids that contribute to IR and the onset of type 2 diabetes mellitus (T2DM). This review summarizes the current understanding of the true involvement of amino acids with IR. Additionally, the involvement of GPCRs in IR will be further discussed in this review.
Collapse
Affiliation(s)
- Nur Fatini Zakaria
- Department of Biochemistry, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia;
| | - Muhajir Hamid
- Department of Microbiology, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia;
| | - Mohd Ezuan Khayat
- Department of Biochemistry, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia;
- Correspondence:
| |
Collapse
|
18
|
Borack MS, Dickinson JM, Fry CS, Reidy PT, Markofski MM, Deer RR, Jennings K, Volpi E, Rasmussen BB. Effect of the lysosomotropic agent chloroquine on mTORC1 activation and protein synthesis in human skeletal muscle. Nutr Metab (Lond) 2021; 18:61. [PMID: 34118944 PMCID: PMC8199655 DOI: 10.1186/s12986-021-00585-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Accepted: 05/28/2021] [Indexed: 11/16/2022] Open
Abstract
Background Previous work in HEK-293 cells demonstrated the importance of amino acid-induced mTORC1 translocation to the lysosomal surface for stimulating mTORC1 kinase activity and protein synthesis. This study tested the conservation of this amino acid sensing mechanism in human skeletal muscle by treating subjects with chloroquine—a lysosomotropic agent that induces in vitro and in vivo lysosome dysfunction. Methods mTORC1 signaling and muscle protein synthesis (MPS) were determined in vivo in a randomized controlled trial of 14 subjects (10 M, 4 F; 26 ± 4 year) that ingested 10 g of essential amino acids (EAA) after receiving 750 mg of chloroquine (CHQ, n = 7) or serving as controls (CON, n = 7; no chloroquine). Additionally, differentiated C2C12 cells were used to assess mTORC1 signaling and myotube protein synthesis (MyPS) in the presence and absence of leucine and the lysosomotropic agent chloroquine. Results mTORC1, S6K1, 4E-BP1 and rpS6 phosphorylation increased in both CON and CHQ 1 h post EAA ingestion (P < 0.05). MPS increased similarly in both groups (CON, P = 0.06; CHQ, P < 0.05). In contrast, in C2C12 cells, 1 mM leucine increased mTORC1 and S6K1 phosphorylation (P < 0.05), which was inhibited by 2 mg/ml chloroquine. Chloroquine (2 mg/ml) was sufficient to disrupt mTORC1 signaling, and MyPS. Conclusions Chloroquine did not inhibit amino acid-induced activation of mTORC1 signaling and skeletal MPS in humans as it does in C2C12 muscle cells. Therefore, different in vivo experimental approaches are required for confirming the precise role of the lysosome and amino acid sensing in human skeletal muscle. Trial registration NCT00891696. Registered 29 April 2009.
Collapse
Affiliation(s)
- Michael S Borack
- Division of Rehabilitation Sciences, University of Texas Medical Branch, 301 University Blvd, Galveston, TX, 77555-1124, USA.,Department of Nutrition and Metabolism, University of Texas Medical Branch, 301 University Blvd, Galveston, TX, 77555-1124, USA.,Center for the Study of Aging and Human Development, Duke University, Durham, NC, USA
| | - Jared M Dickinson
- Department of Nutrition and Metabolism, University of Texas Medical Branch, 301 University Blvd, Galveston, TX, 77555-1124, USA.,Sealy Center On Aging, University of Texas Medical Branch, 301 University Blvd, Galveston, TX, 77555-1124, USA.,Department of Health Sciences, Central Washington University, Ellensburg, WA, USA
| | - Christopher S Fry
- Department of Nutrition and Metabolism, University of Texas Medical Branch, 301 University Blvd, Galveston, TX, 77555-1124, USA.,Sealy Center On Aging, University of Texas Medical Branch, 301 University Blvd, Galveston, TX, 77555-1124, USA.,Department of Athletic Training and Clinical Nutrition, University of Kentucky, Lexington, KY, USA
| | - Paul T Reidy
- Division of Rehabilitation Sciences, University of Texas Medical Branch, 301 University Blvd, Galveston, TX, 77555-1124, USA.,Department of Nutrition and Metabolism, University of Texas Medical Branch, 301 University Blvd, Galveston, TX, 77555-1124, USA.,Department of Kinesiology, Nutrition and Health, Miami of Ohio University, Oxford, OH, USA
| | - Melissa M Markofski
- Sealy Center On Aging, University of Texas Medical Branch, 301 University Blvd, Galveston, TX, 77555-1124, USA.,Department of Health and Human Performance, University of Houston, Houston, TX, USA
| | - Rachel R Deer
- Division of Rehabilitation Sciences, University of Texas Medical Branch, 301 University Blvd, Galveston, TX, 77555-1124, USA.,Sealy Center On Aging, University of Texas Medical Branch, 301 University Blvd, Galveston, TX, 77555-1124, USA
| | - Kristofer Jennings
- Department of Preventive Medicine and Population Health, University of Texas Medical Branch, 301 University Blvd, Galveston, TX, 77555-1124, USA.,Department of Biostatistics, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Elena Volpi
- Department of Internal Medicine/Geriatrics, University of Texas Medical Branch, 301 University Blvd, Galveston, TX, 77555-1124, USA.,Sealy Center On Aging, University of Texas Medical Branch, 301 University Blvd, Galveston, TX, 77555-1124, USA
| | - Blake B Rasmussen
- Department of Nutrition and Metabolism, University of Texas Medical Branch, 301 University Blvd, Galveston, TX, 77555-1124, USA. .,Sealy Center On Aging, University of Texas Medical Branch, 301 University Blvd, Galveston, TX, 77555-1124, USA.
| |
Collapse
|
19
|
Zhao Y, Cholewa J, Shang H, Yang Y, Ding X, Wang Q, Su Q, Zanchi NE, Xia Z. Advances in the Role of Leucine-Sensing in the Regulation of Protein Synthesis in Aging Skeletal Muscle. Front Cell Dev Biol 2021; 9:646482. [PMID: 33869199 PMCID: PMC8047301 DOI: 10.3389/fcell.2021.646482] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2020] [Accepted: 03/04/2021] [Indexed: 12/21/2022] Open
Abstract
Skeletal muscle anabolic resistance (i.e., the decrease in muscle protein synthesis (MPS) in response to anabolic stimuli such as amino acids and exercise) has been identified as a major cause of age-related sarcopenia, to which blunted nutrition-sensing contributes. In recent years, it has been suggested that a leucine sensor may function as a rate-limiting factor in skeletal MPS via small-molecule GTPase. Leucine-sensing and response may therefore have important therapeutic potential in the steady regulation of protein metabolism in aging skeletal muscle. This paper systematically summarizes the three critical processes involved in the leucine-sensing and response process: (1) How the coincidence detector mammalian target of rapamycin complex 1 localizes on the surface of lysosome and how its crucial upstream regulators Rheb and RagB/RagD interact to modulate the leucine response; (2) how complexes such as Ragulator, GATOR, FLCN, and TSC control the nucleotide loading state of Rheb and RagB/RagD to modulate their functional activity; and (3) how the identified leucine sensor leucyl-tRNA synthetase (LARS) and stress response protein 2 (Sestrin2) participate in the leucine-sensing process and the activation of RagB/RagD. Finally, we discuss the potential mechanistic role of exercise and its interactions with leucine-sensing and anabolic responses.
Collapse
Affiliation(s)
- Yan Zhao
- Exercise Physiology and Biochemistry Laboratory, College of Physical Education, Jinggangshan University, Ji'an, China
| | - Jason Cholewa
- Department of Exercise Physiology, University of Lynchburg, Lynchburg, VA, United States
| | - Huayu Shang
- School of Sport Medicine and Health, Chengdu Sport University, Chengdu, China
| | - Yueqin Yang
- Hubei Provincial Collaborative Innovation Center for Exercise and Health Promotion, College of Health Science, Wuhan Sports University, Wuhan, China
| | - Xiaomin Ding
- Exercise Physiology and Biochemistry Laboratory, College of Physical Education, Jinggangshan University, Ji'an, China
| | - Qianjin Wang
- Exercise Physiology and Biochemistry Laboratory, College of Physical Education, Jinggangshan University, Ji'an, China
| | - Quansheng Su
- School of Sport Medicine and Health, Chengdu Sport University, Chengdu, China
| | - Nelo Eidy Zanchi
- Department of Physical Education, Federal University of Maranhão (UFMA), São Luís-MA, Brazil.,Laboratory of Cellular and Molecular Biology of Skeletal Muscle (LABCEMME), São Luís-MA, Brazil
| | - Zhi Xia
- Exercise Physiology and Biochemistry Laboratory, College of Physical Education, Jinggangshan University, Ji'an, China.,School of Sport Medicine and Health, Chengdu Sport University, Chengdu, China
| |
Collapse
|
20
|
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: 3.7] [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
|
21
|
Abstract
Sensing and responding to changes in nutrient levels, including those of glucose, lipids, and amino acids, by the body is necessary for survival. Accordingly, perturbations in nutrient sensing are tightly linked with human pathologies, particularly metabolic diseases such as obesity, type 2 diabetes mellitus, and other complications of metabolic syndromes. The conventional view is that amino acids are fundamental elements for protein and peptide synthesis, while recent studies have revealed that amino acids are also important bioactive molecules that play key roles in signaling pathways and metabolic regulation. Different pathways that sense intracellular and extracellular levels of amino acids are integrated and coordinated at the organismal level, and, together, these pathways maintain whole metabolic homeostasis. In this review, we discuss the studies describing how important sensing signals respond to amino acid availability and how these sensing mechanisms modulate metabolic processes, including energy, glucose, and lipid metabolism. We further discuss whether dysregulation of amino acid sensing signals can be targeted to promote metabolic disorders, and discuss how to translate these mechanisms to treat human diseases. This review will help to enhance our overall understanding of the correlation between amino acid sensing and metabolic homeostasis, which have important implications for human health.
Collapse
Affiliation(s)
- Xiaoming Hu
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Feifan Guo
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| |
Collapse
|
22
|
Deligiannis A, D'Alessandro C, Cupisti A. Exercise training in dialysis patients: impact on cardiovascular and skeletal muscle health. Clin Kidney J 2021; 14:ii25-ii33. [PMID: 33981417 PMCID: PMC8101623 DOI: 10.1093/ckj/sfaa273] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2020] [Accepted: 12/09/2020] [Indexed: 12/13/2022] Open
Abstract
Dialysis patients show a high rate of reduced functional capacity, morbidity and mortality. Cardiovascular disorders, muscle atrophy and malnutrition play an essential role among the aetiological factors. Sedentary lifestyle characterizes them and contributes to the aggravation of the disorders. On the contrary, exercise training is an important preventive and therapeutic tool both for cardiovascular problems and for the appearance of muscle atrophy in dialysis patients. Regular exercise causes both central (cardiac) and peripheral (muscular) adaptations, improving functional capacity. In particular, circulatory system clinical trials in haemodialysis (HD) patients documented that exercise has favourable effects on heart function, promotes balance on the cardiac autonomic nervous system and contributes to the management of arterial hypertension. In the muscular system, it prevents muscle atrophy or contributes significantly to its treatment. The main preventive mechanisms of the beneficial effect of exercise on the muscles constitute the inhibition of the apoptotic processes and protein degradation. Exercise training in HD patients leads to an increase of muscle fibers, mitochondria and capillaries, and the combination of regular exercise and dietary strategies is even more effective in preventing or treating muscle atrophy. Finally, an improvement in functional capacity and quality of life was found also in peritoneal dialysis patients following exercise training.
Collapse
Affiliation(s)
- Asterios Deligiannis
- Sports Medicine Laboratory, School of Physical Education and Sports Science, Aristotle University of Thessaloniki, Thermi, Greece
| | - Claudia D'Alessandro
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Adamasco Cupisti
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| |
Collapse
|
23
|
Morgan PT, Smeuninx B, Breen L. Exploring the Impact of Obesity on Skeletal Muscle Function in Older Age. Front Nutr 2020; 7:569904. [PMID: 33335909 PMCID: PMC7736105 DOI: 10.3389/fnut.2020.569904] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Accepted: 11/06/2020] [Indexed: 12/16/2022] Open
Abstract
Sarcopenia is of important clinical relevance for loss of independence in older adults. The prevalence of obesity in combination with sarcopenia ("sarcopenic-obesity") is increasing at a rapid rate. However, whilst the development of sarcopenia is understood to be multi-factorial and harmful to health, the role of obesity from a protective and damaging perspective on skeletal muscle in aging, is poorly understood. Specifically, the presence of obesity in older age may be accompanied by a greater volume of skeletal muscle mass in weight-bearing muscles compared with lean older individuals, despite impaired physical function and resistance to anabolic stimuli. Collectively, these findings support a potential paradox in which obesity may protect skeletal muscle mass in older age. One explanation for these paradoxical findings may be that the anabolic response to weight-bearing activity could be greater in obese vs. lean older individuals due to a larger mechanical stimulus, compensating for the heightened muscle anabolic resistance. However, it is likely that there is a complex interplay between muscle, adipose, and external influences in the aging process that are ultimately harmful to health in the long-term. This narrative briefly explores some of the potential mechanisms regulating changes in skeletal muscle mass and function in aging combined with obesity and the interplay with sarcopenia, with a particular focus on muscle morphology and the regulation of muscle proteostasis. In addition, whilst highly complex, we attempt to provide an updated summary for the role of obesity from a protective and damaging perspective on muscle mass and function in older age. We conclude with a brief discussion on treatment of sarcopenia and obesity and a summary of future directions for this research field.
Collapse
Affiliation(s)
- Paul T. Morgan
- School of Sport, Exercise and Rehabilitation Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Benoit Smeuninx
- School of Sport, Exercise and Rehabilitation Sciences, University of Birmingham, Birmingham, United Kingdom
- Cellular & Molecular Metabolism Laboratory, Monash Institute of Pharmacological Sciences, Monash University, Parkville, VIC, Australia
| | - Leigh Breen
- School of Sport, Exercise and Rehabilitation Sciences, University of Birmingham, Birmingham, United Kingdom
| |
Collapse
|
24
|
Hodson N, Dent JR, Song Z, O'Leary MF, Nicholson T, Jones SW, Murray JT, Jeromson S, Hamilton DL, Breen L, Philp A. Protein-carbohydrate ingestion alters Vps34 cellular localization independent of changes in kinase activity in human skeletal muscle. Exp Physiol 2020; 105:2178-2189. [PMID: 32965751 DOI: 10.1113/ep088805] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 09/07/2020] [Accepted: 09/18/2020] [Indexed: 02/02/2023]
Abstract
NEW FINDINGS What is the central question of the study? Is Vps34 a nutrient-sensitive activator of mTORC1 in human skeletal muscle? What is the main finding and its importance? We show that altering nutrient availability, via protein-carbohydrate feeding, does not increase Vps34 kinase activity in human skeletal muscle. Instead, feeding increased Vps34-mTORC1 co-localization in parallel to increased mTORC1 activity. These findings may have important implications in the understanding nutrient-induced mTORC1 activation in skeletal muscle via interaction with Vps34. ABSTRACT The Class III PI3Kinase, Vps34, has recently been proposed as a nutrient sensor, essential for activation of the mechanistic target of rapamycin (mTOR) complex 1 (mTORC1). We therefore investigated the effects of increasing nutrient availability through protein-carbohydrate (PRO-CHO) feeding on Vps34 kinase activity and cellular localization in human skeletal muscle. Eight young, healthy males (21 ± 0.5 yrs, 77.7 ± 9.9 kg, 25.9 ± 2.7 kg/m2 , mean ± SD) ingested a PRO-CHO beverage containing 20/44/1 g PRO/CHO/FAT respectively, with skeletal muscle biopsies obtained at baseline and 1 h and 3 h post-feeding. PRO-CHO feeding did not alter Vps34 kinase activity, but did stimulate Vps34 translocation toward the cell periphery (PRE (mean ± SD) - 0.273 ± 0.040, 1 h - 0.348 ± 0.061, Pearson's Coefficient (r)) where it co-localized with mTOR (PRE - 0.312 ± 0.040, 1 h - 0.348 ± 0.069, Pearson's Coefficient (r)). These alterations occurred in parallel to an increase in S6K1 kinase activity (941 ± 466% of PRE at 1 h post-feeding). Subsequent in vitro experiments in C2C12 and human primary myotubes displayed no effect of the Vps34-specific inhibitor SAR405 on mTORC1 signalling responses to elevated nutrient availability. Therefore, in summary, PRO-CHO ingestion does not increase Vps34 activity in human skeletal muscle, whilst pharmacological inhibition of Vps34 does not prevent nutrient stimulation of mTORC1 in vitro. However, PRO-CHO ingestion promotes Vps34 translocation to the cell periphery, enabling Vps34 to associate with mTOR. Therefore, our data suggests that interaction between Vps34 and mTOR, rather than changes in Vps34 activity per se may be involved in PRO-CHO activation of mTORC1 in human skeletal muscle.
Collapse
Affiliation(s)
- Nathan Hodson
- School of Sport, Exercise and Rehabilitation Sciences, University of Birmingham, Birmingham, UK
| | - Jessica R Dent
- School of Sport, Exercise and Rehabilitation Sciences, University of Birmingham, Birmingham, UK
| | - Zhe Song
- School of Sport, Exercise and Rehabilitation Sciences, University of Birmingham, Birmingham, UK
| | - Mary F O'Leary
- School of Sport, Exercise and Rehabilitation Sciences, University of Birmingham, Birmingham, UK
| | - Thomas Nicholson
- Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK
| | - Simon W Jones
- Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK
| | - James T Murray
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College, Dublin, Ireland.,Swansea University Medical School, Swansea University, UK
| | - Stewart Jeromson
- Physiology, Exercise and Nutrition Research Group, School of Sport, Stirling University, Stirling, UK
| | - D Lee Hamilton
- Institute for Physical Activity and Nutrition (IPAN), School of Exercise & Nutrition Sciences, Deakin University, Geelong, 3216, Australia
| | - Leigh Breen
- 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.,Healthy Ageing Research Theme, Garvan Institute of Medical Research, Sydney, NSW, Australia.,St Vincent's Clinical School, UNSW Medicine, UNSW Sydney, Sydney, NSW, Australia
| |
Collapse
|
25
|
Zhu Y, Zhou J, Niu P, Chen H, Shi D. Cardamonin inhibits cell proliferation by caspase-mediated cleavage of Raptor. Naunyn Schmiedebergs Arch Pharmacol 2020; 394:809-817. [PMID: 33043385 DOI: 10.1007/s00210-020-01986-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Accepted: 10/04/2020] [Indexed: 12/16/2022]
Abstract
The antiproliferative effect of cardamonin on mTORC1 is related with downregulation of Raptor. We investigated the mechanism that cardamonin decreases Raptor expression through caspase-mediated protein degradation. SKOV3 cells and HeLa cells were pretreated with caspase inhibitor z-VAD-fmk for 30 min and then exposed to different doses of cardamonin and cisplatin, respectively. We analyzed the gene expression of caspases based on TCGA and GTEx gene expression data in serous cystadenocarcinoma and normal tissues, monitored caspase activity by caspase colorimetric assay kit, detected expression of mTORC1-associated proteins and apoptosis-associated proteins by western blotting, and finally detected cell viability by methyl thiazolyl tetrazolium (MTT) assay. A different expression of caspases except caspase-1 was found between serous cystadenocarcinoma and normal tissues. Raptor was cleaved when caspases were activated by cisplatin and caspase-6/caspase-8 was activated by cardamonin in SKOV3 cells. We further used a monoclonal antibody recognizing the N-terminal part of Raptor to find that Raptor was cleaved into a smaller fragment of about 70 kDa by cardamonin and was rescued by z-VAD-fmk treatment. As a result of Raptor cleavage, mTORC1 activity was decreased and cell viability was inhibited, while cell apoptosis was induced in SKOV3 cells. Notably, similar results are only observed in HeLa cells with a high dose of cardamonin. We concluded that caspase-mediated cleavage of Raptor might be an important mechanism in that cardamonin regulated Raptor and mTORC1 activity.
Collapse
Affiliation(s)
- Yanting Zhu
- Department of Pharmacy, Fujian Maternity and Child Health Hospital, Affiliated Hospital of Fujian Medical University, 18 Daoshan Road, Fuzhou, 350001, Fujian, China
| | - Jintuo Zhou
- Department of Pharmacy, Fujian Maternity and Child Health Hospital, Affiliated Hospital of Fujian Medical University, 18 Daoshan Road, Fuzhou, 350001, Fujian, China
| | - Peiguang Niu
- Department of Pharmacy, Fujian Maternity and Child Health Hospital, Affiliated Hospital of Fujian Medical University, 18 Daoshan Road, Fuzhou, 350001, Fujian, China
| | - Huajiao Chen
- Department of Pharmacy, Fujian Maternity and Child Health Hospital, Affiliated Hospital of Fujian Medical University, 18 Daoshan Road, Fuzhou, 350001, Fujian, China
| | - Daohua Shi
- Department of Pharmacy, Fujian Maternity and Child Health Hospital, Affiliated Hospital of Fujian Medical University, 18 Daoshan Road, Fuzhou, 350001, Fujian, China.
| |
Collapse
|
26
|
Counts BR, Hardee JP, Fix DK, Vanderveen BN, Montalvo RN, Carson JA. Cachexia Disrupts Diurnal Regulation of Activity, Feeding, and Muscle Mechanistic Target of Rapamycin Complex 1 in Mice. Med Sci Sports Exerc 2020; 52:577-587. [PMID: 32058469 DOI: 10.1249/mss.0000000000002166] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
INTRODUCTION Cancer cachexia is characterized by severe skeletal muscle mass loss, which is driven by decreased muscle protein synthesis and increased protein degradation. Daily physical activity and feeding behaviors exhibit diurnal fluctuations in mice that can impact the systemic environment and skeletal muscle signaling. PURPOSE We investigated the effect of cancer cachexia on the diurnal regulation of feeding, physical activity, and skeletal muscle mechanistic target of rapamycin complex 1 (mTORC1) signaling in tumor-bearing mice. We also examined the impact of increased physical activity on diurnal behaviors and skeletal muscle mTROC1 signaling in the cancer environment. METHODS Physical activity and feeding behaviors were measured for four consecutive days before sacrifice in male C57BL/6 (B6; n = 24) and Apc (MIN; n = 22) mice at 7:00 AM and 7:00 PM under ad libitum condition. A subset of B6 (n = 16) and MIN (n = 19) mice were given wheel access for 2 wk before diurnal behavior measurements. Gastrocnemius muscle protein expression was examined. RESULTS The MIN mice demonstrated altered diurnal fluctuations in feeding and activity compared with the B6. Interestingly, cachexia did not alter MIN total food intake, but dramatically reduced cage physical activity. As a measurement of mTORC1 activity, 4E-BP1 phosphorylation increased after the dark cycle in B6 and precachectic MIN mice, whereas rpS6 phosphorylation was only increased after the dark cycle in MIN mice. MIN 4E-BP1 phosphorylation at the end of the light cycle was significantly correlated with cachexia progression and reduced physical activity. Voluntary wheel running increased light cycle MIN 4E-BP1 phosphorylation and attenuated muscle mass loss. CONCLUSIONS The cancer environment can alter diurnal feeding and physical activity behaviors in tumor-bearing mice, which are linked to the progression of cachexia and muscle wasting. Furthermore, suppressed physical activity during cachexia is associated with decreased skeletal muscle mTORC1 signaling.
Collapse
Affiliation(s)
- Brittany R Counts
- Integrative Muscle Biology Laboratory, Division of Rehabilitation Sciences, College of Health Professions, University of Tennessee Health Science Center, Memphis TN
| | - Justin P Hardee
- Integrative Muscle Biology Laboratory, Department of Exercise Science, University of South Carolina, Columbia, SC
| | - Dennis K Fix
- Integrative Muscle Biology Laboratory, Department of Exercise Science, University of South Carolina, Columbia, SC
| | - Brandon N Vanderveen
- Integrative Muscle Biology Laboratory, Department of Exercise Science, University of South Carolina, Columbia, SC
| | - Ryan N Montalvo
- Integrative Muscle Biology Laboratory, Department of Exercise Science, University of South Carolina, Columbia, SC
| | - James A Carson
- Integrative Muscle Biology Laboratory, Division of Rehabilitation Sciences, College of Health Professions, University of Tennessee Health Science Center, Memphis TN
| |
Collapse
|
27
|
Monteyne AJ, Coelho MOC, Porter C, Abdelrahman DR, Jameson TSO, Jackman SR, Blackwell JR, Finnigan TJA, Stephens FB, Dirks ML, Wall BT. Mycoprotein ingestion stimulates protein synthesis rates to a greater extent than milk protein in rested and exercised skeletal muscle of healthy young men: a randomized controlled trial. Am J Clin Nutr 2020; 112:318-333. [PMID: 32438401 DOI: 10.1093/ajcn/nqaa092] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Accepted: 04/09/2020] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Mycoprotein is a fungal-derived sustainable protein-rich food source, and its ingestion results in systemic amino acid and leucine concentrations similar to that following milk protein ingestion. OBJECTIVE We assessed the mixed skeletal muscle protein synthetic response to the ingestion of a single bolus of mycoprotein compared with a leucine-matched bolus of milk protein, in rested and exercised muscle of resistance-trained young men. METHODS Twenty resistance-trained healthy young males (age: 22 ± 1 y, body mass: 82 ± 2 kg, BMI: 25 ± 1 kg·m-2) took part in a randomized, double-blind, parallel-group study. Participants received primed, continuous infusions of L-[ring-2H5]phenylalanine and ingested either 31 g (26.2 g protein: 2.5 g leucine) milk protein (MILK) or 70 g (31.5 g protein: 2.5 g leucine) mycoprotein (MYCO) following a bout of unilateral resistance-type exercise (contralateral leg acting as resting control). Blood and m. vastus lateralis muscle samples were collected before exercise and protein ingestion, and following a 4-h postprandial period to assess mixed muscle fractional protein synthetic rates (FSRs) and myocellular signaling in response to the protein beverages in resting and exercised muscle. RESULTS Mixed muscle FSRs increased following MILK ingestion (from 0.036 ± 0.008 to 0.052 ± 0.006%·h-1 in rested, and 0.035 ± 0.008 to 0.056 ± 0.005%·h-1 in exercised muscle; P <0.01) but to a greater extent following MYCO ingestion (from 0.025 ± 0.006 to 0.057 ± 0.004%·h-1 in rested, and 0.024 ± 0.007 to 0.072 ± 0.005%·h-1 in exercised muscle; P <0.0001) (treatment × time interaction effect; P <0.05). Postprandial FSRs trended to be greater in MYCO compared with MILK (0.065 ± 0.004 compared with 0.054 ± 0.004%·h-1, respectively; P = 0.093) and the postprandial rise in FSRs was greater in MYCO compared with MILK (Delta 0.040 ± 0.006 compared with Delta 0.018 ± 0.005%·h-1, respectively; P <0.01). CONCLUSIONS The ingestion of a single bolus of mycoprotein stimulates resting and postexercise muscle protein synthesis rates, and to a greater extent than a leucine-matched bolus of milk protein, in resistance-trained young men. This trial was registered at clinicaltrials.gov as 660065600.
Collapse
Affiliation(s)
- Alistair J Monteyne
- Department of Sport and Health Sciences, College of Life and Environmental Sciences, Heavitree Road, University of Exeter, Exeter, UK
| | - Mariana O C Coelho
- Department of Sport and Health Sciences, College of Life and Environmental Sciences, Heavitree Road, University of Exeter, Exeter, UK
| | - Craig Porter
- Department of Surgery, University of Texas Medical Branch & Shriners Hospital for Children, Galveston, TX, USA
| | - Doaa R Abdelrahman
- Department of Surgery, University of Texas Medical Branch & Shriners Hospital for Children, Galveston, TX, USA
| | - Thomas S O Jameson
- Department of Sport and Health Sciences, College of Life and Environmental Sciences, Heavitree Road, University of Exeter, Exeter, UK
| | - Sarah R Jackman
- Department of Sport and Health Sciences, College of Life and Environmental Sciences, Heavitree Road, University of Exeter, Exeter, UK
| | - Jamie R Blackwell
- Department of Sport and Health Sciences, College of Life and Environmental Sciences, Heavitree Road, University of Exeter, Exeter, UK
| | | | - Francis B Stephens
- Department of Sport and Health Sciences, College of Life and Environmental Sciences, Heavitree Road, University of Exeter, Exeter, UK
| | - Marlou L Dirks
- Department of Sport and Health Sciences, College of Life and Environmental Sciences, Heavitree Road, University of Exeter, Exeter, UK
| | - Benjamin T Wall
- Department of Sport and Health Sciences, College of Life and Environmental Sciences, Heavitree Road, University of Exeter, Exeter, UK
| |
Collapse
|
28
|
Abou Sawan S, Mazzulla M, Moore DR, Hodson N. More than just a garbage can: emerging roles of the lysosome as an anabolic organelle in skeletal muscle. Am J Physiol Cell Physiol 2020; 319:C561-C568. [PMID: 32726158 DOI: 10.1152/ajpcell.00241.2020] [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] [Indexed: 12/17/2022]
Abstract
Skeletal muscle is a highly plastic tissue capable of remodeling in response to a range of physiological stimuli, including nutrients and exercise. Historically, the lysosome has been considered an essentially catabolic organelle contributing to autophagy, phagocytosis, and exo-/endocytosis in skeletal muscle. However, recent evidence has emerged of several anabolic roles for the lysosome, including the requirement for autophagy in skeletal muscle mass maintenance, the discovery of the lysosome as an intracellular signaling hub for mechanistic target of rapamycin complex 1 (mTORC1) activation, and the importance of transcription factor EB/lysosomal biogenesis-related signaling in the regulation of mTORC1-mediated protein synthesis. We, therefore, propose that the lysosome is an understudied organelle with the potential to underpin the skeletal muscle adaptive response to anabolic stimuli. Within this review, we describe the molecular regulation of lysosome biogenesis and detail the emerging anabolic roles of the lysosome in skeletal muscle with particular emphasis on how these roles may mediate adaptations to chronic resistance exercise. Furthermore, given the well-established role of amino acids to support muscle protein remodeling, we describe how dietary proteins "labeled" with stable isotopes could provide a complementary research tool to better understand how lysosomal biogenesis, autophagy regulation, and/or mTORC1-lysosomal repositioning can mediate the intracellular usage of dietary amino acids in response to anabolic stimuli. Finally, we provide avenues for future research with the aim of elucidating how the regulation of this important organelle could mediate skeletal muscle anabolism.
Collapse
Affiliation(s)
- Sidney Abou Sawan
- Department of Exercise Science, Faculty of Kinesiology and Physical Education, University of Toronto, Toronto, Ontario, Canada
| | - Michael Mazzulla
- Department of Exercise Science, Faculty of Kinesiology and Physical Education, University of Toronto, Toronto, Ontario, Canada
| | - Daniel R Moore
- Department of Exercise Science, Faculty of Kinesiology and Physical Education, University of Toronto, Toronto, Ontario, Canada
| | - Nathan Hodson
- Department of Exercise Science, Faculty of Kinesiology and Physical Education, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
29
|
Hannaian SJ, Hodson N, Abou Sawan S, Mazzulla M, Kato H, Matsunaga K, Waskiw-Ford M, Duncan J, Kumbhare DA, Moore DR. Leucine-enriched amino acids maintain peripheral mTOR-Rheb localization independent of myofibrillar protein synthesis and mTORC1 signaling postexercise. J Appl Physiol (1985) 2020; 129:133-143. [PMID: 32525432 DOI: 10.1152/japplphysiol.00241.2020] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Postexercise protein ingestion can elevate rates of myofibrillar protein synthesis (MyoPS), mTORC1 activity, and mTOR translocation/protein-protein interactions. However, it is unclear if leucine-enriched essential amino acids (LEAA) can similarly facilitate intracellular mTOR trafficking in humans after exercise. The purpose of this study was to determine the effect of postexercise LEAA (4 g total EAAs, 1.6 g leucine) on acute MyoPS and mTORC1 translocation and signaling. Recreationally active men performed lower-body resistance exercise (5 × 8-10 leg press and leg extension) to volitional failure. Following exercise participants consumed LEAA (n = 8) or an isocaloric carbohydrate drink (PLA; n = 10). MyoPS was measured over 1.5-4 h of recovery by oral pulse of l-[ring-2H5]-phenylalanine. Phosphorylation of proteins in the mTORC1 pathway were analyzed via immunoblotting and mTORC1-LAMP2/WGA/Rheb colocalization via immunofluorescence microscopy. There was no difference in MyoPS between groups (LEAA = 0.098 ± 0.01%/h; PL = 0.090 ± 0.01%/h; P > 0.05). Exercise increased (P < 0.05) rpS6Ser240/244(LEAA = 35.3-fold; PLA = 20.6-fold), mTORSer2448(LEAA = 1.8-fold; PLA = 1.2-fold) and 4EBP1Thr37/46(LEAA = 1.5-fold; PLA = 1.4-fold) phosphorylation irrespective of nutrition (P > 0.05). LAT1 and SNAT2 protein expression were not affected by exercise or nutrient ingestion. mTOR-LAMP2 colocalization was greater in LEAA preexercise and decreased following exercise and supplement ingestion (P < 0.05), yet was unchanged in PLA. mTOR-WGA (cell periphery marker) and mTOR-Rheb colocalization was greater in LEAA compared with PLA irrespective of time-point (P < 0.05). In conclusion, the postexercise consumption of 4 g of LEAA maintains mTOR in peripheral regions of muscle fibers, in closer proximity to its direct activator Rheb, during prolonged recovery independent of differences in MyoPS or mTORC1 signaling compared with PLA ingestion. This intracellular localization of mTOR may serve to "prime" the kinase for future anabolic stimuli.NEW & NOTEWORTHY This is the first study to investigate whether postexercise leucine-enriched amino acid (LEAA) ingestion elevates mTORC1 translocation and protein-protein interactions in human skeletal muscle. Here, we observed that although LEAA ingestion did not further elevate postexercise MyoPS or mTORC1 signaling compared with placebo, mTORC1 peripheral location and interaction with Rheb were maintained. This may serve to "prime" mTORC1 for subsequent anabolic stimuli.
Collapse
Affiliation(s)
- Sarkis J Hannaian
- Faculty of Kinesiology and Physical Education, Department of Exercise Science, University of Toronto, Toronto, Canada
| | - Nathan Hodson
- Faculty of Kinesiology and Physical Education, Department of Exercise Science, University of Toronto, Toronto, Canada
| | - Sidney Abou Sawan
- Faculty of Kinesiology and Physical Education, Department of Exercise Science, University of Toronto, Toronto, Canada
| | - Michael Mazzulla
- Faculty of Kinesiology and Physical Education, Department of Exercise Science, University of Toronto, Toronto, Canada
| | - Hiroyuki Kato
- Technology Development Center, Institute of Food Sciences and Technologies, Ajinomoto Co., Inc., Kawasaki, Kanagawa, Japan
| | - Keiko Matsunaga
- Technology Development Center, Institute of Food Sciences and Technologies, Ajinomoto Co., Inc., Kawasaki, Kanagawa, Japan
| | - Marcus Waskiw-Ford
- Faculty of Kinesiology and Physical Education, Department of Exercise Science, University of Toronto, Toronto, Canada
| | - Justin Duncan
- Faculty of Kinesiology and Physical Education, Department of Exercise Science, University of Toronto, Toronto, Canada
| | | | - Daniel R Moore
- Faculty of Kinesiology and Physical Education, Department of Exercise Science, University of Toronto, Toronto, Canada
| |
Collapse
|
30
|
Zhu M, Wang XQ. Regulation of mTORC1 by Small GTPases in Response to Nutrients. J Nutr 2020; 150:1004-1011. [PMID: 31965176 DOI: 10.1093/jn/nxz301] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2019] [Revised: 10/07/2019] [Accepted: 11/18/2019] [Indexed: 12/15/2022] Open
Abstract
Mechanistic target of rapamycin complex 1 (mTORC1) is a highly evolutionarily conserved serine/threonine kinase that regulates cell growth and metabolism in response to multiple environmental cues, such as nutrients, hormones, energy, and stress. Deregulation of mTORC1 can lead to diseases such as diabetes, obesity, and cancer. A series of small GTPases, including Rag, Ras homolog enriched in brain (Rheb), adenosine diphosphate ribosylation factor 1 (Arf1), Ras-related protein Ral-A, Ras homolog (Rho), and Rab, are involved in regulating mTORC1 in response to nutrients, and mTORC1 is differentially regulated via these small GTPases according to specific conditions. Leucine and arginine sensing are considered to be well-confirmed amino acid-sensing signals, activating mTORC1 via a Rag GTPase-dependent mechanism as well as the Ragulator complex and vacuolar H+-adenosine triphosphatase (v-ATPase). Glutamine promotes mTORC1 activation via Arf1 independently of the Rag GTPase. In this review, we summarize current knowledge regarding the regulation of mTORC1 activity by small GTPases in response to nutrients, focusing on the function of small GTPases in mTORC1 activation and how small GTPases are regulated by nutrients.
Collapse
Affiliation(s)
- Min Zhu
- College of Animal Science, South China Agricultural University/Guangdong Provincial Key Laboratory of Animal Nutrition Control/National Engineering Research Center for Breeding Swine Industry, Guangzhou, Guangdong, China
| | - Xiu-Qi Wang
- College of Animal Science, South China Agricultural University/Guangdong Provincial Key Laboratory of Animal Nutrition Control/National Engineering Research Center for Breeding Swine Industry, Guangzhou, Guangdong, China
| |
Collapse
|
31
|
McGlory C, Calder PC, Nunes EA. The Influence of Omega-3 Fatty Acids on Skeletal Muscle Protein Turnover in Health, Disuse, and Disease. Front Nutr 2019; 6:144. [PMID: 31555658 PMCID: PMC6742725 DOI: 10.3389/fnut.2019.00144] [Citation(s) in RCA: 103] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Accepted: 08/19/2019] [Indexed: 12/14/2022] Open
Abstract
Ingestion of omega-3 fatty acids is known to exert favorable health effects on a number of biological processes such as improved immune profile, enhanced cognition, and optimized neuromuscular function. Recently, data have emerged demonstrating a positive influence of omega-3 fatty acid intake on skeletal muscle. For instance, there are reports of clinically-relevant gains in muscle size and strength in healthy older persons with omega-3 fatty acid intake as well as evidence that omega-3 fatty acid ingestion alleviates the loss of muscle mass and prevents decrements in mitochondrial respiration during periods of muscle-disuse. Cancer cachexia that is characterized by a rapid involuntary loss of lean mass may also be attenuated by omega-3 fatty acid provision. The primary means by which omega-3 fatty acids positively impact skeletal muscle mass is via incorporation of eicosapentaenoic acid (EPA; 20:5n−3) and docosahexaenoic acid (DHA; 22:6n−3) into membrane phospholipids of the sarcolemma and intracellular organelles. Enrichment of EPA and DHA in these membrane phospholipids is linked to enhanced rates of muscle protein synthesis, decreased expression of factors that regulate muscle protein breakdown, and improved mitochondrial respiration kinetics. However, exactly how incorporation of EPA and DHA into phospholipid membranes alters these processes remains unknown. In this review, we discuss the interaction between omega-3 fatty acid ingestion and skeletal muscle protein turnover in response to nutrient provision in younger and older adults. Additionally, we examine the role of omega-3 fatty acid supplementation in protecting muscle loss during muscle-disuse and in cancer cachexia, and critically evaluate the molecular mechanisms that underpin the phenotypic changes observed in skeletal muscle with omega-3 fatty acid intake.
Collapse
Affiliation(s)
- Chris McGlory
- School of Kinesiology and Health Studies, Queen's University, Kingston, ON, Canada
| | - Philip C Calder
- Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, United Kingdom.,NIHR Southampton Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust and University of Southampton, Southampton, United Kingdom
| | - Everson A Nunes
- Department of Physiological Sciences, Federal University of Santa Catarina, Florianópolis, Brazil
| |
Collapse
|
32
|
Hodson N, West DWD, Philp A, Burd NA, Moore DR. Molecular regulation of human skeletal muscle protein synthesis in response to exercise and nutrients: a compass for overcoming age-related anabolic resistance. Am J Physiol Cell Physiol 2019; 317:C1061-C1078. [PMID: 31461340 DOI: 10.1152/ajpcell.00209.2019] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Skeletal muscle mass, a strong predictor of longevity and health in humans, is determined by the balance of two cellular processes, muscle protein synthesis (MPS) and muscle protein breakdown. MPS seems to be particularly sensitive to changes in mechanical load and/or nutritional status; therefore, much research has focused on understanding the molecular mechanisms that underpin this cellular process. Furthermore, older individuals display an attenuated MPS response to anabolic stimuli, termed anabolic resistance, which has a negative impact on muscle mass and function, as well as quality of life. Therefore, an understanding of which, if any, molecular mechanisms contribute to anabolic resistance of MPS is of vital importance in formulation of therapeutic interventions for such populations. This review summarizes the current knowledge of the mechanisms that underpin MPS, which are broadly divided into mechanistic target of rapamycin complex 1 (mTORC1)-dependent, mTORC1-independent, and ribosomal biogenesis-related, and describes the evidence that shows how they are regulated by anabolic stimuli (exercise and/or nutrition) in healthy human skeletal muscle. This review also summarizes evidence regarding which of these mechanisms may be implicated in age-related skeletal muscle anabolic resistance and provides recommendations for future avenues of research that can expand our knowledge of this area.
Collapse
Affiliation(s)
- Nathan Hodson
- Faculty of Kinesiology and Physical Education, University of Toronto, Toronto, Ontario, Canada
| | - Daniel W D West
- Faculty of Kinesiology and Physical Education, University of Toronto, Toronto, Ontario, Canada
| | - Andrew Philp
- Garvan Institute of Medical Research, Darlinghurst, Sydney, New South Wales, Australia
| | - Nicholas A Burd
- Department of Kinesiology and Community Health, University of Illinois, Urbana, Illinois
| | - Daniel R Moore
- Faculty of Kinesiology and Physical Education, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
33
|
Boppart MD, Mahmassani ZS. Integrin signaling: linking mechanical stimulation to skeletal muscle hypertrophy. Am J Physiol Cell Physiol 2019; 317:C629-C641. [PMID: 31314586 DOI: 10.1152/ajpcell.00009.2019] [Citation(s) in RCA: 71] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
The α7β1-integrin is a transmembrane adhesion protein that connects laminin in the extracellular matrix (ECM) with actin in skeletal muscle fibers. The α7β1-integrin is highly expressed in skeletal muscle and is concentrated at costameres and myotendious junctions, providing the opportunity to transmit longitudinal and lateral forces across the membrane. Studies have demonstrated that α7-integrin subunit mRNA and protein are upregulated following eccentric contractions as a mechanism to reinforce load-bearing structures and resist injury with repeated bouts of exercise. It has been hypothesized for many years that the integrin can also promote protein turnover in a manner that can promote beneficial adaptations with resistance exercise training, including hypertrophy. This review provides basic information about integrin structure and activation and then explores its potential to serve as a critical mechanosensor and activator of muscle protein synthesis and growth. Overall, the hypothesis is proposed that the α7β1-integrin can contribute to mechanical-load induced skeletal muscle growth via an mammalian target of rapamycin complex 1-independent mechanism.
Collapse
Affiliation(s)
- Marni D Boppart
- Department of Kinesiology and Community Health, University of Illinois at Urbana-Champaign, Urbana, Illinois.,Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, Illinois
| | - Ziad S Mahmassani
- Department of Physical Therapy and Athletic Training, University of Utah, Salt Lake City, Utah
| |
Collapse
|
34
|
Rivas DA, Rice NP, Ezzyat Y, McDonald DJ, Cooper BE, Fielding RA. Sphingosine-1-phosphate analog FTY720 reverses obesity but not age-induced anabolic resistance to muscle contraction. Am J Physiol Cell Physiol 2019; 317:C502-C512. [PMID: 31241988 DOI: 10.1152/ajpcell.00455.2018] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Sarcopenia, the age-associated loss of skeletal muscle mass and function, is coupled with declines in physical functioning leading to subsequent higher rates of disability, frailty, morbidity, and mortality. Aging and obesity independently contribute to muscle atrophy that is assumed to be a result of the activation of mutual physiological pathways. Understanding mechanisms contributing to the induction of skeletal muscle atrophy with aging and obesity is important for determining targets that may have pivotal roles in muscle loss in these conditions. We find that aging and obesity equally induce an anabolic resistance to acute skeletal muscle contraction as observed with decreases in anabolic signaling activation after contraction. Furthermore, treatment with the sphingosine-1-phosphate analog FTY720 for 4 wk increased lean mass and strength, and the anabolic signaling response to contraction was improved in obese but not older animals. To determine the role of chronic inflammation and different fatty acids on anabolic resistance in skeletal muscle cells, we overexpressed IKKβ with and without exposure to saturated fatty acid (SFA; palmitic acid), polyunsaturated fatty acid (eicosapentaenoic acid), and monounsaturated fatty acid (oleic acid). We found that IKKβ overexpression increased inflammation markers in muscle cells, and this chronic inflammation exacerbated anabolic resistance in response to SFA. Pretreatment with FTY720 reversed the inflammatory effects of palmitic acid in the muscle cells. Taken together, these data demonstrate chronic inflammation can induce anabolic resistance, SFA aggravates these effects, and FTY720 can reverse this by decreasing ceramide accumulation in skeletal muscle.
Collapse
Affiliation(s)
- Donato A Rivas
- Nutrition, Exercise Physiology and Sarcopenia Laboratory, Jean Mayer U.S. Department of Agriculture Human Nutrition Research Center on Aging at Tufts University, Boston, Massachusetts
| | - Nicholas P Rice
- Nutrition, Exercise Physiology and Sarcopenia Laboratory, Jean Mayer U.S. Department of Agriculture Human Nutrition Research Center on Aging at Tufts University, Boston, Massachusetts
| | - Yassine Ezzyat
- Nutrition, Exercise Physiology and Sarcopenia Laboratory, Jean Mayer U.S. Department of Agriculture Human Nutrition Research Center on Aging at Tufts University, Boston, Massachusetts
| | - Devin J McDonald
- Nutrition, Exercise Physiology and Sarcopenia Laboratory, Jean Mayer U.S. Department of Agriculture Human Nutrition Research Center on Aging at Tufts University, Boston, Massachusetts
| | - Brittany E Cooper
- Nutrition, Exercise Physiology and Sarcopenia Laboratory, Jean Mayer U.S. Department of Agriculture Human Nutrition Research Center on Aging at Tufts University, Boston, Massachusetts
| | - Roger A Fielding
- Nutrition, Exercise Physiology and Sarcopenia Laboratory, Jean Mayer U.S. Department of Agriculture Human Nutrition Research Center on Aging at Tufts University, Boston, Massachusetts
| |
Collapse
|
35
|
Wu JZ, Ardah M, Haikal C, Svanbergsson A, Diepenbroek M, Vaikath NN, Li W, Wang ZY, Outeiro TF, El-Agnaf OM, Li JY. Dihydromyricetin and Salvianolic acid B inhibit alpha-synuclein aggregation and enhance chaperone-mediated autophagy. Transl Neurodegener 2019; 8:18. [PMID: 31223479 PMCID: PMC6570948 DOI: 10.1186/s40035-019-0159-7] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Accepted: 05/24/2019] [Indexed: 02/02/2023] Open
Abstract
BACKGROUND Progressive accumulation of α-synuclein is a key step in the pathological development of Parkinson's disease. Impaired protein degradation and increased levels of α-synuclein may trigger a pathological aggregation in vitro and in vivo. The chaperone-mediated autophagy (CMA) pathway is involved in the intracellular degradation processes of α-synuclein. Dysfunction of the CMA pathway impairs α-synuclein degradation and causes cytotoxicity. RESULTS In the present study, we investigated the effects on the CMA pathway and α-synuclein aggregation using bioactive ingredients (Dihydromyricetin (DHM) and Salvianolic acid B (Sal B)) extracted from natural medicinal plants. In both cell-free and cellular models of α-synuclein aggregation, after administration of DHM and Sal B, we observed significant inhibition of α-synuclein accumulation and aggregation. Cells were co-transfected with a C-terminal modified α-synuclein (SynT) and synphilin-1, and then treated with DHM (10 μM) and Sal B (50 μM) 16 hours after transfection; levels of α-synuclein aggregation decreased significantly (68% for DHM and 75% for Sal B). Concomitantly, we detected increased levels of LAMP-1 (a marker of lysosomal homeostasis) and LAMP-2A (a key marker of CMA). Immunofluorescence analyses showed increased colocalization between LAMP-1 and LAMP-2A with α-synuclein inclusions after treatment with DHM and Sal B. We also found increased levels of LAMP-1 and LAMP-2A both in vitro and in vivo, along with decreased levels of α-synuclein. Moreover, DHM and Sal B treatments exhibited anti-inflammatory activities, preventing astroglia- and microglia-mediated neuroinflammation in BAC-α-syn-GFP transgenic mice. CONCLUSIONS Our data indicate that DHM and Sal B are effective in modulating α-synuclein accumulation and aggregate formation and augmenting activation of CMA, holding potential for the treatment of Parkinson's disease.
Collapse
Affiliation(s)
- Jia-Zhen Wu
- Institute of Neuroscience, College of Life and Health Sciences, Northeastern University, Shenyang, Liaoning China
- Key Laboratory of Maternal-Fetal Medicine of Liaoning Province and Key Laboratory of Obstetrics and Gynecology of Higher Education of Liaoning Province, Shengjing Hospital of China Medical University, Shenyang, China
| | - Mustafa Ardah
- Department of Biochemistry, College of Medicine and Health Sciences, United Arab Emirates University, PO Box 17666, Al-Ain, United Arab Emirates
| | - Caroline Haikal
- Neural Plasticity and Repair Unit, Wallenberg Neuroscience Center, Department of Experimental Medical Science, BMC A10, 221 84 Lund, Sweden
| | - Alexander Svanbergsson
- Neural Plasticity and Repair Unit, Wallenberg Neuroscience Center, Department of Experimental Medical Science, BMC A10, 221 84 Lund, Sweden
| | - Meike Diepenbroek
- Neural Plasticity and Repair Unit, Wallenberg Neuroscience Center, Department of Experimental Medical Science, BMC A10, 221 84 Lund, Sweden
| | - Nishant N. Vaikath
- Neural Plasticity and Repair Unit, Wallenberg Neuroscience Center, Department of Experimental Medical Science, BMC A10, 221 84 Lund, Sweden
- Neurological Disorders Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Education City, Qatar Foundation, P.O. Box 5825, Doha, Qatar
| | - Wen Li
- Neural Plasticity and Repair Unit, Wallenberg Neuroscience Center, Department of Experimental Medical Science, BMC A10, 221 84 Lund, Sweden
| | - Zhan-You Wang
- Institute of Heath Sciences, China Medical University, 110112 Shenyang, People’s Republic of China
| | - Tiago F. Outeiro
- Department of Experimental Neurodegeneration, Center for Nanoscale Microscopy and Molecular Physiology of the Brain, Center for Biostructural Imaging of Neurodegeneration, University Medical Center Göttingen, 37073 Göttingen, Germany
- Max Planck Institute for Experimental Medicine, Göttingen, Germany
| | - Omar M. El-Agnaf
- Neurological Disorders Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Education City, Qatar Foundation, P.O. Box 5825, Doha, Qatar
| | - Jia-Yi Li
- Institute of Neuroscience, College of Life and Health Sciences, Northeastern University, Shenyang, Liaoning China
- Neural Plasticity and Repair Unit, Wallenberg Neuroscience Center, Department of Experimental Medical Science, BMC A10, 221 84 Lund, Sweden
- Institute of Heath Sciences, China Medical University, 110112 Shenyang, People’s Republic of China
| |
Collapse
|
36
|
Beals JW, Burd NA, Moore DR, van Vliet S. Obesity Alters the Muscle Protein Synthetic Response to Nutrition and Exercise. Front Nutr 2019; 6:87. [PMID: 31263701 PMCID: PMC6584965 DOI: 10.3389/fnut.2019.00087] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Accepted: 05/23/2019] [Indexed: 12/17/2022] Open
Abstract
Improving the health of skeletal muscle is an important component of obesity treatment. Apart from allowing for physical activity, skeletal muscle tissue is fundamental for the regulation of postprandial macronutrient metabolism, a time period that represents when metabolic derangements are most often observed in adults with obesity. In order for skeletal muscle to retain its capacity for physical activity and macronutrient metabolism, its protein quantity and composition must be maintained through the efficient degradation and resynthesis for proper tissue homeostasis. Life-style behaviors such as increasing physical activity and higher protein diets are front-line treatment strategies to enhance muscle protein remodeling by primarily stimulating protein synthesis rates. However, the muscle of individuals with obesity appears to be resistant to the anabolic action of targeted exercise regimes and protein ingestion when compared to normal-weight adults. This indicates impaired muscle protein remodeling in response to the main anabolic stimuli to human skeletal muscle tissue is contributing to poor muscle health with obesity. Deranged anabolic signaling related to insulin resistance, lipid accumulation, and/or systemic/muscle inflammation are likely at the root of the anabolic resistance of muscle protein synthesis rates with obesity. The purpose of this review is to discuss the impact of protein ingestion and exercise on muscle protein remodeling in people with obesity, and the potential mechanisms underlining anabolic resistance of their muscle.
Collapse
Affiliation(s)
- Joseph W Beals
- Center for Human Nutrition, Washington University School of Medicine, St. Louis, MO, United States
| | - Nicholas A Burd
- Department of Kinesiology and Community Health, University of Illinois at Urbana-Champaign, Urbana, IL, United States
| | - Daniel R Moore
- Faculty of Kinesiology and Physical Education, University of Toronto, Toronto, ON, Canada
| | - Stephan van Vliet
- Duke Molecular Physiology Institute, Duke University Medical Center, Durham, NC, United States
| |
Collapse
|
37
|
McKenna CF, Salvador AF, Hendriks FK, Harris APY, van Loon LJC, Burd NA. Exercising to offset muscle mass loss in hemodialysis patients: The disconnect between intention and intervention. Semin Dial 2019; 32:379-385. [DOI: 10.1111/sdi.12805] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
| | - Amadeo F. Salvador
- Department of Kinesiology and Community Health University of Illinois Urbana Illinois
| | - Floris K. Hendriks
- Department of Human Biology NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre+ Maastricht The Netherlands
| | - Alana P. Y. Harris
- Department of Kinesiology and Community Health University of Illinois Urbana Illinois
| | - Luc J. C. van Loon
- Department of Human Biology NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre+ Maastricht The Netherlands
| | - Nicholas A. Burd
- Division of Nutritional Sciences University of Illinois Urbana Illinois
- Department of Kinesiology and Community Health University of Illinois Urbana Illinois
| |
Collapse
|
38
|
Tinline-Goodfellow C, Estafanos S, Adams C, Bruccoleri G, Dellatolla J, McLaughlin M. When bigger isn't better: understanding the anabolic resistance of obese skeletal muscle. J Physiol 2019; 597:2129-2130. [PMID: 30835830 DOI: 10.1113/jp277805] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Affiliation(s)
| | - Stephanie Estafanos
- Faculty of Kinesiology and Physical Education, University of Toronto, Toronto, ON, Canada
| | - Carolyn Adams
- Faculty of Kinesiology and Physical Education, University of Toronto, Toronto, ON, Canada
| | - Giovanni Bruccoleri
- Faculty of Kinesiology and Physical Education, University of Toronto, Toronto, ON, Canada
| | - Jason Dellatolla
- Faculty of Kinesiology and Physical Education, University of Toronto, Toronto, ON, Canada
| | - Mackenzie McLaughlin
- Faculty of Kinesiology and Physical Education, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
39
|
Abou Sawan S, van Vliet S, Parel JT, Beals JW, Mazzulla M, West DWD, Philp A, Li Z, Paluska SA, Burd NA, Moore DR. Translocation and protein complex co-localization of mTOR is associated with postprandial myofibrillar protein synthesis at rest and after endurance exercise. Physiol Rep 2019; 6. [PMID: 29512299 PMCID: PMC5840389 DOI: 10.14814/phy2.13628] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Accepted: 01/31/2018] [Indexed: 01/27/2023] Open
Abstract
Translocation and colocalization of mechanistic target of rapamycin complex 1 (mTORC1) with regulatory proteins represents a critical step in translation initiation of protein synthesis in vitro. However, mechanistic insight into the control of postprandial skeletal muscle protein synthesis rates at rest and after an acute bout of endurance exercise in humans is lacking. In crossover trials, eight endurance‐trained men received primed‐continuous infusions of L‐[ring‐2H5]phenylalanine and consumed a mixed‐macronutrient meal (18 g protein, 60 g carbohydrates, 17 g fat) at rest (REST) and after 60 min of treadmill running at 70% VO2peak (EX). Skeletal muscle biopsies were collected to measure changes in phosphorylation and colocalization in the mTORC1‐pathway, in addition to rates of myofibrillar (MyoPS) and mitochondrial (MitoPS) protein synthesis. MyoPS increased (P < 0.05) above fasted in REST (~2.1‐fold) and EX (~twofold) during the 300 min postprandial period, with no corresponding changes in MitoPS (P > 0.05). TSC2/Rheb colocalization decreased below fasted at 60 and 300 min after feeding in REST and EX (P < 0.01). mTOR colocalization with Rheb increased above fasted at 60 and 300 min after feeding in REST and EX (P < 0.01), which was consistent with an increased phosphorylation 4E‐BP1Thr37/46 and rpS6ser240/244 at 60 min. Our data suggest that MyoPS, but not MitoPS, is primarily nutrient responsive in trained young men at rest and after endurance exercise. The postprandial increase in MyoPS is associated with an increase in mTOR/Rheb colocalization and a reciprocal decrease in TSC2/Rheb colocalization and thus likely represent important regulatory events for in vivo skeletal muscle myofibrillar mRNA translation in humans.
Collapse
Affiliation(s)
- Sidney Abou Sawan
- Faculty of Kinesiology and Physical Education, University of Toronto, Toronto, Ontario
| | - Stephan van Vliet
- Department of Kinesiology and Community Health, University of Illinois, Urbana, Illinois
| | - Justin T Parel
- Department of Kinesiology and Community Health, University of Illinois, Urbana, Illinois
| | - Joseph W Beals
- Division of Nutritional Sciences, University of Illinois, Urbana, Illinois
| | - Michael Mazzulla
- Faculty of Kinesiology and Physical Education, University of Toronto, Toronto, Ontario
| | - Daniel W D West
- Faculty of Kinesiology and Physical Education, University of Toronto, Toronto, Ontario
| | - Andrew Philp
- School of Sport, Exercise and Rehabilitation Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Zhong Li
- Roy J. Carver Biotechnology Center, University of Illinois, Urbana, Illinois
| | - Scott A Paluska
- Department of Family Medicine, University of Illinois, Urbana, Illinois
| | - Nicholas A Burd
- Department of Kinesiology and Community Health, University of Illinois, Urbana, Illinois.,Division of Nutritional Sciences, University of Illinois, Urbana, Illinois
| | - Daniel R Moore
- Faculty of Kinesiology and Physical Education, University of Toronto, Toronto, Ontario
| |
Collapse
|
40
|
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: 7.4] [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
|
41
|
Montalvo RN, Counts BR, Carson JA. Understanding sex differences in the regulation of cancer-induced muscle wasting. Curr Opin Support Palliat Care 2018; 12:394-403. [PMID: 30102621 PMCID: PMC6239206 DOI: 10.1097/spc.0000000000000380] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
PURPOSE OF REVIEW We highlight evidence for sexual dimorphism in preclinical and clinical studies investigating the cause and treatment of cancer cachexia. RECENT FINDINGS Cancer cachexia is unintended bodyweight loss occurring with cancer, and skeletal muscle wasting is a critical predictor of negative outcomes in the cancer patient. Skeletal muscle exhibits sexual dimorphism in fiber type, function, and regeneration capacity. Sex differences have been implicated in skeletal muscle metabolism, mitochondrial function, immune response to injury, and myogenic stem cell regulation. All of these processes have the potential to be involved in cancer-induced muscle wasting. Unfortunately, the vast majority of published studies examining cancer cachexia in preclinical models or cancer patients either have not accounted for sex in their design or have exclusively studied males. Preclinical studies have established that ovarian function and estradiol can affect skeletal muscle function, metabolism and mass; ovarian function has also been implicated in the sensitivity of circulating inflammatory cytokines and the progression of cachexia. SUMMARY Females and males have unique characteristics that effect skeletal muscle's microenvironment and intrinsic signaling. These differences provide a strong rationale for distinct causes for cancer cachexia development and treatment in males and females.
Collapse
Affiliation(s)
- Ryan N Montalvo
- Department of Exercise Science, University of South Carolina, Public Health Research Center, Columbia, USA
| | | | | |
Collapse
|
42
|
McGlory C, Nunes EA, Oikawa SY, Tsakiridis E, Phillips SM. Assessing the mechanistic target of rapamycin complex-1 pathway in response to resistance exercise and feeding in human skeletal muscle by multiplex assay. Appl Physiol Nutr Metab 2018; 43:945-949. [PMID: 29590537 DOI: 10.1139/apnm-2017-0852] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The mechanistic target of rapamycin complex-1 (mTORC-1) is a key nutrient and contraction-sensitive protein that regulates a pathway leading to skeletal muscle growth. Utilizing a multiplex assay, we aimed to examine the phosphorylation status of key mTORC-1-related signalling molecules in response to protein feeding and resistance exercise. Eight healthy men (age, 22.5 ± 3.1 years; mass, 80 ± 9 kg; 1-repetition maximum leg extension, 87 ± 5 kg) performed 4 sets of unilateral leg extensions until volitional failure. Immediately following the final set, all participants consumed a protein-enriched beverage. A single skeletal muscle biopsy was obtained from the vastus lateralis before (Pre) with further bilateral biopsies at 1 h (1 h exercised legs (FEDEX) and 1 h nonexercised legs (FED)) and 3 h (3 h FEDEX and 3 h FED) after drink ingestion. Phosphorylated AktSer473 was significantly elevated from Pre at 1 h FEDEX. Phosphorylated p70S6K1Thr412 was significantly increased above Pre at 1 h FEDEX and 1 h FED and was still significantly elevated at 3 h FEDEX but not 3 h FED. Phosphorylated rpS6Ser235/236 was also significantly increased above Pre at 1 h FEDEX and 1 h FED with 1 h FEDEX greater than 1 h FED. Our data highlight the utility of a multiplex assay to assess anabolic signalling molecules in response to protein feeding and resistance exercise in humans. Importantly, these changes are comparable with those as previously reported using standard immunoblotting and protein activity assays.
Collapse
Affiliation(s)
- Chris McGlory
- a Department of Kinesiology, McMaster University, Hamilton, ON L8S 4K1, Canada
| | - Everson A Nunes
- b Department of Physiological Sciences, Federal University of Santa Catarina, Florianópolis, 88040-900, SC, Brazil
| | - Sara Y Oikawa
- a Department of Kinesiology, McMaster University, Hamilton, ON L8S 4K1, Canada
| | | | - Stuart M Phillips
- a Department of Kinesiology, McMaster University, Hamilton, ON L8S 4K1, Canada
| |
Collapse
|
43
|
Abou Sawan S, van Vliet S, West DWD, Beals JW, Paluska SA, Burd NA, Moore DR. Whole egg, but not egg white, ingestion induces mTOR colocalization with the lysosome after resistance exercise. Am J Physiol Cell Physiol 2018; 315:C537-C543. [PMID: 30133322 DOI: 10.1152/ajpcell.00225.2018] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
We have recently demonstrated that whole egg ingestion induces a greater muscle protein synthetic (MPS) response when compared with isonitrogenous egg white ingestion after resistance exercise in young men. Our aim was to determine whether whole egg or egg white ingestion differentially influenced colocalization of key regulators of mechanistic target of rapamycin complex 1 (mTORC1) as means to explain our previously observed divergent postexercise MPS response. In crossover trials, 10 healthy resistance-trained men (21 ± 1 yr; 88 ± 3 kg; body fat: 16 ± 1%; means ± SE) completed lower body resistance exercise before ingesting whole eggs (18 g protein, 17 g fat) or egg whites (18 g protein, 0 g fat). Muscle biopsies were obtained before exercise and at 120 and 300 min after egg ingestion to assess, by immunofluorescence, protein colocalization of key anabolic signaling molecules. After resistance exercise, tuberous sclerosis 2-Ras homolog enriched in brain (Rheb) colocalization decreased ( P < 0.01) at 120 and 300 min after whole egg and egg white ingestion with concomitant increases ( P < 0.01) in mTOR-Rheb colocalization. After resistance exercise, mTOR-lysosome-associated membrane protein 2 (LAMP2) colocalization significantly increased at 120 and 300 min only after whole egg ingestion ( P < 0.01), and mTOR-LAMP2 colocalization correlated with rates of MPS at rest and after exercise ( r = 0.40, P < 0.05). We demonstrated that the greater postexercise MPS response with whole egg ingestion is related in part to an enhanced recruitment of mTORC1-Rheb complexes to the lysosome during recovery. These data suggest nonprotein dietary factors influence the postexercise regulation of mRNA translation in human skeletal muscle.
Collapse
Affiliation(s)
- Sidney Abou Sawan
- Faculty of Kinesiology and Physical Education, University of Toronto , Toronto, Ontario , Canada
| | - Stephan van Vliet
- Department of Kinesiology and Community Health, University of Illinois , Urbana, Illinois
| | - Daniel W D West
- Faculty of Kinesiology and Physical Education, University of Toronto , Toronto, Ontario , Canada
| | - Joseph W Beals
- Division of Nutritional Sciences, University of Illinois , Urbana, Illinois
| | - Scott A Paluska
- Department of Family Medicine, University of Illinois , Urbana, Illinois
| | - Nicholas A Burd
- Department of Kinesiology and Community Health, University of Illinois , Urbana, Illinois.,Division of Nutritional Sciences, University of Illinois , Urbana, Illinois
| | - Daniel R Moore
- Faculty of Kinesiology and Physical Education, University of Toronto , Toronto, Ontario , Canada
| |
Collapse
|
44
|
Moro T, Brightwell CR, Deer RR, Graber TG, Galvan E, Fry CS, Volpi E, Rasmussen BB. Muscle Protein Anabolic Resistance to Essential Amino Acids Does Not Occur in Healthy Older Adults Before or After Resistance Exercise Training. J Nutr 2018; 148:900-909. [PMID: 29796648 PMCID: PMC6251608 DOI: 10.1093/jn/nxy064] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Accepted: 03/11/2018] [Indexed: 12/22/2022] Open
Abstract
Background The muscle protein anabolic response to contraction and feeding may be blunted in older adults. Acute bouts of exercise can improve the ability of amino acids to stimulate muscle protein synthesis (MPS) by activating mechanistic target of rapamycin complex 1 (mTORC1) signaling, but it is not known whether exercise training may improve muscle sensitivity to amino acid availability. Objective The aim of this study was to determine if muscle protein anabolism is resistant to essential amino acids (EAAs) and whether resistance exercise training (RET) improves muscle sensitivity to EAA in healthy older adults. Methods In a longitudinal study, 19 healthy older adults [mean ± SD age: 71 ± 4 y body mass index (kg/m2): 28 ± 3] were trained for 12 wk with a whole-body program of progressive RET (60-75% 1-repetition maximum). Body composition, strength, and metabolic health were measured pre- and posttraining. We also performed stable isotope infusion experiments with muscle biopsies pre- and posttraining to measure MPS and markers of amino acid sensing in the basal state and in response to 6.8 g of EAA ingestion. Results RET increased muscle strength by 16%, lean mass by 2%, and muscle cross-sectional area by 27% in healthy older adults (P < 0.05). MPS and mTORC1 signaling (i.e., phosphorylation status of protein kinase B, 4E binding protein 1, 70-kDa S6 protein kinase, and ribosomal protein S6) increased after EAA ingestion (P < 0.05) pre- and posttraining. RET increased basal MPS by 36% (P < 0.05); however, RET did not affect the response of MPS and mTORC1 signaling to EAA ingestion. Conclusion RET increases strength and basal MPS, promoting hypertrophy in healthy older adults. In these subjects, a small dose of EAAs stimulates muscle mTORC1 signaling and MPS, and this response to EAAs does not improve after RET. Our data indicate that anabolic resistance to amino acids may not be a problem in healthy older adults. This trial was registered at www.clinicaltrials.gov as NCT02999802.
Collapse
Affiliation(s)
- Tatiana Moro
- Department of Nutrition and Metabolism
- Sealy Center on Aging, University of Texas Medical Branch, Galveston, TX
| | | | | | | | | | - Christopher S Fry
- Department of Nutrition and Metabolism
- Sealy Center on Aging, University of Texas Medical Branch, Galveston, TX
| | - Elena Volpi
- Department of Internal Medicine/Geriatrics
- Sealy Center on Aging, University of Texas Medical Branch, Galveston, TX
| | - Blake B Rasmussen
- Department of Nutrition and Metabolism
- Sealy Center on Aging, University of Texas Medical Branch, Galveston, TX
- Address correspondence to BBR (e-mail: )
| |
Collapse
|