1
|
Kubat GB, Bouhamida E, Ulger O, Turkel I, Pedriali G, Ramaccini D, Ekinci O, Ozerklig B, Atalay O, Patergnani S, Nur Sahin B, Morciano G, Tuncer M, Tremoli E, Pinton P. Mitochondrial dysfunction and skeletal muscle atrophy: Causes, mechanisms, and treatment strategies. Mitochondrion 2023; 72:33-58. [PMID: 37451353 DOI: 10.1016/j.mito.2023.07.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 07/02/2023] [Accepted: 07/11/2023] [Indexed: 07/18/2023]
Abstract
Skeletal muscle, which accounts for approximately 40% of total body weight, is one of the most dynamic and plastic tissues in the human body and plays a vital role in movement, posture and force production. More than just a component of the locomotor system, skeletal muscle functions as an endocrine organ capable of producing and secreting hundreds of bioactive molecules. Therefore, maintaining healthy skeletal muscles is crucial for supporting overall body health. Various pathological conditions, such as prolonged immobilization, cachexia, aging, drug-induced toxicity, and cardiovascular diseases (CVDs), can disrupt the balance between muscle protein synthesis and degradation, leading to skeletal muscle atrophy. Mitochondrial dysfunction is a major contributing mechanism to skeletal muscle atrophy, as it plays crucial roles in various biological processes, including energy production, metabolic flexibility, maintenance of redox homeostasis, and regulation of apoptosis. In this review, we critically examine recent knowledge regarding the causes of muscle atrophy (disuse, cachexia, aging, etc.) and its contribution to CVDs. Additionally, we highlight the mitochondrial signaling pathways involvement to skeletal muscle atrophy, such as the ubiquitin-proteasome system, autophagy and mitophagy, mitochondrial fission-fusion, and mitochondrial biogenesis. Furthermore, we discuss current strategies, including exercise, mitochondria-targeted antioxidants, in vivo transfection of PGC-1α, and the potential use of mitochondrial transplantation as a possible therapeutic approach.
Collapse
Affiliation(s)
- Gokhan Burcin Kubat
- Department of Mitochondria and Cellular Research, Gulhane Health Sciences Institute, University of Health Sciences, 06010 Ankara, Turkey.
| | - Esmaa Bouhamida
- Translational Research Center, Maria Cecilia Hospital GVM Care & Research, 48033 Cotignola, Italy
| | - Oner Ulger
- Department of Mitochondria and Cellular Research, Gulhane Health Sciences Institute, University of Health Sciences, 06010 Ankara, Turkey
| | - Ibrahim Turkel
- Department of Exercise and Sport Sciences, Faculty of Sport Sciences, Hacettepe University, 06800 Ankara, Turkey
| | - Gaia Pedriali
- Translational Research Center, Maria Cecilia Hospital GVM Care & Research, 48033 Cotignola, Italy
| | - Daniela Ramaccini
- Translational Research Center, Maria Cecilia Hospital GVM Care & Research, 48033 Cotignola, Italy
| | - Ozgur Ekinci
- Department of Pathology, Gazi University, 06500 Ankara, Turkey
| | - Berkay Ozerklig
- Department of Exercise and Sport Sciences, Faculty of Sport Sciences, Hacettepe University, 06800 Ankara, Turkey
| | - Ozbeyen Atalay
- Department of Physiology, Faculty of Medicine, Hacettepe University, 06230 Ankara, Turkey
| | - Simone Patergnani
- Translational Research Center, Maria Cecilia Hospital GVM Care & Research, 48033 Cotignola, Italy; Department of Medical Sciences, Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, 44121 Ferrara, Italy
| | - Beyza Nur Sahin
- Department of Physiology, Faculty of Medicine, Hacettepe University, 06230 Ankara, Turkey
| | - Giampaolo Morciano
- Translational Research Center, Maria Cecilia Hospital GVM Care & Research, 48033 Cotignola, Italy; Department of Medical Sciences, Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, 44121 Ferrara, Italy
| | - Meltem Tuncer
- Department of Physiology, Faculty of Medicine, Hacettepe University, 06230 Ankara, Turkey
| | - Elena Tremoli
- Translational Research Center, Maria Cecilia Hospital GVM Care & Research, 48033 Cotignola, Italy
| | - Paolo Pinton
- Translational Research Center, Maria Cecilia Hospital GVM Care & Research, 48033 Cotignola, Italy; Department of Medical Sciences, Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, 44121 Ferrara, Italy.
| |
Collapse
|
2
|
Lu W, Feng W, Lai J, Yuan D, Xiao W, Li Y. Role of adipokines in sarcopenia. Chin Med J (Engl) 2023; 136:1794-1804. [PMID: 37442757 PMCID: PMC10406092 DOI: 10.1097/cm9.0000000000002255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Indexed: 07/15/2023] Open
Abstract
ABSTRACT Sarcopenia is an age-related disease that mainly involves decreases in muscle mass, muscle strength and muscle function. At the same time, the body fat content increases with aging, especially the visceral fat content. Adipose tissue is an endocrine organ that secretes biologically active factors called adipokines, which act on local and distant tissues. Studies have revealed that some adipokines exert regulatory effects on muscle, such as higher serum leptin levels causing a decrease in muscle function and adiponectin inhibits the transcriptional activity of Forkhead box O3 (FoxO3) by activating peroxisome proliferators-activated receptor-γ coactivator -1α (PGC-1α) and sensitizing cells to insulin, thereby repressing atrophy-related genes (atrogin-1 and muscle RING finger 1 [MuRF1]) to prevent the loss of muscle mass. Here, we describe the effects on muscle of adipokines produced by adipose tissue, such as leptin, adiponectin, resistin, mucin and lipocalin-2, and discuss the importance of these adipokines for understanding the development of sarcopenia.
Collapse
Affiliation(s)
- Wenhao Lu
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Wenjie Feng
- Department of Burns and Plastic Surgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Jieyu Lai
- Xiangya School of Medicine, Central South University, Changsha, Hunan 410083, China
| | - Dongliang Yuan
- Xiangya School of Medicine, Central South University, Changsha, Hunan 410083, China
| | - Wenfeng Xiao
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Yusheng Li
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| |
Collapse
|
3
|
Keeble AR, Brightwell CR, Latham CM, Thomas NT, Mobley CB, Murach KA, Johnson DL, Noehren B, Fry CS. Depressed Protein Synthesis and Anabolic Signaling Potentiate ACL Tear-Resultant Quadriceps Atrophy. Am J Sports Med 2023; 51:81-96. [PMID: 36475881 PMCID: PMC9813974 DOI: 10.1177/03635465221135769] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND Anterior cruciate ligament (ACL) tear (ACLT) leads to protracted quadriceps muscle atrophy. Protein turnover largely dictates muscle size and is highly responsive to injury and loading. Regulation of quadriceps molecular protein synthetic machinery after ACLT has largely been unexplored, limiting development of targeted therapies. PURPOSE To define the effect of ACLT on (1) the activation of protein synthetic and catabolic signaling within quadriceps biopsy specimens from human participants and (2) the time course of alterations to protein synthesis and its molecular regulation in a mouse ACL injury model. STUDY DESIGN Descriptive laboratory study. METHODS Muscle biopsy specimens were obtained from the ACL-injured and noninjured vastus lateralis of young adult humans after an overnight fast (N = 21; mean ± SD, 19 ± 5 years). Mice had their limbs assigned to ACLT or control, and whole quadriceps were collected 6 hours or 1, 3, or 7 days after injury with puromycin injected before tissue collection for assessment of relative protein synthesis. Muscle fiber size and expression and phosphorylation of protein anabolic and catabolic signaling proteins were assessed at the protein and transcript levels (RNA sequencing). RESULTS Human quadriceps showed reduced phosphorylation of ribosomal protein S6 (-41%) in the ACL-injured limb (P = .008), in addition to elevated phosphorylation of eukaryotic initiation factor 2α (+98%; P = .006), indicative of depressed protein anabolic signaling in the injured limb. No differences in E3 ubiquitin ligase expression were noted. Protein synthesis was lower at 1 day (P = .01 vs control limb) and 3 days (P = .002 vs control limb) after ACLT in mice. Pathway analyses revealed shared molecular alterations between human and mouse quadriceps after ACLT. CONCLUSION (1) Global protein synthesis and anabolic signaling deficits occur in the quadriceps in response to ACL injury, without notable changes in measured markers of muscle protein catabolism. (2) Importantly, these deficits occur before the onset of significant atrophy, underscoring the need for early intervention. CLINICAL RELEVANCE These findings suggest that blunted protein anabolism as opposed to increased catabolism likely mediates quadriceps atrophy after ACL injury. Thus, future interventions should aim to restore muscle protein anabolism rapidly after ACLT.
Collapse
Affiliation(s)
- Alexander R. Keeble
- Department of Physiology, College of Medicine, University of Kentucky
- Center for Muscle Biology, University of Kentucky
| | - Camille R. Brightwell
- Center for Muscle Biology, University of Kentucky
- Department of Athletic Training and Clinical Nutrition, University of Kentucky
| | - Christine M. Latham
- Center for Muscle Biology, University of Kentucky
- Department of Athletic Training and Clinical Nutrition, University of Kentucky
| | - Nicholas T. Thomas
- Center for Muscle Biology, University of Kentucky
- Department of Athletic Training and Clinical Nutrition, University of Kentucky
| | - C. Brooks Mobley
- Department of Physiology, College of Medicine, University of Kentucky
- Center for Muscle Biology, University of Kentucky
| | - Kevin A. Murach
- Center for Muscle Biology, University of Kentucky
- Department of Physical Therapy, University of Kentucky
| | - Darren L. Johnson
- Department of Orthopaedic Surgery & Sports Medicine, University of Kentucky
| | - Brian Noehren
- Center for Muscle Biology, University of Kentucky
- Department of Physical Therapy, University of Kentucky
- Department of Orthopaedic Surgery & Sports Medicine, University of Kentucky
| | - Christopher S. Fry
- Center for Muscle Biology, University of Kentucky
- Department of Athletic Training and Clinical Nutrition, University of Kentucky
| |
Collapse
|
4
|
Vechetti IJ, Norrbom J, Alkner B, Hjalmarsson E, Palmcrantz A, Pontén E, Pingel J, von Walden F, Fernandez-Gonzalo R. Extracellular vesicle characteristics and microRNA content in cerebral palsy and typically developed individuals at rest and in response to aerobic exercise. Front Physiol 2022; 13:1072040. [PMID: 36620222 PMCID: PMC9811128 DOI: 10.3389/fphys.2022.1072040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Accepted: 12/06/2022] [Indexed: 12/24/2022] Open
Abstract
In this study, the properties of circulating extracellular vesicles (EVs) were examined in cerebral palsy (CP) and typically developed (TD) individuals at rest and after aerobic exercise, focusing on the size, concentration, and microRNA cargo of EVs. Nine adult individuals with CP performed a single exercise bout consisting of 45 min of Frame Running, and TD participants completed either 45 min of cycling (n = 10; TD EX) or were enrolled as controls with no exercise (n = 10; TD CON). Blood was drawn before and 30 min after exercise and analyzed for EV concentration, size, and microRNA content. The size of EVs was similar in CP vs. TD, and exercise had no effect. Individuals with CP had an overall lower concentration (∼25%, p < 0.05) of EVs. At baseline, let-7a, let-7b and let-7e were downregulated in individuals with CP compared to TD (p < 0.05), while miR-100 expression was higher, and miR-877 and miR-4433 lower in CP compared to TD after exercise (p < 0.05). Interestingly, miR-486 was upregulated ∼2-fold in the EVs of CP vs. TD both at baseline and after exercise. We then performed an in silico analysis of miR-486 targets and identified the satellite cell stemness factor Pax7 as a target of miR-486. C2C12 myoblasts were cultured with a miR-486 mimetic and RNA-sequencing was performed. Gene enrichment analysis revealed that several genes involved in sarcomerogenesis and extracellular matrix (ECM) were downregulated. Our data suggest that circulating miR-486 transported by EVs is elevated in individuals with CP and that miR-486 alters the transcriptome of myoblasts affecting both ECM- and sarcomerogenesis-related genes, providing a link to the skeletal muscle alterations observed in individuals with CP.
Collapse
Affiliation(s)
- Ivan J. Vechetti
- Department of Nutrition and Health Sciences, College of Education and Human Sciences, University of Nebraska-Lincoln, Lincoln, NE, United States
| | - Jessica Norrbom
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Björn Alkner
- Department of Orthopaedics, Eksjö, Region Jönköping County and Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Emma Hjalmarsson
- Division of Pediatric Neurology, Department of Women’s and Children’s Health, Karolinska Institutet, Stockholm, Sweden
| | - Alexandra Palmcrantz
- Division of Pediatric Neurology, Department of Women’s and Children’s Health, Karolinska Institutet, Stockholm, Sweden
| | - Eva Pontén
- Division of Pediatric Neurology, Department of Women’s and Children’s Health, Karolinska Institutet, Stockholm, Sweden,Department of Pediatric Orthopedic Surgery, Karolinska University Hospital, Stockholm, Sweden
| | - Jessica Pingel
- Department of Neuroscience, University of Copenhagen, Copenhagen, Denmark
| | - Ferdinand von Walden
- Division of Pediatric Neurology, Department of Women’s and Children’s Health, Karolinska Institutet, Stockholm, Sweden
| | - Rodrigo Fernandez-Gonzalo
- Division of Clinical Physiology, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden,Unit of Clinical Physiology, Karolinska University Hospital, Stockholm, Sweden,*Correspondence: Rodrigo Fernandez-Gonzalo,
| |
Collapse
|
5
|
Gonzalez A, Abrigo J, Achiardi O, Simon F, Cabello-Verrugio C. Intensive care unit-acquired weakness: From molecular mechanisms to its impact in COVID-2019. Eur J Transl Myol 2022; 32. [PMID: 36036350 PMCID: PMC9580540 DOI: 10.4081/ejtm.2022.10511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Accepted: 08/08/2022] [Indexed: 01/08/2023] Open
Abstract
Intensive Care Unit-Acquired Weakness (ICU-AW) is a generalized and symmetric neuromuscular dysfunction associated with critical illness and its treatments. Its incidence is approximately 80% in intensive care unit patients, and it manifests as critical illness polyneuropathy, critical illness myopathy, and muscle atrophy. Intensive care unit patients can lose an elevated percentage of their muscle mass in the first days after admission, producing short- and long-term sequelae that affect patients’ quality of life, physical health, and mental health. In 2019, the world was faced with coronavirus disease 2019 (COVID-19), caused by the acute respiratory syndrome coronavirus 2. COVID-19 produces severe respiratory disorders, such as acute respiratory distress syndrome, which increases the risk of developing ICU-AW. COVID-19 patients treated in intensive care units have shown early diffuse and symmetrical muscle weakness, polyneuropathy, and myalgia, coinciding with the clinical presentation of ICU-AW. Besides, these patients require prolonged intensive care unit stays, invasive mechanical ventilation, and intensive care unit pharmacological therapy, which are risk factors for ICU-AW. Thus, the purposes of this review are to discuss the features of ICU-AW and its effects on skeletal muscle. Further, we will describe the mechanisms involved in the probable development of ICU-AW in severe COVID-19 patients.
Collapse
|
6
|
Liu Q, Tang Q, Liao L, Li D, Zhu W, Zhao C. Translational therapy from preclinical animal models for muscle degeneration after rotator cuff injury. J Orthop Translat 2022; 35:13-22. [PMID: 35846726 PMCID: PMC9260436 DOI: 10.1016/j.jot.2022.03.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2021] [Revised: 02/28/2022] [Accepted: 03/01/2022] [Indexed: 11/29/2022] Open
Abstract
Chronic rotator cuff tears are debilitating diseases which significantly affect patients’ quality of life and pose substantial financial burden to the society. The intraoperative reparability of injured tendon and postoperative probability of tendon retear are highly associated with the quality of torn muscles, specifically, the severity of muscle atrophy and fatty infiltration. Animal models that reproduce the characteristic muscle pathology after rotator cuff injury have been developed and used to provide insight into the underlying biology and pathophysiology. In this review, we briefly summarize the current information obtained from preclinical animal studies regarding the degenerative change of cuff muscle subsequent to tendon release and/or suprascapular nerve denervation. Importantly, we focus on the potential translational therapeutic targets or agents for the prevention or reversal of muscle atrophy and fatty infiltration. While further studies are warranted to assess the safety and efficacy of novel therapies derived from these preclinical animal research, we believe that their clinical translation for the treatment of rotator cuff disorders is on the horizon. The Translational potential of this article Novel therapeutic strategies described in this review from preclinical animal studies hold a great translational potential for preventing or reversing rotator cuff muscle pathology, while further assessments on their safety and efficacy are warranted.
Collapse
|
7
|
Duerre DJ, Galmozzi A. Deconstructing Adipose Tissue Heterogeneity One Cell at a Time. Front Endocrinol (Lausanne) 2022; 13:847291. [PMID: 35399946 PMCID: PMC8990929 DOI: 10.3389/fendo.2022.847291] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/01/2022] [Accepted: 02/28/2022] [Indexed: 12/26/2022] Open
Abstract
As a central coordinator of physiologic metabolism, adipose tissue has long been appreciated as a highly plastic organ that dynamically responds to environmental cues. Once thought of as a homogenous storage depot, recent advances have enabled deep characterizations of the underlying structure and composition of adipose tissue depots. As the obesity and metabolic disease epidemics continue to accelerate due to modern lifestyles and an aging population, elucidation of the underlying mechanisms that control adipose and systemic homeostasis are of critical importance. Within the past decade, the emergence of deep cell profiling at tissue- and, recently, single-cell level has furthered our understanding of the complex dynamics that contribute to tissue function and their implications in disease development. Although many paradigm-shifting findings may lie ahead, profound advances have been made to forward our understanding of the adipose tissue niche in both health and disease. Now widely accepted as a highly heterogenous organ with major roles in metabolic homeostasis, endocrine signaling, and immune function, the study of adipose tissue dynamics has reached a new frontier. In this review, we will provide a synthesis of the latest advances in adipose tissue biology made possible by the use of single-cell technologies, the impact of epigenetic mechanisms on adipose function, and suggest what next steps will further our understanding of the role that adipose tissue plays in systemic physiology.
Collapse
Affiliation(s)
- Dylan J. Duerre
- Department of Medicine, University of Wisconsin-Madison, School of Medicine and Public Health, Madison, WI, United States
| | - Andrea Galmozzi
- Department of Medicine, University of Wisconsin-Madison, School of Medicine and Public Health, Madison, WI, United States
- Department of Biomolecular Chemistry, University of Wisconsin-Madison, School of Medicine and Public Health, Madison, WI, United States
- University of Wisconsin Carbone Cancer Center, University of Wisconsin-Madison, School of Medicine and Public Health, Madison, WI, United States
| |
Collapse
|
8
|
Sanchez D, Ganfornina MD. The Lipocalin Apolipoprotein D Functional Portrait: A Systematic Review. Front Physiol 2021; 12:738991. [PMID: 34690812 PMCID: PMC8530192 DOI: 10.3389/fphys.2021.738991] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Accepted: 08/30/2021] [Indexed: 12/18/2022] Open
Abstract
Apolipoprotein D is a chordate gene early originated in the Lipocalin protein family. Among other features, regulation of its expression in a wide variety of disease conditions in humans, as apparently unrelated as neurodegeneration or breast cancer, have called for attention on this gene. Also, its presence in different tissues, from blood to brain, and different subcellular locations, from HDL lipoparticles to the interior of lysosomes or the surface of extracellular vesicles, poses an interesting challenge in deciphering its physiological function: Is ApoD a moonlighting protein, serving different roles in different cellular compartments, tissues, or organisms? Or does it have a unique biochemical mechanism of action that accounts for such apparently diverse roles in different physiological situations? To answer these questions, we have performed a systematic review of all primary publications where ApoD properties have been investigated in chordates. We conclude that ApoD ligand binding in the Lipocalin pocket, combined with an antioxidant activity performed at the rim of the pocket are properties sufficient to explain ApoD association with different lipid-based structures, where its physiological function is better described as lipid-management than by long-range lipid-transport. Controlling the redox state of these lipid structures in particular subcellular locations or extracellular structures, ApoD is able to modulate an enormous array of apparently diverse processes in the organism, both in health and disease. The new picture emerging from these data should help to put the physiological role of ApoD in new contexts and to inspire well-focused future research.
Collapse
Affiliation(s)
- Diego Sanchez
- Instituto de Biologia y Genetica Molecular, Unidad de Excelencia, Universidad de Valladolid-Consejo Superior de Investigaciones Cientificas, Valladolid, Spain
| | - Maria D Ganfornina
- Instituto de Biologia y Genetica Molecular, Unidad de Excelencia, Universidad de Valladolid-Consejo Superior de Investigaciones Cientificas, Valladolid, Spain
| |
Collapse
|
9
|
Willis CRG, Gallagher IJ, Wilkinson DJ, Brook MS, Bass JJ, Phillips BE, Smith K, Etheridge T, Stokes T, McGlory C, Gorissen SHM, Szewczyk NJ, Phillips SM, Atherton PJ. Transcriptomic links to muscle mass loss and declines in cumulative muscle protein synthesis during short-term disuse in healthy younger humans. FASEB J 2021; 35:e21830. [PMID: 34342902 DOI: 10.1096/fj.202100276rr] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 07/05/2021] [Accepted: 07/19/2021] [Indexed: 12/13/2022]
Abstract
Muscle disuse leads to a rapid decline in muscle mass, with reduced muscle protein synthesis (MPS) considered the primary physiological mechanism. Here, we employed a systems biology approach to uncover molecular networks and key molecular candidates that quantitatively link to the degree of muscle atrophy and/or extent of decline in MPS during short-term disuse in humans. After consuming a bolus dose of deuterium oxide (D2 O; 3 mL.kg-1 ), eight healthy males (22 ± 2 years) underwent 4 days of unilateral lower-limb immobilization. Bilateral muscle biopsies were obtained post-intervention for RNA sequencing and D2 O-derived measurement of MPS, with thigh lean mass quantified using dual-energy X-ray absorptiometry. Application of weighted gene co-expression network analysis identified 15 distinct gene clusters ("modules") with an expression profile regulated by disuse and/or quantitatively connected to disuse-induced muscle mass or MPS changes. Module scans for candidate targets established an experimentally tractable set of candidate regulatory molecules (242 hub genes, 31 transcriptional regulators) associated with disuse-induced maladaptation, many themselves potently tied to disuse-induced reductions in muscle mass and/or MPS and, therefore, strong physiologically relevant candidates. Notably, we implicate a putative role for muscle protein breakdown-related molecular networks in impairing MPS during short-term disuse, and further establish DEPTOR (a potent mTOR inhibitor) as a critical mechanistic candidate of disuse driven MPS suppression in humans. Overall, these findings offer a strong benchmark for accelerating mechanistic understanding of short-term muscle disuse atrophy that may help expedite development of therapeutic interventions.
Collapse
Affiliation(s)
- Craig R G Willis
- Department of Sport and Health Sciences, College of Life and Environmental Sciences, University of Exeter, Exeter, UK
| | - Iain J Gallagher
- Faculty of Health Sciences and Sport, University of Stirling, Stirling, UK
| | - Daniel J Wilkinson
- MRC-Versus Arthritis Centre for Musculoskeletal Ageing Research and National Institute of Health Research, Nottingham Biomedical Research Centre, Royal Derby Hospital Centre, School of Medicine, University of Nottingham, Derby, UK
| | - Matthew S Brook
- MRC-Versus Arthritis Centre for Musculoskeletal Ageing Research and National Institute of Health Research, Nottingham Biomedical Research Centre, Royal Derby Hospital Centre, School of Medicine, University of Nottingham, Derby, UK
| | - Joseph J Bass
- MRC-Versus Arthritis Centre for Musculoskeletal Ageing Research and National Institute of Health Research, Nottingham Biomedical Research Centre, Royal Derby Hospital Centre, School of Medicine, University of Nottingham, Derby, UK
| | - Bethan E Phillips
- MRC-Versus Arthritis Centre for Musculoskeletal Ageing Research and National Institute of Health Research, Nottingham Biomedical Research Centre, Royal Derby Hospital Centre, School of Medicine, University of Nottingham, Derby, UK
| | - Kenneth Smith
- MRC-Versus Arthritis Centre for Musculoskeletal Ageing Research and National Institute of Health Research, Nottingham Biomedical Research Centre, Royal Derby Hospital Centre, School of Medicine, University of Nottingham, Derby, UK
| | - Timothy Etheridge
- Department of Sport and Health Sciences, College of Life and Environmental Sciences, University of Exeter, Exeter, UK
| | - Tanner Stokes
- Department of Kinesiology, McMaster University, Hamilton, ON, Canada
| | - Chris McGlory
- Department of Kinesiology, McMaster University, Hamilton, ON, Canada
| | | | - Nathaniel J Szewczyk
- MRC-Versus Arthritis Centre for Musculoskeletal Ageing Research and National Institute of Health Research, Nottingham Biomedical Research Centre, Royal Derby Hospital Centre, School of Medicine, University of Nottingham, Derby, UK.,Ohio Musculoskeletal and Neurological Institute (OMNI) and Department of Biomedical Sciences, Ohio University, Athens, OH, USA
| | - Stuart M Phillips
- Department of Kinesiology, McMaster University, Hamilton, ON, Canada
| | - Philip J Atherton
- MRC-Versus Arthritis Centre for Musculoskeletal Ageing Research and National Institute of Health Research, Nottingham Biomedical Research Centre, Royal Derby Hospital Centre, School of Medicine, University of Nottingham, Derby, UK
| |
Collapse
|
10
|
Desmarais F, Hervé V, Bergeron KF, Ravaut G, Perrotte M, Fyfe-Desmarais G, Rassart E, Ramassamy C, Mounier C. Cerebral Apolipoprotein D Exits the Brain and Accumulates in Peripheral Tissues. Int J Mol Sci 2021; 22:ijms22084118. [PMID: 33923459 PMCID: PMC8073497 DOI: 10.3390/ijms22084118] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Revised: 04/08/2021] [Accepted: 04/14/2021] [Indexed: 12/12/2022] Open
Abstract
Apolipoprotein D (ApoD) is a secreted lipocalin associated with neuroprotection and lipid metabolism. In rodent, the bulk of its expression occurs in the central nervous system. Despite this, ApoD has profound effects in peripheral tissues, indicating that neural ApoD may reach peripheral organs. We endeavor to determine if cerebral ApoD can reach the circulation and accumulate in peripheral tissues. Three hours was necessary for over 40% of all the radiolabeled human ApoD (hApoD), injected bilaterally, to exit the central nervous system (CNS). Once in circulation, hApoD accumulates mostly in the kidneys/urine, liver, and muscles. Accumulation specificity of hApoD in these tissues was strongly correlated with the expression of lowly glycosylated basigin (BSG, CD147). hApoD was observed to pass through bEnd.3 blood brain barrier endothelial cells monolayers. However, cyclophilin A did not impact hApoD internalization rates in bEnd.3, indicating that ApoD exit from the brain is either independent of BSG or relies on additional cell types. Overall, our data showed that ApoD can quickly and efficiently exit the CNS and reach the liver and kidneys/urine, organs linked to the recycling and excretion of lipids and toxins. This indicated that cerebral overexpression during neurodegenerative episodes may serve to evacuate neurotoxic ApoD ligands from the CNS.
Collapse
Affiliation(s)
- Frederik Desmarais
- Laboratoire du Métabolisme Moléculaire des Lipides, Centre de Recherches CERMO-FC, Département des Sciences Biologiques, Université du Québec à Montréal (UQAM), 141 av. du Président-Kennedy, Montréal, QC H2X 1Y4, Canada; (F.D.); (K.F.B.); (G.R.); (G.F.-D.)
- Laboratoire de Biologie Moléculaire, Département des Sciences Biologiques, Université du Québec à Montréal (UQAM), 141 av. du Président-Kennedy, Montréal, QC H2X 1Y4, Canada; (V.H.); (E.R.)
| | - Vincent Hervé
- Laboratoire de Biologie Moléculaire, Département des Sciences Biologiques, Université du Québec à Montréal (UQAM), 141 av. du Président-Kennedy, Montréal, QC H2X 1Y4, Canada; (V.H.); (E.R.)
- Centre Armand-Frappier Santé Biotechnologie, Institut National de la Recherche Scientifique (INRS), 531 boul. des Prairies, Laval, QC H7V 1B7, Canada;
| | - Karl F. Bergeron
- Laboratoire du Métabolisme Moléculaire des Lipides, Centre de Recherches CERMO-FC, Département des Sciences Biologiques, Université du Québec à Montréal (UQAM), 141 av. du Président-Kennedy, Montréal, QC H2X 1Y4, Canada; (F.D.); (K.F.B.); (G.R.); (G.F.-D.)
| | - Gaétan Ravaut
- Laboratoire du Métabolisme Moléculaire des Lipides, Centre de Recherches CERMO-FC, Département des Sciences Biologiques, Université du Québec à Montréal (UQAM), 141 av. du Président-Kennedy, Montréal, QC H2X 1Y4, Canada; (F.D.); (K.F.B.); (G.R.); (G.F.-D.)
| | - Morgane Perrotte
- Centre Armand-Frappier Santé Biotechnologie, Institut National de la Recherche Scientifique (INRS), 531 boul. des Prairies, Laval, QC H7V 1B7, Canada;
| | - Guillaume Fyfe-Desmarais
- Laboratoire du Métabolisme Moléculaire des Lipides, Centre de Recherches CERMO-FC, Département des Sciences Biologiques, Université du Québec à Montréal (UQAM), 141 av. du Président-Kennedy, Montréal, QC H2X 1Y4, Canada; (F.D.); (K.F.B.); (G.R.); (G.F.-D.)
- Laboratoire de Biologie Moléculaire, Département des Sciences Biologiques, Université du Québec à Montréal (UQAM), 141 av. du Président-Kennedy, Montréal, QC H2X 1Y4, Canada; (V.H.); (E.R.)
| | - Eric Rassart
- Laboratoire de Biologie Moléculaire, Département des Sciences Biologiques, Université du Québec à Montréal (UQAM), 141 av. du Président-Kennedy, Montréal, QC H2X 1Y4, Canada; (V.H.); (E.R.)
| | - Charles Ramassamy
- Centre Armand-Frappier Santé Biotechnologie, Institut National de la Recherche Scientifique (INRS), 531 boul. des Prairies, Laval, QC H7V 1B7, Canada;
- Correspondence: (C.R.); (C.M.)
| | - Catherine Mounier
- Laboratoire du Métabolisme Moléculaire des Lipides, Centre de Recherches CERMO-FC, Département des Sciences Biologiques, Université du Québec à Montréal (UQAM), 141 av. du Président-Kennedy, Montréal, QC H2X 1Y4, Canada; (F.D.); (K.F.B.); (G.R.); (G.F.-D.)
- Correspondence: (C.R.); (C.M.)
| |
Collapse
|
11
|
Effects of acute and chronic strength training on skeletal muscle autophagy in frail elderly men and women. Exp Gerontol 2020; 142:111122. [PMID: 33132146 DOI: 10.1016/j.exger.2020.111122] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 10/09/2020] [Accepted: 10/14/2020] [Indexed: 12/19/2022]
Abstract
Aging is associated with alterations in skeletal muscle autophagy, potentially affecting both muscle mass and quality in a negative manner. Strength training with protein supplementation has been reported to improve both muscle mass and quality in frail elderly individuals, but whether improvements are accompanied by alterations in protein quality control is not known. To address this issue, we investigated protein degradation markers in skeletal muscle biopsies (m. vastus lateralis) from twenty-four frail elderly men and women (86 ± 7 yr) after acute and chronic (10 weeks) strength training with protein supplementation (ST + PRO) or protein supplementation alone (PRO). Acute increases in mRNA expression of genes related to the ubiquitin proteasome system (MuRF-1, MUSA1), autophagy (ATG7, LC3, p62), and mitochondrial fission (DRP1) were observed after the first, but not after the last training session in ST + PRO. Acute changes in gene expression were accompanied by changes in protein levels of both LC3-I and LC3-II. Hence, the acute training-induced activation of proteasomal degradation and autophagy seems to depend on training status, with activation in the untrained, but not trained state. The ten-week training intervention did not affect basal levels of autophagy mRNAs and proteins, and neither markers of the ubiquitin-proteasome system. This suggests that a relatively short period of strength training may not be sufficient to increase the basal rate of protein degradation in frail elderly.
Collapse
|
12
|
Studies of ApoD -/- and ApoD -/-ApoE -/- mice uncover the APOD significance for retinal metabolism, function, and status of chorioretinal blood vessels. Cell Mol Life Sci 2020; 78:963-983. [PMID: 32440710 DOI: 10.1007/s00018-020-03546-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Revised: 04/15/2020] [Accepted: 05/07/2020] [Indexed: 12/26/2022]
Abstract
Apolipoprotein D (APOD) is an atypical apolipoprotein with unknown significance for retinal structure and function. Conversely, apolipoprotein E (APOE) is a typical apolipoprotein with established roles in retinal cholesterol transport. Herein, we immunolocalized APOD to the photoreceptor inner segments and conducted ophthalmic characterizations of ApoD-/- and ApoD-/-ApoE-/- mice. ApoD-/- mice had normal levels of retinal sterols but changes in the chorioretinal blood vessels and impaired retinal function. The whole-body glucose disposal was impaired in this genotype but the retinal glucose metabolism was unchanged. ApoD-/-ApoE-/- mice had altered sterol profile in the retina but apparently normal chorioretinal vasculature and function. The whole-body glucose disposal and retinal glucose utilization were enhanced in this genotype. OB-Rb, both leptin and APOD receptor, was found to be expressed in the photoreceptor inner segments and was at increased abundance in the ApoD-/- and ApoD-/-ApoE-/- retinas. Retinal levels of Glut4 and Cd36, the glucose transporter and scavenger receptor, respectively, were increased as well, thus linking APOD to retinal glucose and fatty acid metabolism and suggesting the APOD-OB-Rb-GLUT4/CD36 axis. In vivo isotopic labeling, transmission electron microscopy, and retinal proteomics provided additional insights into the mechanism underlying the retinal phenotypes of ApoD-/- and ApoD-/-ApoE-/- mice. Collectively, our data suggest that the APOD roles in the retina are context specific and could determine retinal glucose fluxes into different pathways. APOD and APOE do not play redundant, complementary or opposing roles in the retina, rather their interplay is more complex and reflects retinal responses elicited by lack of these apolipoproteins.
Collapse
|
13
|
Flück M, Fitze D, Ruoss S, Valdivieso P, von Rechenberg B, Bratus-Neuenschwander A, Opitz L, Hu J, Laczko E, Wieser K, Gerber C. Down-Regulation of Mitochondrial Metabolism after Tendon Release Primes Lipid Accumulation in Rotator Cuff Muscle. THE AMERICAN JOURNAL OF PATHOLOGY 2020; 190:1513-1529. [PMID: 32305353 DOI: 10.1016/j.ajpath.2020.03.019] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Revised: 02/14/2020] [Accepted: 03/26/2020] [Indexed: 12/12/2022]
Abstract
Atrophy and fat accumulation are debilitating aspects of muscle diseases and are rarely prevented. Using a vertical approach combining anatomic techniques with omics methodology in a tenotomy-induced sheep model of rotator cuff disease, we tested whether mitochondrial dysfunction is implicated in muscle wasting and perturbed lipid metabolism, speculating that both can be prevented by the stimulation of β-oxidation with l-carnitine. The infraspinatus muscle lost 22% of its volume over the first 6 weeks after tenotomy before the area-percentage of lipid increased from 8% to 18% at week 16. Atrophy was associated with the down-regulation of mitochondrial transcripts and protein and a slow-to-fast shift in muscle composition. Correspondingly, amino acid levels were increased 2 weeks after tendon release, when the levels of high-energy phosphates and glycerophospholipids were lowered. l-Carnitine administration (0.9 g/kg per day) prevented atrophy over the first 2 weeks, and mitigated alterations of glutamate, glycerophospholipids, and carnitine levels in released muscle, but did not prevent the level decrease in high-energy phosphates or protein constituents of mitochondrial respiration, promoting the accumulation of longer lipids with an increasing saturation. We conclude that the early phase of infraspinatus muscle degeneration after tendon release involves the elimination of oxidative characteristics associated with an aberrant accumulation of lipid species but is largely unrelated to the prevention of atrophy with oral l-carnitine administration.
Collapse
Affiliation(s)
- Martin Flück
- Laboratory for Muscle Plasticity, Balgrist University Hospital, University of Zurich, Zurich, Switzerland.
| | - Daniel Fitze
- Laboratory for Muscle Plasticity, Balgrist University Hospital, University of Zurich, Zurich, Switzerland
| | - Severin Ruoss
- Laboratory for Muscle Plasticity, Balgrist University Hospital, University of Zurich, Zurich, Switzerland
| | - Paola Valdivieso
- Laboratory for Muscle Plasticity, Balgrist University Hospital, University of Zurich, Zurich, Switzerland
| | - Brigitte von Rechenberg
- Vetsuisse Faculty, Musculoskeletal Research Unit, Competence Center for Applied Biotechnology, University of Zurich, Zurich, Switzerland
| | | | - Lennart Opitz
- Functional Genomics Center Zurich, ETH Zurich, University of Zurich, Zurich, Switzerland
| | - Junmin Hu
- Functional Genomics Center Zurich, ETH Zurich, University of Zurich, Zurich, Switzerland
| | - Endre Laczko
- Functional Genomics Center Zurich, ETH Zurich, University of Zurich, Zurich, Switzerland
| | - Karl Wieser
- Department of Orthopedics, Balgrist University Hospital, Zurich, Switzerland
| | - Christian Gerber
- Department of Orthopedics, Balgrist University Hospital, Zurich, Switzerland
| |
Collapse
|
14
|
Fernandez‐Gonzalo R, Tesch PA, Lundberg TR, Alkner BA, Rullman E, Gustafsson T. Three months of bed rest induce a residual transcriptomic signature resilient to resistance exercise countermeasures. FASEB J 2020; 34:7958-7969. [DOI: 10.1096/fj.201902976r] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Revised: 02/19/2020] [Accepted: 03/29/2020] [Indexed: 01/01/2023]
Affiliation(s)
- Rodrigo Fernandez‐Gonzalo
- Department of Laboratory Medicine, Division of Clinical Physiology, Karolinska Institutet, and Unit of Clinical Physiology Karolinska University Hospital Stockholm Sweden
| | - Per A. Tesch
- Department of Physiology & Pharmacology Karolinska Institutet Stockholm Sweden
| | - Tommy R. Lundberg
- Department of Laboratory Medicine, Division of Clinical Physiology, Karolinska Institutet, and Unit of Clinical Physiology Karolinska University Hospital Stockholm Sweden
| | - Björn A. Alkner
- Department of Orthopaedics Region Jönköping County Eksjö Sweden
- Department of Biomedical and Clinical Sciences Linköping University Linköping Sweden
| | - Eric Rullman
- Department of Laboratory Medicine, Division of Clinical Physiology, Karolinska Institutet, and Unit of Clinical Physiology Karolinska University Hospital Stockholm Sweden
| | - Thomas Gustafsson
- Department of Laboratory Medicine, Division of Clinical Physiology, Karolinska Institutet, and Unit of Clinical Physiology Karolinska University Hospital Stockholm Sweden
| |
Collapse
|
15
|
Abstract
The incidence of muscle atrophy is increasing with each passing year, which imposes a huge burden on the quality of life of patients. It is a public health issue that causes a growing concern around the world. Exercise is one of the key strategies to prevent and treat various diseases. Appropriate exercise is conducive to compensatory muscle hypertrophy, to improve muscle strength and elasticity, and to train muscle coordination, which is also beneficial to the recovery of skeletal muscle function and the regeneration of muscle cells. Sequelae of paralysis of patients with limb dyskinesia caused by muscle atrophy will be significantly alleviated after regular exercise therapy. Furthermore, exercise therapy can slow down or even reverse muscle atrophy. This article aims to introduce the characteristics of muscle atrophy and summarize the role and mechanism of exercise in the treatment of muscle atrophy in the existing studies, in order to further explore the mechanism of exercise to protect muscle atrophy and provide protection for patients with muscular atrophy.
Collapse
Affiliation(s)
- Nana He
- Department of Cardiology, Huamei Hospital, (previously named Ningbo No. 2 Hospital), University of Chinese Academy of Sciences, Ningbo, China
- Department of Experimental Medical Science, HwaMei Hospital, University of Chinese Academy of Sciences, Ningbo, China
- Key Laboratory of Diagnosis and Treatment of Digestive System Tumors of Zhejiang Province, Ningbo, China
| | - Honghua Ye
- Department of Cardiology, Huamei Hospital, (previously named Ningbo No. 2 Hospital), University of Chinese Academy of Sciences, Ningbo, China
| |
Collapse
|
16
|
Mugahid DA, Sengul TG, You X, Wang Y, Steil L, Bergmann N, Radke MH, Ofenbauer A, Gesell-Salazar M, Balogh A, Kempa S, Tursun B, Robbins CT, Völker U, Chen W, Nelson L, Gotthardt M. Proteomic and Transcriptomic Changes in Hibernating Grizzly Bears Reveal Metabolic and Signaling Pathways that Protect against Muscle Atrophy. Sci Rep 2019; 9:19976. [PMID: 31882638 PMCID: PMC6934745 DOI: 10.1038/s41598-019-56007-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Accepted: 12/05/2019] [Indexed: 12/31/2022] Open
Abstract
Muscle atrophy is a physiological response to disuse and malnutrition, but hibernating bears are largely resistant to this phenomenon. Unlike other mammals, they efficiently reabsorb amino acids from urine, periodically activate muscle contraction, and their adipocytes differentially responds to insulin. The contribution of myocytes to the reduced atrophy remains largely unknown. Here we show how metabolism and atrophy signaling are regulated in skeletal muscle of hibernating grizzly bear. Metabolic modeling of proteomic changes suggests an autonomous increase of non-essential amino acids (NEAA) in muscle and treatment of differentiated myoblasts with NEAA is sufficient to induce hypertrophy. Our comparison of gene expression in hibernation versus muscle atrophy identified several genes differentially regulated during hibernation, including Pdk4 and Serpinf1. Their trophic effects extend to myoblasts from non-hibernating species (including C. elegans), as documented by a knockdown approach. Together, these changes reflect evolutionary favored adaptations that, once translated to the clinics, could help improve atrophy treatment.
Collapse
Affiliation(s)
- D A Mugahid
- Neuromuscular and Cardiovascular Cell Biology, Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - T G Sengul
- Neuromuscular and Cardiovascular Cell Biology, Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - X You
- Berlin Institute for Medical Systems Biology, Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Y Wang
- Berlin Institute for Medical Systems Biology, Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - L Steil
- Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, Germany
| | - N Bergmann
- Neuromuscular and Cardiovascular Cell Biology, Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - M H Radke
- Neuromuscular and Cardiovascular Cell Biology, Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - A Ofenbauer
- Berlin Institute for Medical Systems Biology, Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - M Gesell-Salazar
- Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, Germany
| | - A Balogh
- Experimental and Clinical Research Center, Charité & Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - S Kempa
- Berlin Institute for Medical Systems Biology, Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - B Tursun
- Berlin Institute for Medical Systems Biology, Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - C T Robbins
- School of the Environment and School of Biological Sciences, Washington State University, Pullman, Washington, USA
| | - U Völker
- Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, Germany.,DZHK (German Centre for Cardiovascular Research), partner site Greifswald, Greifswald, Germany
| | - W Chen
- Berlin Institute for Medical Systems Biology, Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - L Nelson
- College of Veterinary Medicine and Department of Veterinary Clinical Science, Washington State University, Pullman, Washington, USA
| | - M Gotthardt
- Neuromuscular and Cardiovascular Cell Biology, Max Delbrück Center for Molecular Medicine, Berlin, Germany. .,Charité Universitätsmedizin Berlin, Berlin, Germany. .,DZHK (German Center for Cardiovascular Research), partner site Berlin, Berlin, Germany.
| |
Collapse
|
17
|
Anekwe DE, Biswas S, Bussières A, Spahija J. Early rehabilitation reduces the likelihood of developing intensive care unit-acquired weakness: a systematic review and meta-analysis. Physiotherapy 2019; 107:1-10. [PMID: 32135387 DOI: 10.1016/j.physio.2019.12.004] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Indexed: 10/25/2022]
Abstract
BACKGROUND Intensive care unit-acquired weakness (ICUAW) is associated with significant impairments in body structure and function, activity limitation, and participation restriction. The etiology and management of ICUAW remain uncertain. OBJECTIVE To estimate the extent to which early rehabilitation interventions (early mobilization [EM] and/or neuromuscular electrical stimulation [NMES]) compared to usual care reduce the incidence of ICUAW in critically ill patients. DATA SOURCES We searched MEDLINE, EMBASE, CINAHL, Cochrane Central and Physiotherapy Evidence Database databases from inception to May 1st, 2017. ELIGIBILITY CRITERIA Randomized controlled trials of EM and/or NMES interventions in critically ill adults. DATA EXTRACTION AND DATA SYNTHESIS Data on the incidence of ICUAW and secondary outcomes were extracted. Both odds and risk ratios for ICUAW were pooled using the random-effects model. RESULTS We identified 1421 reports after duplicate removal. Nine studies including 841 patients (419 intervention and 422 usual care) were included in the final analysis. The interventions involved EM in five trials, NMES in three trials, and both EM and NMES in one trial. Early rehabilitation decreased the likelihood of developing ICUAW: odds ratio of 0.63 (95% CI: 0.43 to 0.92) in the screened population, and 0.71 (95% CI: 0.53 to 0.95) in the randomized population. CONCLUSION, IMPLICATIONS OF KEY FINDINGS Early rehabilitation was associated with a decreased likelihood of developing ICUAW. Our findings support early rehabilitation in the ICU. While results were consistent in both the screened and randomized populations, the wide confidence intervals suggest that well-conducted trials are needed to validate our findings. SYSTEMATIC REVIEW REGISTRATION NUMBER PROSPERO registration ID: CRD42017065031.
Collapse
Affiliation(s)
- David E Anekwe
- School of Physical and Occupational Therapy, McGill University, Montreal, Quebec, Canada; Research Center, CIUSSS du Nord-de-l'Ile-de-Montréal, Sacré-Coeur Hospital, Université de Montréal, Montréal, Quebec, Canada; Center for Interdisciplinary Research in Rehabilitation in Montreal, CISS du Nord-de-l'Île-de-Montréal, Jewish Rehabilitation Hospital, Laval, Quebec, Canada
| | - Sharmistha Biswas
- Respiratory Epidemiology and Clinical Research Unit, Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada; Epidemiology, Biostatistics and Occupational Health, McGill University, Montreal, Quebec, Canada
| | - André Bussières
- School of Physical and Occupational Therapy, McGill University, Montreal, Quebec, Canada; Center for Interdisciplinary Research in Rehabilitation in Montreal, CISS du Nord-de-l'Île-de-Montréal, Jewish Rehabilitation Hospital, Laval, Quebec, Canada
| | - Jadranka Spahija
- School of Physical and Occupational Therapy, McGill University, Montreal, Quebec, Canada; Research Center, CIUSSS du Nord-de-l'Ile-de-Montréal, Sacré-Coeur Hospital, Université de Montréal, Montréal, Quebec, Canada; Center for Interdisciplinary Research in Rehabilitation in Montreal, CISS du Nord-de-l'Île-de-Montréal, Jewish Rehabilitation Hospital, Laval, Quebec, Canada.
| |
Collapse
|
18
|
Leermakers PA, Kneppers AEM, Schols AMWJ, Kelders MCJM, de Theije CC, Verdijk LB, van Loon LJC, Langen RCJ, Gosker HR. Skeletal muscle unloading results in increased mitophagy and decreased mitochondrial biogenesis regulation. Muscle Nerve 2019; 60:769-778. [PMID: 31495926 PMCID: PMC6900132 DOI: 10.1002/mus.26702] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Revised: 09/03/2019] [Accepted: 09/03/2019] [Indexed: 12/31/2022]
Abstract
Introduction Physical inactivity significantly contributes to loss of muscle mass and performance in bed‐bound patients. Loss of skeletal muscle mitochondrial content has been well‐established in muscle unloading models, but the underlying molecular mechanism remains unclear. We hypothesized that apparent unloading‐induced loss of muscle mitochondrial content is preceded by increased mitophagy‐ and decreased mitochondrial biogenesis‐signaling during the early stages of unloading. Methods We analyzed a comprehensive set of molecular markers involved in mitochondrial‐autophagy, −biogenesis, −dynamics, and ‐content, in the gastrocnemius muscle of C57BL/6J mice subjected to 0‐ and 3‐days hind limb suspension, and in biopsies from human vastus lateralis muscle obtained before and after 7 days of one‐leg immobilization. Results In both mice and men, short‐term skeletal muscle unloading results in molecular marker patterns indicative of increased receptor‐mediated mitophagy and decreased mitochondrial biogenesis regulation, before apparent loss of mitochondrial content. Discussion These results emphasize the early‐onset of skeletal muscle disuse‐induced mitochondrial remodeling.
Collapse
Affiliation(s)
- Pieter A Leermakers
- Department of Respiratory Medicine, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Anita E M Kneppers
- Department of Respiratory Medicine, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Annemie M W J Schols
- Department of Respiratory Medicine, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Marco C J M Kelders
- Department of Respiratory Medicine, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Chiel C de Theije
- Department of Respiratory Medicine, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Lex B Verdijk
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - 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
| | - Ramon C J Langen
- Department of Respiratory Medicine, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Harry R Gosker
- Department of Respiratory Medicine, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre+, Maastricht, The Netherlands
| |
Collapse
|
19
|
Abstract
Skeletal muscle atrophy is a common side effect of most human diseases. Muscle loss is not only detrimental for the quality of life but it also dramatically impairs physiological processes of the organism and decreases the efficiency of medical treatments. While hypothesized for years, the existence of an atrophying programme common to all pathologies is still incompletely solved despite the discovery of several actors and key regulators of muscle atrophy. More than a decade ago, the discovery of a set of genes, whose expression at the mRNA levels were similarly altered in different catabolic situations, opened the way of a new concept: the presence of atrogenes, i.e. atrophy-related genes. Importantly, the atrogenes are referred as such on the basis of their mRNA content in atrophying muscles, the regulation at the protein level being sometimes more complicate to elucidate. It should be noticed that the atrogenes are markers of atrophy and that their implication as active inducers of atrophy is still an open question for most of them. While the atrogene family has grown over the years, it has mostly been incremented based on data coming from rodent models. Whether the rodent atrogenes are valid for humans still remain to be established. An "atrogene" was originally defined as a gene systematically up- or down-regulated in several catabolic situations. Even if recent works often restrict this notion to the up-regulation of a limited number of proteolytic enzymes, it is important to keep in mind the big picture view. In this review, we provide an update of the validated and potential rodent atrogenes and the metabolic pathways they belong, and based on recent work, their relevance in human physio-pathological situations. We also propose a more precise definition of the atrogenes that integrates rapid recovery when catabolic stimuli are stopped or replaced by anabolic ones.
Collapse
Affiliation(s)
- Daniel Taillandier
- Université Clermont Auvergne, INRA, UNH, Unité de Nutrition Humaine, CRNH Auvergne, F-63000, Clermont-Ferrand, France.
| | - Cécile Polge
- Université Clermont Auvergne, INRA, UNH, Unité de Nutrition Humaine, CRNH Auvergne, F-63000, Clermont-Ferrand, France
| |
Collapse
|
20
|
Buso A, Comelli M, Picco R, Isola M, Magnesa B, Pišot R, Rittweger J, Salvadego D, Šimunič B, Grassi B, Mavelli I. Mitochondrial Adaptations in Elderly and Young Men Skeletal Muscle Following 2 Weeks of Bed Rest and Rehabilitation. Front Physiol 2019; 10:474. [PMID: 31118897 PMCID: PMC6504794 DOI: 10.3389/fphys.2019.00474] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Accepted: 04/04/2019] [Indexed: 12/21/2022] Open
Abstract
The aim of the study was to evaluate the expression levels of proteins related to mitochondrial biogenesis regulation and bioenergetics in vastus lateralis muscle biopsies from 16 elderly and 7 young people subjected to 14 days of bed-rest, causing atrophy, and subsequent 14 days of exercise training. Based on quantitative immunoblot analyses, in both groups a reduction of two key regulators of mitochondrial biogenesis/remodeling and activity, namely PGC-1α and Sirt3, was revealed during bed-rest, with a subsequent up-regulation after rehabilitation, indicating an involvement of PGC-1α-Sirt3 axis in response to the treatments. A difference was observed comparing the young and elderly subjects as, for both proteins, the abundance in the elderly was more affected by immobility and less responsive to exercise. The expression levels of TOM20 and Citrate Synthase, assayed as markers of outer mitochondrial membrane and mitochondrial mass, showed a noticeable sensitivity in the elderly group, where they were affected by bed-rest and rehabilitation recalling the pattern of PGC-1α. TOM20 and CS remained unchanged in young subjects. Single OXPHOS complexes showed peculiar patterns, which were in some cases dissimilar from PGC-1α, and suggest different influences on protein biogenesis and degradation. Overall, exercise was capable to counteract the effect of immobility, when present, except for complex V, which was markedly downregulated by bed-rest, but remained unaffected after rehabilitation, maybe as result of greater extent of degradation processes over biogenesis. Phosphorylation extent of AMPK, and its upstream activator LKB1, did not change after bed-rest and rehabilitation in either young or elderly subjects, suggesting that the activation of energy-sensing LKB1-AMPK signaling pathway was “missed” due to its transient nature, or was not triggered under our conditions. Our study demonstrates that, as far as the expression of various proteins related to mitochondrial biogenesis/remodeling, adaptations to bed-rest and rehabilitation in the two populations were different. The impact of bed-rest was greater in the elderly subjects, where the pattern (decrease after bed rest and recovery following rehabilitation) was accompanied by changes of mitochondrial mass. Modifications of protein abundance were matched with data obtained from gene expression analyses of four public human datasets focusing on related genes.
Collapse
Affiliation(s)
- Alessia Buso
- Department of Medicine, University of Udine, Udine, Italy
| | - Marina Comelli
- Department of Medicine, University of Udine, Udine, Italy
| | | | - Miriam Isola
- Department of Medicine, University of Udine, Udine, Italy
| | | | - Rado Pišot
- Institute for Kinesiology Research, Science and Research Centre, Koper, Slovenia
| | - Joern Rittweger
- Department of Pediatrics and Adolescent Medicine, University of Cologne, Cologne, Germany.,Institute of Aerospace Medicine, German Aerospace Center (DLR), Cologne, Germany
| | - Desy Salvadego
- Department of Medicine, University of Udine, Udine, Italy
| | - Boštjan Šimunič
- Institute for Kinesiology Research, Science and Research Centre, Koper, Slovenia
| | - Bruno Grassi
- Department of Medicine, University of Udine, Udine, Italy.,Institute of Bioimaging and Molecular Physiology, National Research Council, Milan, Italy
| | - Irene Mavelli
- Department of Medicine, University of Udine, Udine, Italy.,INBB Istituto Nazionale Biostrutture e Biosistemi, Rome, Italy
| |
Collapse
|
21
|
Hyatt H, Deminice R, Yoshihara T, Powers SK. Mitochondrial dysfunction induces muscle atrophy during prolonged inactivity: A review of the causes and effects. Arch Biochem Biophys 2018; 662:49-60. [PMID: 30452895 DOI: 10.1016/j.abb.2018.11.005] [Citation(s) in RCA: 117] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2018] [Accepted: 11/05/2018] [Indexed: 02/08/2023]
Abstract
Prolonged skeletal muscle inactivity (e.g. limb immobilization, bed rest, mechanical ventilation, spinal cord injury, etc.) results in muscle atrophy that manifests into a decreased quality of life and in select patient populations, a higher risk of morbidity and mortality. Thus, understanding the processes that contribute to muscle atrophy during prolonged periods of muscle disuse is an important area of research. In this regard, mitochondrial dysfunction has been directly linked to the muscle wasting that occurs during extended periods of skeletal muscle inactivity. While the concept that mitochondrial dysfunction contributes to disuse muscle atrophy has been contemplated for nearly 50 years, the mechanisms connecting mitochondrial signaling events to skeletal muscle atrophy remained largely unexplained until recently. Indeed, emerging evidence reveals that mitochondrial dysfunction and the associated mitochondrial signaling events are a requirement for several forms of inactivity-induced skeletal muscle atrophy. Specifically, inactivity-induced alterations in skeletal muscle mitochondria phenotype and increased ROS emission, impaired Ca2+ handling, and release of mitochondria-specific proteolytic activators are established occurrences that promote fiber atrophy during prolonged periods of muscle inactivity. This review highlights the evidence that directly connects mitochondrial dysfunction and aberrant mitochondrial signaling with skeletal muscle atrophy and discusses the mechanisms linking these interconnected phenomena.
Collapse
Affiliation(s)
- Hayden Hyatt
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, FL, USA.
| | - Rafael Deminice
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, FL, USA; Department of Physical Education, University of Estadual of Londrina, Londrina, Brazil
| | - Toshinori Yoshihara
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, FL, USA; Department of Exercise Physiology, Juntendo University, Tokyo, Japan
| | - Scott K Powers
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, FL, USA
| |
Collapse
|
22
|
Rullman E, Fernandez-Gonzalo R, Mekjavić IB, Gustafsson T, Eiken O. MEF2 as upstream regulator of the transcriptome signature in human skeletal muscle during unloading. Am J Physiol Regul Integr Comp Physiol 2018; 315:R799-R809. [PMID: 29995456 DOI: 10.1152/ajpregu.00452.2017] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Our understanding of skeletal muscle structural and functional alterations during unloading has increased in recent decades, yet the molecular mechanisms underpinning these changes have only started to be unraveled. The purpose of the current investigation was to assess changes in skeletal muscle gene expression after 21 days of bed rest, with a particular focus on predicting upstream regulators of muscle disuse. Additionally, the association between differential microRNA expression and the transcriptome signature of bed rest were investigated. mRNAs from musculus vastus lateralis biopsies obtained from 12 men before and after the bed rest were analyzed using a microarray. There were 54 significantly upregulated probesets after bed rest, whereas 103 probesets were downregulated (false discovery rate 10%; fold-change cutoff ≥1.5). Among the upregulated genes, transcripts related to denervation-induced alterations in skeletal muscle were identified, e.g., acetylcholine receptor subunit delta and perinatal myosin. The most downregulated transcripts were functionally enriched for mitochondrial genes and genes involved in mitochondrial biogenesis, followed by a large number of contractile fiber components. Upstream regulator analysis identified a robust inhibition of the myocyte enhancer factor-2 (MEF2) family, in particular MEF2C, which was suggested to act upstream of several key downregulated genes, most notably peroxisome proliferator-activated receptor γ coactivator 1-α (PGC-1α)/peroxisome proliferator-activated receptors (PPARs) and CRSP3. Only a few microRNAs were identified as playing a role in the overall transcriptome picture induced by sustained bed rest. Our results suggest that the MEF2 family is a key regulator underlying the transcriptional signature of bed rest and, hence, ultimately also skeletal muscle alterations induced by systemic unloading in humans.
Collapse
Affiliation(s)
- Eric Rullman
- Department of Laboratory Medicine, Clinical Physiology, Karolinska Institutet and Karolinska University Hospital , Stockholm , Sweden.,Department of Cardiology, Karolinska University Hospital , Stockholm , Sweden
| | - Rodrigo Fernandez-Gonzalo
- Department of Laboratory Medicine, Clinical Physiology, Karolinska Institutet and Karolinska University Hospital , Stockholm , Sweden
| | - Igor B Mekjavić
- Department of Automation, Biocybernetics and Robotics, Jozef Stefan Institute , Ljubljana , Slovenia
| | - Thomas Gustafsson
- Department of Laboratory Medicine, Clinical Physiology, Karolinska Institutet and Karolinska University Hospital , Stockholm , Sweden
| | - Ola Eiken
- Department of Environmental Physiology, Swedish Aerospace Physiology Centre, KTH Royal Institute of Technology , Stockholm , Sweden
| |
Collapse
|
23
|
Jung J, Kwon M, Bae S, Yim S, Lee D. Petri net-based prediction of therapeutic targets that recover abnormally phosphorylated proteins in muscle atrophy. BMC SYSTEMS BIOLOGY 2018; 12:26. [PMID: 29506508 PMCID: PMC5838966 DOI: 10.1186/s12918-018-0555-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Accepted: 02/21/2018] [Indexed: 11/23/2022]
Abstract
Background Muscle atrophy, an involuntary loss of muscle mass, is involved in various diseases and sometimes leads to mortality. However, therapeutics for muscle atrophy thus far have had limited effects. Here, we present a new approach for therapeutic target prediction using Petri net simulation of the status of phosphorylation, with a reasonable assumption that the recovery of abnormally phosphorylated proteins can be a treatment for muscle atrophy. Results The Petri net model was employed to simulate phosphorylation status in three states, i.e. reference, atrophic and each gene-inhibited state based on the myocyte-specific phosphorylation network. Here, we newly devised a phosphorylation specific Petri net that involves two types of transitions (phosphorylation or de-phosphorylation) and two types of places (activation with or without phosphorylation). Before predicting therapeutic targets, the simulation results in reference and atrophic states were validated by Western blotting experiments detecting five marker proteins, i.e. RELA, SMAD2, SMAD3, FOXO1 and FOXO3. Finally, we determined 37 potential therapeutic targets whose inhibition recovers the phosphorylation status from an atrophic state as indicated by the five validated marker proteins. In the evaluation, we confirmed that the 37 potential targets were enriched for muscle atrophy-related terms such as actin and muscle contraction processes, and they were also significantly overlapping with the genes associated with muscle atrophy reported in the Comparative Toxicogenomics Database (p-value < 0.05). Furthermore, we noticed that they included several proteins that could not be characterized by the shortest path analysis. The three potential targets, i.e. BMPR1B, ROCK, and LEPR, were manually validated with the literature. Conclusions In this study, we suggest a new approach to predict potential therapeutic targets of muscle atrophy with an analysis of phosphorylation status simulated by Petri net. We generated a list of the potential therapeutic targets whose inhibition recovers abnormally phosphorylated proteins in an atrophic state. They were evaluated by various approaches, such as Western blotting, GO terms, literature, known muscle atrophy-related genes and shortest path analysis. We expect the new proposed strategy to provide an understanding of phosphorylation status in muscle atrophy and to provide assistance towards identifying new therapies. Electronic supplementary material The online version of this article (10.1186/s12918-018-0555-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Jinmyung Jung
- Bio-Synergy Research Center, 291 Daehak-ro, Yuseong-gu, 305-701, Daejeon, Republic of Korea.,Department of Applied Statistics, College of Economics and Business, The University of Suwon, Hwaseong-si, Gyeonggi-do, 18323, Republic of Korea
| | - Mijin Kwon
- Department of Bio and Brain Engineering, KAIST, 291 Daehak-ro, Yuseong-gu, Daejeon, 305-701, Republic of Korea
| | - Sunghwa Bae
- Department of Bio and Brain Engineering, KAIST, 291 Daehak-ro, Yuseong-gu, Daejeon, 305-701, Republic of Korea
| | - Soorin Yim
- Department of Bio and Brain Engineering, KAIST, 291 Daehak-ro, Yuseong-gu, Daejeon, 305-701, Republic of Korea
| | - Doheon Lee
- Bio-Synergy Research Center, 291 Daehak-ro, Yuseong-gu, 305-701, Daejeon, Republic of Korea. .,Department of Bio and Brain Engineering, KAIST, 291 Daehak-ro, Yuseong-gu, Daejeon, 305-701, Republic of Korea.
| |
Collapse
|
24
|
Exercise Altered the Skeletal Muscle MicroRNAs and Gene Expression Profiles in Burn Rats With Hindlimb Unloading. J Burn Care Res 2018; 38:11-19. [PMID: 27753701 DOI: 10.1097/bcr.0000000000000444] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
This study investigated microRNA and target gene profiles under different conditions of burn, bed rest, and exercise training. Male Sprague-Dawley rats (n = 48) were assigned to sham ambulatory, sham hindlimb unloading, burn ambulatory, or burn plus hindlimb unloading groups. Rats received a 40% TBSA scald burn or sham treatments and were ambulatory or hindlimb unloaded. Rats were further assigned to exercise or no exercise. Plantaris tissues were harvested on day 14 and pooled to analyze for microRNA and gene expression profiles. Compared with the sham ambulatory-no exercise group, 73, 79, and 80 microRNAs were altered 2-fold in the burn ambulatory, sham hindlimb unloading, and burn hindlimb unloading groups, all with no exercise, respectively. More than 70% of microRNAs were upregulated in response to burn and hindlimb unloading, whereas 60% microRNA of the profile decreased in hindlimb unloaded burn rats with exercise training. MiR-182 was the most affected in rat muscle. Gene ontology biological process and pathway analysis showed that the oxidative stress pathway was most stimulated in the hindlimb unloaded burn rats; while in response to exercise training, all genes in related pathways such as hypermetabolic, inflammation, and blood coagulation were alleviated. MicroRNAs and transcript gene profiles were altered in burn and hindlimb unloading groups, with additive effects on hindlimb unloaded burn rats. The altered genes' signal pathways were associated with muscle mass loss and function impairment. Muscle improvement with exercise training was observed in gene levels with microRNA alterations as well.
Collapse
|
25
|
Perez-Suarez I, Ponce-González JG, de La Calle-Herrero J, Losa-Reyna J, Martin-Rincon M, Morales-Alamo D, Santana A, Holmberg HC, Calbet JAL. Severe energy deficit upregulates leptin receptors, leptin signaling, and PTP1B in human skeletal muscle. J Appl Physiol (1985) 2017; 123:1276-1287. [DOI: 10.1152/japplphysiol.00454.2017] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Revised: 06/22/2017] [Accepted: 07/19/2017] [Indexed: 11/22/2022] Open
Abstract
In obesity, leptin receptors (OBR) and leptin signaling in skeletal muscle are downregulated. To determine whether OBR and leptin signaling are upregulated with a severe energy deficit, 15 overweight men were assessed before the intervention (PRE), after 4 days of caloric restriction (3.2 kcal·kg body wt−1·day−1) in combination with prolonged exercise (CRE; 8 h walking + 45 min single-arm cranking/day) to induce an energy deficit of ~5,500 kcal/day, and following 3 days of control diet (isoenergetic) and reduced exercise (CD). During CRE, the diet consisted solely of whey protein ( n = 8) or sucrose ( n = 7; 0.8 g·kg body wt−1·day−1). Muscle biopsies were obtained from the exercised and the nonexercised deltoid muscles and from the vastus lateralis. From PRE to CRE, serum glucose, insulin, and leptin were reduced. OBR expression was augmented in all examined muscles associated with increased maximal fat oxidation. Compared with PRE, after CD, phospho-Tyr1141OBR, phospho-Tyr985OBR, JAK2, and phospho-Tyr1007/1008JAK2 protein expression were increased in all muscles, whereas STAT3 and phospho-Tyr705STAT3 were increased only in the arms. The expression of protein tyrosine phosphatase 1B (PTP1B) in skeletal muscle was increased by 18 and 45% after CRE and CD, respectively ( P < 0.05). Suppressor of cytokine signaling 3 (SOCS3) tended to increase in the legs and decrease in the arm muscles (ANOVA interaction: P < 0.05). Myosin heavy chain I isoform was associated with OBR protein expression ( r = −0.75), phospho-Tyr985OBR ( r = 0.88), and phospho-Tyr705STAT3/STAT3 ( r = 0.74). In summary, despite increased PTP1B expression, skeletal muscle OBR and signaling are upregulated by a severe energy deficit with greater response in the arm than in the legs likely due to SOCS3 upregulation in the leg muscles. NEW & NOTEWORTHY This study shows that the skeletal muscle leptin receptors and their corresponding signaling cascade are upregulated in response to a severe energy deficit, contributing to increase maximal fat oxidation. The responses are more prominent in the arm muscles than in the legs but partly blunted by whey protein ingestion and high volume of exercise. This occurs despite an increase of protein tyrosine phosphatase 1B protein expression, a known inhibitor of insulin and leptin signaling.
Collapse
Affiliation(s)
- Ismael Perez-Suarez
- Department of Physical Education, University of Las Palmas de Gran Canaria, Las Palmas de Gran Canaria, Spain
- Research Institute of Biomedical and Health Sciences, Las Palmas de Gran Canaria, Spain; and
| | | | - Jaime de La Calle-Herrero
- Department of Physical Education, University of Las Palmas de Gran Canaria, Las Palmas de Gran Canaria, Spain
| | - Jose Losa-Reyna
- Department of Physical Education, University of Las Palmas de Gran Canaria, Las Palmas de Gran Canaria, Spain
| | - Marcos Martin-Rincon
- Department of Physical Education, University of Las Palmas de Gran Canaria, Las Palmas de Gran Canaria, Spain
- Research Institute of Biomedical and Health Sciences, Las Palmas de Gran Canaria, Spain; and
| | - David Morales-Alamo
- Department of Physical Education, University of Las Palmas de Gran Canaria, Las Palmas de Gran Canaria, Spain
- Research Institute of Biomedical and Health Sciences, Las Palmas de Gran Canaria, Spain; and
| | - Alfredo Santana
- Clinical Genetics Unit, Complejo Hospitalario Universitario Insular-Materno Infantil de Las Palmas de Gran Canaria, Las Palmas de Gran Canaria, Spain
- Research Institute of Biomedical and Health Sciences, Las Palmas de Gran Canaria, Spain; and
| | - Hans-Christer Holmberg
- Swedish Winter Sports Research Centre, Department of Health Sciences, Mid Sweden University, Östersund, Sweden
| | - Jose A. L. Calbet
- Department of Physical Education, University of Las Palmas de Gran Canaria, Las Palmas de Gran Canaria, Spain
- Research Institute of Biomedical and Health Sciences, Las Palmas de Gran Canaria, Spain; and
| |
Collapse
|
26
|
Dirks ML, Wall BT, van Loon LJC. Interventional strategies to combat muscle disuse atrophy in humans: focus on neuromuscular electrical stimulation and dietary protein. J Appl Physiol (1985) 2017; 125:850-861. [PMID: 28970205 DOI: 10.1152/japplphysiol.00985.2016] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Numerous situations, such as the recovery from illness or rehabilitation after injury, necessitate a period of muscle disuse in otherwise healthy individuals. Even a few days of immobilization or bed rest can lead to substantial loss of skeletal muscle tissue and compromise metabolic health. The decline in muscle mass is attributed largely to a decline in postabsorptive and postprandial muscle protein synthesis rates. Reintroduction of some level of muscle contraction by the application of neuromuscular electrical stimulation (NMES) can augment both postabsorptive and postprandial muscle protein synthesis rates and, as such, prevent or attenuate muscle loss during short-term disuse in various clinical populations. Whereas maintenance of habitual dietary protein consumption is a prerequisite for muscle mass maintenance, supplementing dietary protein above habitual intake levels does not prevent muscle loss during disuse in otherwise healthy humans. Combining the anabolic properties of physical activity (or surrogates) with appropriate nutritional support likely further increases the capacity to preserve skeletal muscle mass during a period of disuse. Therefore, effective interventional strategies to prevent or alleviate muscle disuse atrophy should include both exercise (mimetics) and appropriate nutritional support.
Collapse
Affiliation(s)
- Marlou L Dirks
- NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre+, Maastricht , The Netherlands
| | - Benjamin T Wall
- NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre+, Maastricht , The Netherlands
| | - Luc J C van Loon
- NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre+, Maastricht , The Netherlands
| |
Collapse
|
27
|
Herzig KH, Leppäluoto J, Jokelainen J, Meugnier E, Pesenti S, Selänne H, Mäkelä KA, Ahola R, Jämsä T, Vidal H, Keinänen-Kiukaanniemi S. Low level activity thresholds for changes in NMR biomarkers and genes in high risk subjects for Type 2 Diabetes. Sci Rep 2017; 7:11267. [PMID: 28924247 PMCID: PMC5603534 DOI: 10.1038/s41598-017-09753-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2017] [Accepted: 07/28/2017] [Indexed: 01/26/2023] Open
Abstract
Our objectives were to determine if there are quantitative associations between amounts and intensities of physical activities (PA) on NMR biomarkers and changes in skeletal muscle gene expressions in subjects with high risk for type 2 diabetes (T2D) performing a 3-month PA intervention. We found that PA was associated with beneficial biomarker changes in a factor containing several VLDL and HDL subclasses and lipids in principal component analysis (P = <0.01). Division of PA into quartiles demonstrated significant changes in NMR biomarkers in the 2nd - 4th quartiles compared to the 1st quartile representing PA of less than 2850 daily steps (P = 0.0036). Mediation analysis of PA-related reductions in lipoproteins showed that the effects of PA was 4-15 times greater than those of body weight or fat mass reductions. In a subset study in highly active subjects' gene expressions of oxidative fiber markers, Apo D, and G0/G1 Switch Gene 2, controlling insulin signaling and glucose metabolism were significantly increased. Slow walking at speeds of 2-3 km/h exceeding 2895 steps/day attenuated several circulating lipoprotein lipids. The effects were mediated rather by PA than body weight or fat loss. Thus, lower thresholds for PA may exist for long term prevention of cardio-metabolic diseases in sedentary overweight subjects.
Collapse
Affiliation(s)
- Karl-Heinz Herzig
- Research Unit of Biomedicine, and Biocenter of Oulu, Oulu University, 90014, Oulu, Finland. .,Department of Gastroenterology and Metabolism, Poznan University of Medical Sciences, Poznan, Poland. .,Medical Research Center and Oulu University Hospital, University of Oulu and Oulu University Hospital, Oulu, Finland.
| | - Juhani Leppäluoto
- Research Unit of Biomedicine, and Biocenter of Oulu, Oulu University, 90014, Oulu, Finland
| | - Jari Jokelainen
- Center for Life Course Health Research, Faculty of Medicine, University of Oulu, 90014, Oulu, Finland.,Oulu University Hospital, Unit of General Practice, and Health Center of Oulu, Oulu, Finland
| | - Emmanuelle Meugnier
- CarMeN Laboratory, INSERM U1060, INRA U1397, University of Lyon, 69600, Oullins, France
| | - Sandra Pesenti
- CarMeN Laboratory, INSERM U1060, INRA U1397, University of Lyon, 69600, Oullins, France
| | - Harri Selänne
- Department of Education and Psychology, University of Jyväskylä, Jyväskylä, Finland
| | - Kari A Mäkelä
- Research Unit of Biomedicine, and Biocenter of Oulu, Oulu University, 90014, Oulu, Finland
| | - Riikka Ahola
- Research Unit of Medical Imaging, Physics and Technology, University of Oulu, 90014, Oulu, Finland
| | - Timo Jämsä
- Medical Research Center and Oulu University Hospital, University of Oulu and Oulu University Hospital, Oulu, Finland.,Research Unit of Medical Imaging, Physics and Technology, University of Oulu, 90014, Oulu, Finland.,Department of Diagnostic Imaging, Oulu University Hospital, Oulu, Finland
| | - Hubert Vidal
- CarMeN Laboratory, INSERM U1060, INRA U1397, University of Lyon, 69600, Oullins, France
| | - Sirkka Keinänen-Kiukaanniemi
- Center for Life Course Health Research, Faculty of Medicine, University of Oulu, 90014, Oulu, Finland.,Oulu University Hospital, Unit of General Practice, and Health Center of Oulu, Oulu, Finland
| |
Collapse
|
28
|
Chen YW, Gregory C, Ye F, Harafuji N, Lott D, Lai SH, Mathur S, Scarborough M, Gibbs P, Baligand C, Vandenborne K. Molecular signatures of differential responses to exercise trainings during rehabilitation. ACTA ACUST UNITED AC 2017; 2. [PMID: 28845464 PMCID: PMC5568829 DOI: 10.15761/bgg.1000127] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
The loss and recovery of muscle mass and function following injury and during rehabilitation varies among individuals. While recent expression profiling studies have illustrated transcriptomic responses to muscle disuse and remodeling, how these changes contribute to the physiological responses are not clear. In this study, we quantified the effects of immobilization and subsequent rehabilitation training on muscle size and identified molecular pathways associated with muscle responsiveness in an orthopaedic patient cohort study. The injured leg of 16 individuals with ankle injury was immobilized for a minimum of 4 weeks, followed by a 6-week rehabilitation program. The maximal cross-sectional area (CSA) of the medial gastrocnemius muscle of the immobilized and control legs were determined by T1-weighted axial MRI images. Genome-wide mRNA profiling data were used to identify molecular signatures that distinguish the patients who responded to immobilization and rehabilitation and those who were considered minimal responders. RESULTS: Using 6% change as the threshold to define responsiveness, a greater degree of changes in muscle size was noted in high responders (−14.9 ± 3.6%) compared to low responders (0.1 ± 0.0%) during immobilization. In addition, a greater degree of changes in muscle size was observed in high responders (20.5 ± 3.2%) compared to low responders (2.5 ± 0.9%) at 6-week rehabilitation. Microarray analysis showed a higher number of genes differentially expressed in the responders compared to low responders in general; with more expression changes observed at the acute stage of rehabilitation in both groups. Pathways analysis revealed top molecular pathways differentially affected in the groups, including genes involved in mitochondrial function, protein turn over, integrin signaling and inflammation. This study confirmed the extent of muscle atrophy due to immobilization and recovery by exercise training is associated with distinct remodeling signature, which can potentially be used for evaluating and predicting clinical outcomes.
Collapse
Affiliation(s)
- Yi-Wen Chen
- Research Center for Genetic Medicine, Children's National Medical Center, Washington DC, USA.,Department of Integrative Systems Biology, George Washington University, Washington DC, USA
| | - Chris Gregory
- Department of Health Sciences and Research, Medical University of South Carolina, Charleston, SC, USA
| | - Fan Ye
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, FL, USA
| | - Naoe Harafuji
- Research Center for Genetic Medicine, Children's National Medical Center, Washington DC, USA
| | - Donovan Lott
- Department of Physical Therapy, University of Florida, Gainesville, FL, USA
| | - San-Huei Lai
- Research Center for Genetic Medicine, Children's National Medical Center, Washington DC, USA
| | - Sunita Mathur
- Department of Physical Therapy, University of Toronto, Toronto, Ontario, USA
| | - Mark Scarborough
- Department of Orthopaedics and Rehabilitation, University of Florida, Gainesville, FL, USA
| | - Parker Gibbs
- Department of Orthopaedics and Rehabilitation, University of Florida, Gainesville, FL, USA
| | - Celine Baligand
- Department of Physiology and Functional Genomics, University of Florida, Gainesville, FL, USA
| | - Krista Vandenborne
- Department of Physical Therapy, University of Florida, Gainesville, FL, USA
| |
Collapse
|
29
|
Hamrick MW. Role of the Cytokine-like Hormone Leptin in Muscle-bone Crosstalk with Aging. J Bone Metab 2017; 24:1-8. [PMID: 28326295 PMCID: PMC5357607 DOI: 10.11005/jbm.2017.24.1.1] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2016] [Accepted: 12/20/2016] [Indexed: 12/14/2022] Open
Abstract
The cytokine-like hormone leptin is a classic adipokine that is secreted by adipocytes, increases with weight gain, and decreases with weight loss. Additional studies have, however, shown that leptin is also produced by skeletal muscle, and leptin receptors are abundant in both skeletal muscle and bone-derived mesenchymal (stromal) stem cells. These findings suggest that leptin may play an important role in muscle-bone crosstalk. Leptin treatment in vitro increases the expression of myogenic genes in primary myoblasts, and leptin treatment in vivo increases the expression of microRNAs involved in myogenesis. Bone marrow adipogenesis is associated with low bone mass in humans and rodents, and leptin can reduce marrow adipogenesis centrally through its receptors in the hypothalamus as well as directly via its receptors in bone marrow stem cells. Yet, central leptin resistance can increase with age, and low circulating levels of leptin have been observed among the frail elderly. Thus, aging appears to significantly alter leptin-mediated crosstalk among various organs and tissues. Aging is associated with bone loss and muscle atrophy, contributing to frailty, postural instability, and the incidence of falls. Therapeutic interventions such as protein and amino acid supplementation that can increase muscle mass and muscle-derived leptin may have multiple benefits for the elderly that can potentially reduce the incidence of falls and fractures.
Collapse
Affiliation(s)
- Mark W. Hamrick
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, GA, USA
| |
Collapse
|
30
|
Wall BT, Burd NA, Franssen R, Gorissen SHM, Snijders T, Senden JM, Gijsen AP, van Loon LJC. Presleep protein ingestion does not compromise the muscle protein synthetic response to protein ingested the following morning. Am J Physiol Endocrinol Metab 2016; 311:E964-E973. [PMID: 27780822 DOI: 10.1152/ajpendo.00325.2016] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2016] [Revised: 10/11/2016] [Accepted: 10/24/2016] [Indexed: 11/22/2022]
Abstract
Protein ingestion before sleep augments postexercise muscle protein synthesis during overnight recovery. It is unknown whether postexercise and presleep protein consumption modulates postprandial protein handling and myofibrillar protein synthetic responses the following morning. Sixteen healthy young (24 ± 1 yr) men performed unilateral resistance-type exercise (contralateral leg acting as a resting control) at 2000. Participants ingested 20 g of protein immediately after exercise plus 60 g of protein presleep (PRO group; n = 8) or equivalent boluses of carbohydrate (CON; n = 8). The subsequent morning participants received primed, continuous infusions of l-[ring-2H5]phenylalanine and l-[1-13C]leucine combined with ingestion of 20 g intrinsically l-[1-13C]phenylalanine- and l-[1-13C]leucine-labeled protein to assess postprandial protein handling and myofibrillar protein synthesis in the rested and exercised leg in CON and PRO. Exercise increased postabsorptive myofibrillar protein synthesis rates the subsequent day (P < 0.001), with no differences between CON and PRO. Protein ingested in the morning increased myofibrillar protein synthesis in both the exercised and rested leg (P < 0.01), with no differences between treatments. Myofibrillar protein bound l-[1-13C]phenylalanine enrichments were greater in the exercised (0.016 ± 0.002 and 0.015 ± 0.002 MPE in CON and PRO, respectively) vs. rested (0.010 ± 0.002 and 0.009 ± 0.002 MPE in CON and PRO, respectively) leg (P < 0.05), with no differences between treatments (P > 0.05). The additive effects of resistance-type exercise and protein ingestion on myofibrillar protein synthesis persist for more than 12 h after exercise and are not modulated by protein consumption during acute postexercise recovery. This work provides evidence of an extended window of opportunity where presleep protein supplementation can be an effective nutrient timing strategy to optimize skeletal muscle reconditioning.
Collapse
Affiliation(s)
- Benjamin T Wall
- Department of Human Biology and Movement Sciences, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Nicholas A Burd
- Department of Human Biology and Movement Sciences, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Rinske Franssen
- Department of Human Biology and Movement Sciences, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Stefan H M Gorissen
- Department of Human Biology and Movement Sciences, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Tim Snijders
- Department of Human Biology and Movement Sciences, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Joan M Senden
- Department of Human Biology and Movement Sciences, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Annemie P Gijsen
- Department of Human Biology and Movement Sciences, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Luc J C van Loon
- Department of Human Biology and Movement Sciences, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre, Maastricht, The Netherlands
| |
Collapse
|
31
|
Kononikhin AS, Starodubtseva NL, Pastushkova LK, Kashirina DN, Fedorchenko KY, Brhozovsky AG, Popov IA, Larina IM, Nikolaev EN. Spaceflight induced changes in the human proteome. Expert Rev Proteomics 2016; 14:15-29. [PMID: 27817217 DOI: 10.1080/14789450.2017.1258307] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
INTRODUCTION Spaceflight is one of the most extreme conditions encountered by humans: Individuals are exposed to radiation, microgravity, hypodynamia, and will experience isolation. A better understanding of the molecular processes induced by these factors may allow us to develop personalized countermeasures to minimize risks to astronauts. Areas covered: This review is a summary of literature searches from PubMed, NASA, Roskosmos and the authors' research experiences and opinions. The review covers the available proteomic data on the effects of spaceflight factors on the human body, including both real space missions and ground-based model experiments. Expert commentary: Overall, the authors believe that the present background, methodology and equipment improvements will enhance spaceflight safety and support accumulation of new knowledge on how organisms adapt to extreme conditions.
Collapse
Affiliation(s)
- Alexey S Kononikhin
- a Institute of Biomedical Problems - Russian Federation State Scientific Research Center, Laboratory of proteomics , Russian Academy of Sciences , Moscow , Russia.,b Moscow Institute of Physics and Technology , Laboratory of ion and molecular physics , Moscow , Russia.,d V.L. Talrose Institute for Energy Problems of Chemical Physics , Laboratory of ion and molecular physics, Russian Academy of Sciences , Moscow , Russia
| | - Natalia L Starodubtseva
- b Moscow Institute of Physics and Technology , Laboratory of ion and molecular physics , Moscow , Russia.,c V. I. Kulakov Research Center for Obstetrics, Gynecology and Perinatology , Laboratory of proteomics and metabolomics, Ministry of Healthcare of the Russian Federation , Moscow , Russia.,d V.L. Talrose Institute for Energy Problems of Chemical Physics , Laboratory of ion and molecular physics, Russian Academy of Sciences , Moscow , Russia
| | - Lyudmila Kh Pastushkova
- a Institute of Biomedical Problems - Russian Federation State Scientific Research Center, Laboratory of proteomics , Russian Academy of Sciences , Moscow , Russia
| | - Daria N Kashirina
- a Institute of Biomedical Problems - Russian Federation State Scientific Research Center, Laboratory of proteomics , Russian Academy of Sciences , Moscow , Russia
| | | | - Alexander G Brhozovsky
- a Institute of Biomedical Problems - Russian Federation State Scientific Research Center, Laboratory of proteomics , Russian Academy of Sciences , Moscow , Russia
| | - Igor A Popov
- b Moscow Institute of Physics and Technology , Laboratory of ion and molecular physics , Moscow , Russia.,c V. I. Kulakov Research Center for Obstetrics, Gynecology and Perinatology , Laboratory of proteomics and metabolomics, Ministry of Healthcare of the Russian Federation , Moscow , Russia.,d V.L. Talrose Institute for Energy Problems of Chemical Physics , Laboratory of ion and molecular physics, Russian Academy of Sciences , Moscow , Russia
| | - Irina M Larina
- a Institute of Biomedical Problems - Russian Federation State Scientific Research Center, Laboratory of proteomics , Russian Academy of Sciences , Moscow , Russia
| | - Evgeny N Nikolaev
- d V.L. Talrose Institute for Energy Problems of Chemical Physics , Laboratory of ion and molecular physics, Russian Academy of Sciences , Moscow , Russia.,e Emanuel Institute for Biochemical Physics , Russian Academy of Sciences , Moscow , Russia.,f Skolkovo Institute of Science and Technology, Space Cluster , Skolkovo , Russia
| |
Collapse
|
32
|
Reilly BD, Franklin CE. Prevention of muscle wasting and osteoporosis: the value of examining novel animal models. J Exp Biol 2016; 219:2582-95. [DOI: 10.1242/jeb.128348] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
ABSTRACT
Bone mass and skeletal muscle mass are controlled by factors such as genetics, diet and nutrition, growth factors and mechanical stimuli. Whereas increased mechanical loading of the musculoskeletal system stimulates an increase in the mass and strength of skeletal muscle and bone, reduced mechanical loading and disuse rapidly promote a decrease in musculoskeletal mass, strength and ultimately performance (i.e. muscle atrophy and osteoporosis). In stark contrast to artificially immobilised laboratory mammals, animals that experience natural, prolonged bouts of disuse and reduced mechanical loading, such as hibernating mammals and aestivating frogs, consistently exhibit limited or no change in musculoskeletal performance. What factors modulate skeletal muscle and bone mass, and what physiological and molecular mechanisms protect against losses of muscle and bone during dormancy and following arousal? Understanding the events that occur in different organisms that undergo natural periods of prolonged disuse and suffer negligible musculoskeletal deterioration could not only reveal novel regulatory factors but also might lead to new therapeutic options. Here, we review recent work from a diverse array of species that has revealed novel information regarding physiological and molecular mechanisms that dormant animals may use to conserve musculoskeletal mass despite prolonged inactivity. By highlighting some of the differences and similarities in musculoskeletal biology between vertebrates that experience disparate modes of dormancy, it is hoped that this Review will stimulate new insights and ideas for future studies regarding the regulation of atrophy and osteoporosis in both natural and clinical models of muscle and bone disuse.
Collapse
Affiliation(s)
- Beau D. Reilly
- School of Biological Sciences, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Craig E. Franklin
- School of Biological Sciences, The University of Queensland, Brisbane, Queensland 4072, Australia
| |
Collapse
|
33
|
Changes in Pre- and Post-Exercise Gene Expression among Patients with Chronic Kidney Disease and Kidney Transplant Recipients. PLoS One 2016; 11:e0160327. [PMID: 27518102 PMCID: PMC4982681 DOI: 10.1371/journal.pone.0160327] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2016] [Accepted: 07/18/2016] [Indexed: 11/19/2022] Open
Abstract
Introduction Decreased insulin sensitivity blunts the normal increase in gene expression from skeletal muscle after exercise. In addition, chronic inflammation decreases insulin sensitivity. Chronic kidney disease (CKD) is an inflammatory state. How CKD and, subsequently, kidney transplantation affects skeletal muscle gene expression after exercise are unknown. Methods Study cohort: non-diabetic male/female 4/1, age 52±2 years, with end-stage CKD who underwent successful kidney transplantation. The following were measured both pre-transplant and post-transplant and compared to normals: Inflammatory markers, euglycemic insulin clamp studies determine insulin sensitivity, and skeletal muscle biopsies performed before and within 30 minutes after an acute exercise protocol. Microarray analyses were performed on the skeletal muscle using the 4x44K Whole Human Genome Microarrays. Since nuclear factor of activated T cells (NFAT) plays an important role in T cell activation and calcineurin inhibitors are mainstay immunosuppression, calcineurin/NFAT pathway gene expression was compared at rest and after exercise. Log transformation was performed to prevent skewing of data and regression analyses comparing measures pre- and post-transplant performed. Result Markers of inflammation significantly improved post-transplantation. Insulin infusion raised glucose disposal slightly lower post-transplant compared to pre-transplant, but not significantly, thus concluding differences in insulin sensitivity were similar. The overall pattern of gene expression in response to exercise was reduced both pre-and post-transplant compared to healthy volunteers. Although significant changes were observed among NFAT/Calcineurin gene at rest and after exercise in normal cohort, there were no significant differences comparing NFAT/calcineurin pathway gene expression pre- and post-transplant. Conclusions Despite an improvement in serum inflammatory markers, no significant differences in glucose disposal were observed post-transplant. The reduced skeletal muscle gene expression, including NFAT/calcineurin gene expression, in response to a single bout of exercise was not improved post-transplant. This study suggests that the improvements in inflammatory mediators post-transplant are unrelated to changes of NFAT/calcineurin gene expression.
Collapse
|
34
|
Gallagher IJ, Jacobi C, Tardif N, Rooyackers O, Fearon K. Omics/systems biology and cancer cachexia. Semin Cell Dev Biol 2016; 54:92-103. [DOI: 10.1016/j.semcdb.2015.12.022] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2015] [Accepted: 12/30/2015] [Indexed: 10/22/2022]
|
35
|
Stern JH, Rutkowski JM, Scherer PE. Adiponectin, Leptin, and Fatty Acids in the Maintenance of Metabolic Homeostasis through Adipose Tissue Crosstalk. Cell Metab 2016; 23:770-84. [PMID: 27166942 PMCID: PMC4864949 DOI: 10.1016/j.cmet.2016.04.011] [Citation(s) in RCA: 682] [Impact Index Per Article: 85.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Metabolism research has made tremendous progress over the last several decades in establishing the adipocyte as a central rheostat in the regulation of systemic nutrient and energy homeostasis. Operating at multiple levels of control, the adipocyte communicates with organ systems to adjust gene expression, glucoregulatory hormone exocytosis, enzymatic reactions, and nutrient flux to equilibrate the metabolic demands of a positive or negative energy balance. The identification of these mechanisms has great potential to identify novel targets for the treatment of diabetes and related metabolic disorders. Herein, we review the central role of the adipocyte in the maintenance of metabolic homeostasis, highlighting three critical mediators: adiponectin, leptin, and fatty acids.
Collapse
Affiliation(s)
- Jennifer H Stern
- Touchstone Diabetes Center, Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Joseph M Rutkowski
- Touchstone Diabetes Center, Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Philipp E Scherer
- Touchstone Diabetes Center, Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.
| |
Collapse
|
36
|
Wall BT, Dirks ML, Snijders T, van Dijk JW, Fritsch M, Verdijk LB, van Loon LJC. Short-term muscle disuse lowers myofibrillar protein synthesis rates and induces anabolic resistance to protein ingestion. Am J Physiol Endocrinol Metab 2016; 310:E137-47. [PMID: 26578714 DOI: 10.1152/ajpendo.00227.2015] [Citation(s) in RCA: 101] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2015] [Accepted: 11/11/2015] [Indexed: 01/05/2023]
Abstract
Disuse leads to rapid loss of skeletal muscle mass and function. It has been hypothesized that short successive periods of muscle disuse throughout the lifespan play an important role in the development of sarcopenia. The physiological mechanisms underlying short-term muscle disuse atrophy remain to be elucidated. We assessed the impact of 5 days of muscle disuse on postabsorptive and postprandial myofibrillar protein synthesis rates in humans. Twelve healthy young (22 ± 1 yr) men underwent a 5-day period of one-legged knee immobilization (full leg cast). Quadriceps cross-sectional area (CSA) of both legs was assessed before and after immobilization. Continuous infusions of l-[ring-(2)H5]phenylalanine and l-[1-(13)C]leucine were combined with the ingestion of a 25-g bolus of intrinsically l-[1-(13)C]phenylalanine- and l-[1-(13)C]leucine-labeled dietary protein to assess myofibrillar muscle protein fractional synthetic rates in the immobilized and nonimmobilized control leg. Immobilization led to a 3.9 ± 0.6% decrease in quadriceps muscle CSA of the immobilized leg. Based on the l-[ring-(2)H5]phenylalanine tracer, immobilization reduced postabsorptive myofibrillar protein synthesis rates by 41 ± 13% (0.015 ± 0.002 vs. 0.032 ± 0.005%/h, P < 0.01) and postprandial myofibrillar protein synthesis rates by 53 ± 4% (0.020 ± 0.002 vs. 0.044 ± 0.003%/h, P < 0.01). Comparable results were found using the l-[1-(13)C]leucine tracer. Following protein ingestion, myofibrillar protein bound l-[1-(13)C]phenylalanine enrichments were 53 ± 18% lower in the immobilized compared with the control leg (0.007 ± 0.002 and 0.015 ± 0.002 mole% excess, respectively, P < 0.05). We conclude that 5 days of muscle disuse substantially lowers postabsorptive myofibrillar protein synthesis rates and induces anabolic resistance to protein ingestion.
Collapse
Affiliation(s)
- Benjamin T Wall
- Top Institute Food and Nutrition, Wageningen, The Netherlands; NUTRIM School for Nutrition, Toxicology, and Metabolism, Maastricht University Medical Centre, Maastricht, The Netherlands; and
| | - Marlou L Dirks
- NUTRIM School for Nutrition, Toxicology, and Metabolism, Maastricht University Medical Centre, Maastricht, The Netherlands; and
| | - Tim Snijders
- NUTRIM School for Nutrition, Toxicology, and Metabolism, Maastricht University Medical Centre, Maastricht, The Netherlands; and
| | - Jan-Willem van Dijk
- Top Institute Food and Nutrition, Wageningen, The Netherlands; NUTRIM School for Nutrition, Toxicology, and Metabolism, Maastricht University Medical Centre, Maastricht, The Netherlands; and
| | - Mario Fritsch
- Department of Surgery, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Lex B Verdijk
- Top Institute Food and Nutrition, Wageningen, The Netherlands; NUTRIM School for Nutrition, Toxicology, and Metabolism, Maastricht University Medical Centre, Maastricht, The Netherlands; and
| | - Luc J C van Loon
- Top Institute Food and Nutrition, Wageningen, The Netherlands; NUTRIM School for Nutrition, Toxicology, and Metabolism, Maastricht University Medical Centre, Maastricht, The Netherlands; and
| |
Collapse
|
37
|
Malavaki CJ, Sakkas GK, Mitrou GI, Kalyva A, Stefanidis I, Myburgh KH, Karatzaferi C. Skeletal muscle atrophy: disease-induced mechanisms may mask disuse atrophy. J Muscle Res Cell Motil 2016; 36:405-21. [DOI: 10.1007/s10974-015-9439-8] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2015] [Accepted: 12/08/2015] [Indexed: 01/07/2023]
|
38
|
Vibration mechanosignals superimposed to resistive exercise result in baseline skeletal muscle transcriptome profiles following chronic disuse in bed rest. Sci Rep 2015; 5:17027. [PMID: 26596638 PMCID: PMC4657004 DOI: 10.1038/srep17027] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2015] [Accepted: 10/21/2015] [Indexed: 01/08/2023] Open
Abstract
Disuse-induced muscle atrophy is a major concern in aging, in neuromuscular diseases, post-traumatic injury and in microgravity life sciences affecting health and fitness also of crew members in spaceflight. By using a laboratory analogue to body unloading we perform for the first time global gene expression profiling joined to specific proteomic analysis to map molecular adaptations in disused (60 days of bed rest) human soleus muscle (CTR) and in response to a resistive exercise (RE) countermeasure protocol without and with superimposed vibration mechanosignals (RVE). Adopting Affymetrix GeneChip technology we identified 235 differently transcribed genes in the CTR group (end- vs. pre-bed rest). RE comprised 206 differentially expressed genes, whereas only 51 changed gene transcripts were found in RVE. Most gene transcription and proteomic changes were linked to various key metabolic pathways (glycolysis, oxidative phosphorylation, tricarboxylic acid (TCA) cycle, lipid metabolism) and to functional contractile structures. Gene expression profiling in bed rest identified a novel set of genes explicitly responsive to vibration mechanosignals in human soleus. This new finding highlights the efficacy of RVE protocol in reducing key signs of disuse maladaptation and atrophy, and to maintain a close-to-normal skeletal muscle quality outcome following chronic disuse in bed rest.
Collapse
|
39
|
De Naeyer H, Lamon S, Russell AP, Everaert I, De Spaey A, Jamart C, Vanheel B, Taes Y, Derave W. Effects of tail suspension on serum testosterone and molecular targets regulating muscle mass. Muscle Nerve 2015; 52:278-88. [PMID: 25524358 DOI: 10.1002/mus.24542] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2013] [Revised: 10/31/2014] [Accepted: 12/08/2014] [Indexed: 11/07/2022]
Abstract
INTRODUCTION The contribution of reduced testosterone levels to tail suspension (TS)-induced muscle atrophy remains equivocal. The molecular mechanism by which testosterone regulates muscle mass during TS has not been investigated. METHODS Effects of TS on serum testosterone levels, muscle mass, and expression of muscle atrophy- and hypertrophy-inducing targets were measured in soleus (SOL) and extensor digitorum longus (EDL) muscles after testosterone administration during 1, 5, and 14 days of TS in male mice. RESULTS TS produced an increase followed by a transient drop in testosterone levels. Muscle atrophy was associated with downregulation of Igf1 and upregulation of Mstn, Redd1, Atrogin-1, and MuRF1 mRNA with clear differences in Igf1, Mstn, and MAFbx/Atrogin-1 gene expression between SOL and EDL. Testosterone supplementation did not affect muscle mass or protein expression levels during TS. Conclusions The known anabolic effects of testosterone are not sufficient to ameliorate loss of muscle mass during TS.
Collapse
Affiliation(s)
- Hélène De Naeyer
- Department of Movement and Sports Sciences, Ghent University, Watersportlaan 2, 9000, Ghent, Belgium
| | - Séverine Lamon
- Centre for Physical Activity and Nutrition Research, Deakin University, Melbourne, Victoria, Australia
| | - Aaron P Russell
- Centre for Physical Activity and Nutrition Research, Deakin University, Melbourne, Victoria, Australia
| | - Inge Everaert
- Department of Movement and Sports Sciences, Ghent University, Watersportlaan 2, 9000, Ghent, Belgium
| | - Annelies De Spaey
- Department of Endocrinology, Ghent University Hospital, Ghent, Belgium
| | - Cécile Jamart
- Institute of Neuroscience, Université Catholique de Louvain, Louvain-la-Neuve, Belgium
| | - Bert Vanheel
- Department of Basic Medical Sciences, Division of Physiology, Ghent University, Ghent, Belgium
| | - Youri Taes
- Department of Endocrinology, Ghent University Hospital, Ghent, Belgium
| | - Wim Derave
- Department of Movement and Sports Sciences, Ghent University, Watersportlaan 2, 9000, Ghent, Belgium
| |
Collapse
|
40
|
Banerji CRS, Knopp P, Moyle LA, Severini S, Orrell RW, Teschendorff AE, Zammit PS. β-Catenin is central to DUX4-driven network rewiring in facioscapulohumeral muscular dystrophy. J R Soc Interface 2015; 12:20140797. [PMID: 25551153 DOI: 10.1098/rsif.2014.0797] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Facioscapulohumeral muscular dystrophy (FSHD) is an incurable disease, characterized by skeletal muscle weakness and wasting. Genetically, FSHD is characterized by contraction or hypomethylation of repeat D4Z4 units on chromosome 4, which causes aberrant expression of the transcription factor DUX4 from the last repeat. Many genes have been implicated in FSHD pathophysiology, but an integrated molecular model is currently lacking. We developed a novel differential network methodology, Interactome Sparsification and Rewiring (InSpiRe), which detects network rewiring between phenotypes by integrating gene expression data with known protein interactions. Using InSpiRe, we performed a meta-analysis of multiple microarray datasets from FSHD muscle biopsies, then removed secondary rewiring using non-FSHD datasets, to construct a unified network of rewired interactions. Our analysis identified β-catenin as the main coordinator of FSHD-associated protein interaction signalling, with pathways including canonical Wnt, HIF1-α and TNF-α clearly perturbed. To detect transcriptional changes directly elicited by DUX4, gene expression profiling was performed using microarrays on murine myoblasts. This revealed that DUX4 significantly modified expression of the genes in our FSHD network. Furthermore, we experimentally confirmed that Wnt/β-catenin signalling is affected by DUX4 in murine myoblasts. Thus, we provide the first unified molecular map of FSHD signalling, capable of uncovering pathomechanisms and guiding therapeutic development.
Collapse
|
41
|
Slimani L, Vazeille E, Deval C, Meunier B, Polge C, Dardevet D, Béchet D, Taillandier D, Micol D, Listrat A, Attaix D, Combaret L. The delayed recovery of the remobilized rat tibialis anterior muscle reflects a defect in proliferative and terminal differentiation that impairs early regenerative processes. J Cachexia Sarcopenia Muscle 2015; 6:73-83. [PMID: 26136414 PMCID: PMC4435099 DOI: 10.1002/jcsm.12011] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2014] [Revised: 10/09/2014] [Accepted: 10/15/2014] [Indexed: 01/13/2023] Open
Abstract
BACKGROUND The immobilization-induced tibialis anterior (TA) muscle atrophy worsens after cast removal and is associated with altered extracellular matrix (ECM) composition. The secreted protein acidic and rich in cysteine (Sparc) is an ECM component involved in Akt activation and in β-catenin stabilization, which controls protein turnover and induces muscle regulatory factors (MRFs), respectively. We hypothesized that ECM alterations may influence these intracellular signalling pathways controlling TA muscle mass. METHODS Six-month-old Wistar rats were subjected to hindlimb cast immobilization for 8 days (I8) or not (I0) and allowed to recover for 1 to 10 days (R1-10). RESULTS The TA atrophy during remobilization correlated with reduced fibre cross-sectional area and thickening of endomysium. mRNA levels for Sparc increased during remobilization until R10 and for integrin-α7 and -β1 at I8 and R1. Integrin-linked kinase protein levels increased during immobilization and remobilization until R10. This was inversely correlated with changes in Akt phosphorylation. β-Catenin protein levels increased in the remobilized TA at R1 and R10. mRNA levels of the proliferative MRFs (Myf5 and MyoD) increased at I8 and R1, respectively, without changes in Myf5 protein levels. In contrast, myogenin mRNA levels (a terminal differentiation MRF) decreased at R1, but only increased at R10 in remobilized muscles, as for protein levels. CONCLUSIONS Altogether, this suggests that the TA inefficiently attempted to preserve regeneration during immobilization by increasing transcription of proliferative MRFs, and that the TA could engage recovery during remobilization only when the terminal differentiation step of regeneration is enhanced.
Collapse
Affiliation(s)
- Lamia Slimani
- INRA, UMR 1019, UNH, CRNH, F-63000, Auvergne, Clermont-Ferrand, France.,Clermont Université, Université d'Auvergne, Unité de Nutrition Humaine, BP 10448, F-63000, Clermont-Ferrand, France
| | - Emilie Vazeille
- Centre Hospitalier Universitaire, 63000, Clermont-Ferrand, France
| | - Christiane Deval
- INRA, UMR 1019, UNH, CRNH, F-63000, Auvergne, Clermont-Ferrand, France.,Clermont Université, Université d'Auvergne, Unité de Nutrition Humaine, BP 10448, F-63000, Clermont-Ferrand, France
| | - Bruno Meunier
- INRA, UMR 1213 Herbivores, 63122, Saint Genès Champanelle, France
| | - Cécile Polge
- INRA, UMR 1019, UNH, CRNH, F-63000, Auvergne, Clermont-Ferrand, France.,Clermont Université, Université d'Auvergne, Unité de Nutrition Humaine, BP 10448, F-63000, Clermont-Ferrand, France
| | - Dominique Dardevet
- INRA, UMR 1019, UNH, CRNH, F-63000, Auvergne, Clermont-Ferrand, France.,Clermont Université, Université d'Auvergne, Unité de Nutrition Humaine, BP 10448, F-63000, Clermont-Ferrand, France
| | - Daniel Béchet
- INRA, UMR 1019, UNH, CRNH, F-63000, Auvergne, Clermont-Ferrand, France.,Clermont Université, Université d'Auvergne, Unité de Nutrition Humaine, BP 10448, F-63000, Clermont-Ferrand, France
| | - Daniel Taillandier
- INRA, UMR 1019, UNH, CRNH, F-63000, Auvergne, Clermont-Ferrand, France.,Clermont Université, Université d'Auvergne, Unité de Nutrition Humaine, BP 10448, F-63000, Clermont-Ferrand, France
| | - Didier Micol
- INRA, UMR 1213 Herbivores, 63122, Saint Genès Champanelle, France
| | - Anne Listrat
- INRA, UMR 1213 Herbivores, 63122, Saint Genès Champanelle, France
| | - Didier Attaix
- INRA, UMR 1019, UNH, CRNH, F-63000, Auvergne, Clermont-Ferrand, France.,Clermont Université, Université d'Auvergne, Unité de Nutrition Humaine, BP 10448, F-63000, Clermont-Ferrand, France
| | - Lydie Combaret
- INRA, UMR 1019, UNH, CRNH, F-63000, Auvergne, Clermont-Ferrand, France.,Clermont Université, Université d'Auvergne, Unité de Nutrition Humaine, BP 10448, F-63000, Clermont-Ferrand, France
| |
Collapse
|
42
|
Wall BT, Dirks ML, Snijders T, Stephens FB, Senden JM, Verscheijden ML, van Loon LJ. Short-term muscle disuse atrophy is not associated with increased intramuscular lipid deposition or a decline in the maximal activity of key mitochondrial enzymes in young and older males. Exp Gerontol 2015; 61:76-83. [DOI: 10.1016/j.exger.2014.11.019] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2014] [Revised: 11/11/2014] [Accepted: 11/27/2014] [Indexed: 01/06/2023]
|
43
|
Reilly BD, Cramp RL, Franklin CE. Activity, abundance and expression of Ca2+-activated proteases in skeletal muscle of the aestivating frog, Cyclorana alboguttata. J Comp Physiol B 2014; 185:243-55. [DOI: 10.1007/s00360-014-0880-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2014] [Revised: 11/18/2014] [Accepted: 11/27/2014] [Indexed: 10/24/2022]
|
44
|
MacNeil LG, Glover E, Bergstra TG, Safdar A, Tarnopolsky MA. The order of exercise during concurrent training for rehabilitation does not alter acute genetic expression, mitochondrial enzyme activity or improvements in muscle function. PLoS One 2014; 9:e109189. [PMID: 25289940 PMCID: PMC4188604 DOI: 10.1371/journal.pone.0109189] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2014] [Accepted: 09/08/2014] [Indexed: 01/09/2023] Open
Abstract
Concurrent exercise combines different modes of exercise (e.g., aerobic and resistance) into one training protocol, providing stimuli meant to increase muscle strength, aerobic capacity and mass. As disuse is associated with decrements in strength, aerobic capacity and muscle size concurrent training is an attractive modality for rehabilitation. However, interference between the signaling pathways may result in preferential improvements for one of the exercise modes. We recruited 18 young adults (10 ♂, 8 ♀) to determine if order of exercise mode during concurrent training would differentially affect gene expression, protein content and measures of strength and aerobic capacity after 2 weeks of knee-brace induced disuse. Concurrent exercise sessions were performed 3x/week for 6 weeks at gradually increasing intensities either with endurance exercise preceding (END>RES) or following (RES>END) resistance exercise. Biopsies were collected from the vastus lateralis before, 3 h after the first exercise bout and 48 h after the end of training. Concurrent exercise altered the expression of genes involved in mitochondrial biogenesis (PGC-1α, PRC, PPARγ), hypertrophy (PGC-1α4, REDD2, Rheb) and atrophy (MuRF-1, Runx1), increased electron transport chain complex protein content, citrate synthase and mitochondrial cytochrome c oxidase enzyme activity, muscle mass, maximum isometric strength and VO2peak. However, the order in which exercise was completed (END>RES or RES>END) only affected the protein content of mitochondrial complex II subunit. In conclusion, concurrent exercise training is an effective modality for the rehabilitation of the loss of skeletal muscle mass, maximum strength, and peak aerobic capacity resulting from disuse, regardless of the order in which the modes of exercise are performed.
Collapse
Affiliation(s)
- Lauren G. MacNeil
- Department of Pediatrics, McMaster University, Hamilton, Ontario, Canada
| | - Elisa Glover
- Department of Pediatrics, McMaster University, Hamilton, Ontario, Canada
| | - T. Graham Bergstra
- Department of Health Sciences, McMaster University, Hamilton, Ontario, Canada
| | - Adeel Safdar
- Department of Pediatrics, McMaster University, Hamilton, Ontario, Canada
| | - Mark A. Tarnopolsky
- Department of Pediatrics, McMaster University, Hamilton, Ontario, Canada
- Department of Medicine, McMaster University, Hamilton, Ontario, Canada
- * E-mail:
| |
Collapse
|
45
|
Trudel G, Laneuville O, Coletta E, Goudreau L, Uhthoff HK. Quantitative and temporal differential recovery of articular and muscular limitations of knee joint contractures; results in a rat model. J Appl Physiol (1985) 2014; 117:730-7. [DOI: 10.1152/japplphysiol.00409.2014] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Joint contractures alter the mechanical properties of articular and muscular structures. Reversibility of a contracture depends on the restoration of the elasticity of both structures. We determined the differential contribution of articular and muscular structures to knee flexion contractures during spontaneous recovery. Rats (250, divided into 24 groups) had one knee joint surgically fixed in flexion for six different durations, from 1 to 32 wk, creating joint contractures of various severities. After the fixation was removed, the animals were left to spontaneously recover for 1 to 48 wk. After the recovery periods, animals were killed and the knee extension was measured before and after division of the transarticular posterior muscles using a motorized arthrometer. No articular limitation had developed in contracture of recent onset (≤2 wk of fixation, P > 0.05); muscular limitations were responsible for the majority of the contracture (34 ± 8° and 38 ± 6°, respectively; both P < 0.05). Recovery for 1 and 8 wk reversed the muscular limitation of contractures of recent onset (1 and 2 wk of fixation, respectively). Long-lasting contractures (≥4 wk of fixation) presented articular limitations, irreversible in all 12 durations of recovery compared with controls (all 12 P < 0.05). Knee flexion contractures of recent onset were primarily due to muscular structures, and they were reversible during spontaneous recovery. Long-lasting contractures were primarily due to articular structures and were irreversible. Comprehensive temporal and quantitative data on the differential reversibility of mechanically significant alterations in articular and muscular structures represent novel evidence on which to base clinical practice.
Collapse
Affiliation(s)
- Guy Trudel
- Division of Physical Medicine and Rehabilitation, Department of Medicine, University of Ottawa, Ottawa, Ontario, Canada
- Bone and Joint Research Laboratory, University of Ottawa, Ottawa, Ontario, Canada
| | - Odette Laneuville
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, Ontario, Canada
| | - Elizabeth Coletta
- Bone and Joint Research Laboratory, University of Ottawa, Ottawa, Ontario, Canada
| | - Louis Goudreau
- Biomedical Engineering, The Ottawa Hospital Rehabilitation Centre, Ottawa, Ontario, Canada; and
| | - Hans K. Uhthoff
- Bone and Joint Research Laboratory, University of Ottawa, Ottawa, Ontario, Canada
- Division of Orthopedic Surgery, Department of Surgery, University of Ottawa, Ottawa, Ontario, Canada
| |
Collapse
|
46
|
Early changes in costameric and mitochondrial protein expression with unloading are muscle specific. BIOMED RESEARCH INTERNATIONAL 2014; 2014:519310. [PMID: 25313365 PMCID: PMC4182083 DOI: 10.1155/2014/519310] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/09/2014] [Accepted: 08/23/2014] [Indexed: 11/25/2022]
Abstract
We hypothesised that load-sensitive expression of costameric proteins, which hold the sarcomere in place and position the mitochondria, contributes to the early adaptations of antigravity muscle to unloading and would depend on muscle fibre composition and chymotrypsin activity of the proteasome. Biopsies were obtained from vastus lateralis (VL) and soleus (SOL) muscles of eight men before and after 3 days of unilateral lower limb suspension (ULLS) and subjected to fibre typing and measures for costameric (FAK and FRNK), mitochondrial (NDUFA9, SDHA, UQCRC1, UCP3, and ATP5A1), and MHCI protein and RNA content. Mean cross-sectional area (MCSA) of types I and II muscle fibres in VL and type I fibres in SOL demonstrated a trend for a reduction after ULLS (0.05 ≤ P < 0.10). FAK phosphorylation at tyrosine 397 showed a 20% reduction in VL muscle (P = 0.029). SOL muscle demonstrated a specific reduction in UCP3 content (−23%; P = 0.012). Muscle-specific effects of ULLS were identified for linear relationships between measured proteins, chymotrypsin activity and fibre MCSA. The molecular modifications in costamere turnover and energy homoeostasis identify that aspects of atrophy and fibre transformation are detectable at the protein level in weight-bearing muscles within 3 days of unloading.
Collapse
|
47
|
Abstract
Muscle RING finger 1 (MuRF1) and muscle atrophy F-box (MAFbx)/atrogin-1 were identified more than 10 years ago as two muscle-specific E3 ubiquitin ligases that are increased transcriptionally in skeletal muscle under atrophy-inducing conditions, making them excellent markers of muscle atrophy. In the past 10 years much has been published about MuRF1 and MAFbx with respect to their mRNA expression patterns under atrophy-inducing conditions, their transcriptional regulation, and their putative substrates. However, much remains to be learned about the physiological role of both genes in the regulation of mass and other cellular functions in striated muscle. Although both MuRF1 and MAFbx are enriched in skeletal, cardiac, and smooth muscle, this review will focus on the current understanding of MuRF1 and MAFbx in skeletal muscle, highlighting the critical questions that remain to be answered.
Collapse
Affiliation(s)
- Sue C Bodine
- Departments of Neurobiology, Physiology, and Behavior and Physiology and Membrane Biology, University of California Davis, Davis, California; and Northern California Veterans Affairs Health Systems, Mather, California
| | - Leslie M Baehr
- Membrane Biology, University of California Davis, Davis, California; and
| |
Collapse
|
48
|
Mobley CB, Fox CD, Ferguson BS, Amin RH, Dalbo VJ, Baier S, Rathmacher JA, Wilson JM, Roberts MD. L-leucine, beta-hydroxy-beta-methylbutyric acid (HMB) and creatine monohydrate prevent myostatin-induced Akirin-1/Mighty mRNA down-regulation and myotube atrophy. J Int Soc Sports Nutr 2014; 11:38. [PMID: 25132809 PMCID: PMC4134516 DOI: 10.1186/1550-2783-11-38] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2014] [Accepted: 06/30/2014] [Indexed: 12/03/2022] Open
Abstract
Background The purpose of this study was to examine if L-leucine (Leu), β-hydroxy-β-methylbutyrate (HMB), or creatine monohydrate (Crea) prevented potential atrophic effects of myostatin (MSTN) on differentiated C2C12 myotubes. Methods After four days of differentiation, myotubes were treated with MSTN (10 ng/ml) for two additional days and four treatment groups were studied: 1) 3x per day 10 mM Leu, 2) 3x per day 10 mM HMB, 3) 3x per day 10 mM Crea, 4) DM only. Myotubes treated with DM without MSTN were analyzed as the control condition (DM/CTL). Following treatment, cells were analyzed for total protein, DNA content, RNA content, muscle protein synthesis (MPS, SUnSET method), and fiber diameter. Separate batch treatments were analyzed for mRNA expression patterns of myostatin-related genes (Akirin-1/Mighty, Notch-1, Ski, MyoD) as well as atrogenes (MuRF-1, and MAFbx/Atrogin-1). Results MSTN decreased fiber diameter approximately 30% compared to DM/CTL myotubes (p < 0.001). Leu, HMB and Crea prevented MSTN-induced atrophy. MSTN did not decrease MPS levels compared to DM/CTL myotubes, but MSTN treatment decreased the mRNA expression of Akirin-1/Mighty by 27% (p < 0.001) and MyoD by 26% (p < 0.01) compared to DM/CTL myotubes. shRNA experiments confirmed that Mighty mRNA knockdown reduced myotube size, linking MSTN treatment to atrophy independent of MPS. Remarkably, MSTN + Leu and MSTN + HMB myotubes had similar Akirin-1/Mighty and MyoD mRNA levels compared to DM/CTL myotubes. Furthermore, MSTN + Crea myotubes exhibited a 36% (p < 0.05) and 86% (p < 0.001) increase in Akirin-1/Mighty mRNA compared to DM/CTL and MSTN-only treated myotubes, respectively. Conclusions Leu, HMB and Crea may reduce MSTN-induced muscle fiber atrophy by influencing Akirin-1/Mighty mRNA expression patterns. Future studies are needed to examine if Leu, HMB and Crea independently or synergistically affect Akirin-1/Mighty expression, and how Akirin-1/Mighty expression mechanistically relates to skeletal muscle hypertrophy in vivo.
Collapse
Affiliation(s)
- Christopher Brooks Mobley
- School of Kinesiology, Molecular and Applied Sciences Laboratory, Auburn University, 301 Wire Road, Office 286, Auburn, AL 36849, USA
| | - Carlton D Fox
- School of Kinesiology, Molecular and Applied Sciences Laboratory, Auburn University, 301 Wire Road, Office 286, Auburn, AL 36849, USA
| | - Brian S Ferguson
- School of Kinesiology, Molecular and Applied Sciences Laboratory, Auburn University, 301 Wire Road, Office 286, Auburn, AL 36849, USA
| | - Rajesh H Amin
- Harrison School of Pharmacy, Auburn University, Auburn, AL, USA
| | - Vincent J Dalbo
- School of Medical and Applied Sciences, Central Queensland University, Rockhampton, QLD, Australia
| | | | | | - Jacob M Wilson
- Department of Health Sciences and Human Performance, University of Tampa, Tampa, FL, USA
| | - Michael D Roberts
- School of Kinesiology, Molecular and Applied Sciences Laboratory, Auburn University, 301 Wire Road, Office 286, Auburn, AL 36849, USA
| |
Collapse
|
49
|
Vendelbo MH, Møller AB, Christensen B, Nellemann B, Clasen BFF, Nair KS, Jørgensen JOL, Jessen N, Møller N. Fasting increases human skeletal muscle net phenylalanine release and this is associated with decreased mTOR signaling. PLoS One 2014; 9:e102031. [PMID: 25020061 PMCID: PMC4096723 DOI: 10.1371/journal.pone.0102031] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2013] [Accepted: 06/13/2014] [Indexed: 12/12/2022] Open
Abstract
Aim Fasting is characterised by profound changes in energy metabolism including progressive loss of body proteins. The underlying mechanisms are however unknown and we therefore determined the effects of a 72-hour-fast on human skeletal muscle protein metabolism and activation of mammalian target of rapamycin (mTOR), a key regulator of cell growth. Methods Eight healthy male volunteers were studied twice: in the postabsorptive state and following 72 hours of fasting. Regional muscle amino acid kinetics was measured in the forearm using amino acid tracers. Signaling to protein synthesis and breakdown were assessed in skeletal muscle biopsies obtained during non-insulin and insulin stimulated conditions on both examination days. Results Fasting significantly increased forearm net phenylalanine release and tended to decrease phenylalanine rate of disappearance. mTOR phosphorylation was decreased by ∼50% following fasting, together with reduced downstream phosphorylation of 4EBP1, ULK1 and rpS6. In addition, the insulin stimulated increase in mTOR and rpS6 phosphorylation was significantly reduced after fasting indicating insulin resistance in this part of the signaling pathway. Autophagy initiation is in part regulated by mTOR through ULK1 and fasting increased expression of the autophagic marker LC3B-II by ∼30%. p62 is degraded during autophagy but was increased by ∼10% during fasting making interpretation of autophagic flux problematic. MAFbx and MURF1 ubiquitin ligases remained unaltered after fasting indicating no change in protesomal protein degradation. Conclusions Our results show that during fasting increased net phenylalanine release in skeletal muscle is associated to reduced mTOR activation and concomitant decreased downstream signaling to cell growth.
Collapse
Affiliation(s)
- Mikkel Holm Vendelbo
- Department of Endocrinology and Internal Medicine, Aarhus University Hospital, Aarhus, Denmark
- * E-mail:
| | - Andreas Buch Møller
- Research Laboratory for Biochemical Pathology, Aarhus University Hospital, Aarhus, Denmark
| | - Britt Christensen
- Department of Endocrinology and Internal Medicine, Aarhus University Hospital, Aarhus, Denmark
| | - Birgitte Nellemann
- Department of Endocrinology and Internal Medicine, Aarhus University Hospital, Aarhus, Denmark
| | - Berthil Frederik Forrest Clasen
- Department of Endocrinology and Internal Medicine, Aarhus University Hospital, Aarhus, Denmark
- Research Laboratory for Biochemical Pathology, Aarhus University Hospital, Aarhus, Denmark
| | - K. Sreekumaran Nair
- Division of Endocrinology, Endocrine Research Unit, Mayo Clinic, Rochester, Minnesota, United States of America
| | | | - Niels Jessen
- Department of Endocrinology and Internal Medicine, Aarhus University Hospital, Aarhus, Denmark
- Research Laboratory for Biochemical Pathology, Aarhus University Hospital, Aarhus, Denmark
- Department of Molecular Medicine, Aarhus University Hospital, Aarhus, Denmark
| | - Niels Møller
- Department of Endocrinology and Internal Medicine, Aarhus University Hospital, Aarhus, Denmark
| |
Collapse
|
50
|
Romacho T, Elsen M, Röhrborn D, Eckel J. Adipose tissue and its role in organ crosstalk. Acta Physiol (Oxf) 2014; 210:733-53. [PMID: 24495317 DOI: 10.1111/apha.12246] [Citation(s) in RCA: 194] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2013] [Revised: 12/18/2013] [Accepted: 01/28/2014] [Indexed: 12/13/2022]
Abstract
The discovery of adipokines has revealed adipose tissue as a central node in the interorgan crosstalk network, which mediates the regulation of multiple organs and tissues. Adipose tissue is a true endocrine organ that produces and secretes a wide range of mediators regulating adipose tissue function in an auto-/paracrine manner and important distant targets, such as the liver, skeletal muscle, the pancreas and the cardiovascular system. In metabolic disorders such as obesity, enlargement of adipocytes leads to adipose tissue dysfunction and a shift in the secretory profile with an increased release of pro-inflammatory adipokines. Adipose tissue dysfunction has a central role in the development of insulin resistance, type 2 diabetes, and cardiovascular diseases. Besides the well-acknowledged role of adipokines in metabolic diseases, and the increasing number of adipokines being discovered in the last years, the mechanisms underlying the release of many adipokines from adipose tissue remain largely unknown. To combat metabolic diseases, it is crucial to better understand how adipokines can modulate adipose tissue growth and function. Therefore, we will focus on adipokines with a prominent role in auto-/paracrine crosstalk within the adipose tissue such as RBP4, HO-1, WISP2, SFRPs and chemerin. To depict the endocrine crosstalk between adipose tissue with skeletal muscle, the cardiovascular system and the pancreas, we will report the main findings regarding the direct effects of adiponectin, leptin, DPP4 and visfatin on skeletal muscle insulin resistance, cardiovascular function and β-cell growth and function.
Collapse
Affiliation(s)
- T. Romacho
- Paul-Langerhans-Group for Integrative Physiology; German Diabetes Center; Düsseldorf Germany
| | - M. Elsen
- Paul-Langerhans-Group for Integrative Physiology; German Diabetes Center; Düsseldorf Germany
| | - D. Röhrborn
- Paul-Langerhans-Group for Integrative Physiology; German Diabetes Center; Düsseldorf Germany
| | - J. Eckel
- Paul-Langerhans-Group for Integrative Physiology; German Diabetes Center; Düsseldorf Germany
- German Center for Diabetes Research (DZD e.V.); Düsseldorf Germany
| |
Collapse
|