1
|
Xu X, Talifu Z, Zhang CJ, Gao F, Ke H, Pan YZ, Gong H, Du HY, Yu Y, Jing YL, Du LJ, Li JJ, Yang DG. Mechanism of skeletal muscle atrophy after spinal cord injury: A narrative review. Front Nutr 2023; 10:1099143. [PMID: 36937344 PMCID: PMC10020380 DOI: 10.3389/fnut.2023.1099143] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Accepted: 02/20/2023] [Indexed: 03/06/2023] Open
Abstract
Spinal cord injury leads to loss of innervation of skeletal muscle, decreased motor function, and significantly reduced load on skeletal muscle, resulting in atrophy. Factors such as braking, hormone level fluctuation, inflammation, and oxidative stress damage accelerate skeletal muscle atrophy. The atrophy process can result in skeletal muscle cell apoptosis, protein degradation, fat deposition, and other pathophysiological changes. Skeletal muscle atrophy not only hinders the recovery of motor function but is also closely related to many systemic dysfunctions, affecting the prognosis of patients with spinal cord injury. Extensive research on the mechanism of skeletal muscle atrophy and intervention at the molecular level has shown that inflammation and oxidative stress injury are the main mechanisms of skeletal muscle atrophy after spinal cord injury and that multiple pathways are involved. These may become targets of future clinical intervention. However, most of the experimental studies are still at the basic research stage and still have some limitations in clinical application, and most of the clinical treatments are focused on rehabilitation training, so how to develop more efficient interventions in clinical treatment still needs to be further explored. Therefore, this review focuses mainly on the mechanisms of skeletal muscle atrophy after spinal cord injury and summarizes the cytokines and signaling pathways associated with skeletal muscle atrophy in recent studies, hoping to provide new therapeutic ideas for future clinical work.
Collapse
Affiliation(s)
- Xin Xu
- School of Rehabilitation, Capital Medical University, Beijing, China
- Department of Spinal and Neural Functional Reconstruction, China Rehabilitation Research Center, Beijing, China
- Chinese Institute of Rehabilitation Science, Beijing, China
- Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, China
- Beijing Key Laboratory of Neural Injury and Rehabilitation, Beijing, China
| | - Zuliyaer Talifu
- School of Rehabilitation, Capital Medical University, Beijing, China
- Department of Spinal and Neural Functional Reconstruction, China Rehabilitation Research Center, Beijing, China
- Chinese Institute of Rehabilitation Science, Beijing, China
- Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, China
- Beijing Key Laboratory of Neural Injury and Rehabilitation, Beijing, China
- School of Rehabilitation Sciences and Engineering, University of Health and Rehabilitation Sciences, Qingdao, Shandong, China
| | - Chun-Jia Zhang
- School of Rehabilitation, Capital Medical University, Beijing, China
- Department of Spinal and Neural Functional Reconstruction, China Rehabilitation Research Center, Beijing, China
- Chinese Institute of Rehabilitation Science, Beijing, China
- Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, China
- Beijing Key Laboratory of Neural Injury and Rehabilitation, Beijing, China
| | - Feng Gao
- School of Rehabilitation, Capital Medical University, Beijing, China
- Department of Spinal and Neural Functional Reconstruction, China Rehabilitation Research Center, Beijing, China
- Chinese Institute of Rehabilitation Science, Beijing, China
- Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, China
- Beijing Key Laboratory of Neural Injury and Rehabilitation, Beijing, China
| | - Han Ke
- School of Rehabilitation, Capital Medical University, Beijing, China
- Department of Spinal and Neural Functional Reconstruction, China Rehabilitation Research Center, Beijing, China
- Chinese Institute of Rehabilitation Science, Beijing, China
- Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, China
- Beijing Key Laboratory of Neural Injury and Rehabilitation, Beijing, China
- School of Rehabilitation Sciences and Engineering, University of Health and Rehabilitation Sciences, Qingdao, Shandong, China
| | - Yun-Zhu Pan
- School of Rehabilitation, Capital Medical University, Beijing, China
- Department of Spinal and Neural Functional Reconstruction, China Rehabilitation Research Center, Beijing, China
- Chinese Institute of Rehabilitation Science, Beijing, China
- Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, China
- Beijing Key Laboratory of Neural Injury and Rehabilitation, Beijing, China
- School of Rehabilitation Sciences and Engineering, University of Health and Rehabilitation Sciences, Qingdao, Shandong, China
| | - Han Gong
- School of Rehabilitation, Capital Medical University, Beijing, China
- Department of Spinal and Neural Functional Reconstruction, China Rehabilitation Research Center, Beijing, China
- Chinese Institute of Rehabilitation Science, Beijing, China
- Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, China
- Beijing Key Laboratory of Neural Injury and Rehabilitation, Beijing, China
| | - Hua-Yong Du
- School of Rehabilitation, Capital Medical University, Beijing, China
- Department of Spinal and Neural Functional Reconstruction, China Rehabilitation Research Center, Beijing, China
- Chinese Institute of Rehabilitation Science, Beijing, China
- Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, China
- Beijing Key Laboratory of Neural Injury and Rehabilitation, Beijing, China
| | - Yan Yu
- School of Rehabilitation, Capital Medical University, Beijing, China
- Chinese Institute of Rehabilitation Science, Beijing, China
- Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, China
- Beijing Key Laboratory of Neural Injury and Rehabilitation, Beijing, China
| | - Ying-Li Jing
- School of Rehabilitation, Capital Medical University, Beijing, China
- Chinese Institute of Rehabilitation Science, Beijing, China
- Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, China
- Beijing Key Laboratory of Neural Injury and Rehabilitation, Beijing, China
| | - Liang-Jie Du
- School of Rehabilitation, Capital Medical University, Beijing, China
- Department of Spinal and Neural Functional Reconstruction, China Rehabilitation Research Center, Beijing, China
- Chinese Institute of Rehabilitation Science, Beijing, China
- Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, China
- Beijing Key Laboratory of Neural Injury and Rehabilitation, Beijing, China
| | - Jian-Jun Li
- School of Rehabilitation, Capital Medical University, Beijing, China
- Department of Spinal and Neural Functional Reconstruction, China Rehabilitation Research Center, Beijing, China
- Chinese Institute of Rehabilitation Science, Beijing, China
- Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, China
- Beijing Key Laboratory of Neural Injury and Rehabilitation, Beijing, China
- School of Rehabilitation Sciences and Engineering, University of Health and Rehabilitation Sciences, Qingdao, Shandong, China
- *Correspondence: Jian-Jun Li
| | - De-Gang Yang
- School of Rehabilitation, Capital Medical University, Beijing, China
- Department of Spinal and Neural Functional Reconstruction, China Rehabilitation Research Center, Beijing, China
- Chinese Institute of Rehabilitation Science, Beijing, China
- Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, China
- Beijing Key Laboratory of Neural Injury and Rehabilitation, Beijing, China
- De-Gang Yang
| |
Collapse
|
2
|
Pingel J, Kjer HM, Biering-Sørensen F, Feidenhans'l R, Dyrby TB. 3D synchrotron imaging of muscle tissues at different atrophic stages in stroke and spinal cord injury: a proof-of-concept study. Sci Rep 2022; 12:17289. [PMID: 36241693 PMCID: PMC9568578 DOI: 10.1038/s41598-022-21741-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Accepted: 09/30/2022] [Indexed: 01/10/2023] Open
Abstract
Synchrotron X-ray computed tomography (SXCT) allows 3D imaging of tissue with a very large field of view and an excellent micron resolution and enables the investigation of muscle fiber atrophy in 3D. The study aimed to explore the 3D micro-architecture of healthy skeletal muscle fibers and muscle fibers at different stages of atrophy (stroke sample = muscle atrophy; spinal cord injury (SCI) sample = severe muscle atrophy). Three muscle samples: a healthy control sample; a stroke sample (atrophic sample), and an SCI sample (severe atrophic sample) were imaged using SXCT, and muscle fiber populations were segmented and quantified for microarchitecture and morphology differences. The volume fraction of muscle fibers was 74.7%, 70.2%, and 35.3% in the healthy, stroke (atrophic), and SCI (severe atrophic) muscle fiber population samples respectively. In the SCI (severe atrophic sample), 3D image analysis revealed fiber splitting and fiber swelling. In the stroke sample (atrophic sample) muscle fiber buckling was observed but was only visible in the 3D analysis. 3D muscle fiber population analysis revealed new insights into the different stages of muscle fiber atrophy not to be observed nor quantified with a 2D histological analysis including fiber buckling, loss of fibers and fiber splitting.
Collapse
Affiliation(s)
- Jessica Pingel
- Department of Neuroscience, University of Copenhagen, Copenhagen, Denmark.
| | - Hans Martin Kjer
- Department of Applied Mathematics and Computer Science, Technical University of Denmark, Lyngby, Denmark
| | - Fin Biering-Sørensen
- Section for Spinal Cord Injuries, Department for Brain and Spinal Cord Injuries, Rigshospitalet and Institute of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Robert Feidenhans'l
- Niels Bohr Institute, University of Copenhagen, Copenhagen, Denmark
- European X-Ray Free Electron Laser, Schenefeld, Germany
| | - Tim B Dyrby
- Department of Applied Mathematics and Computer Science, Technical University of Denmark, Lyngby, Denmark
- Danish Research Centre for Magnetic Resonance, Copenhagen University Hospital Hvidovre and Amager, Hvidovre, Denmark
| |
Collapse
|
3
|
Yu W, Zha W, Peng H, Wang Q, Zhang S, Ren J. Trehalose Protects against Insulin Resistance-Induced Tissue Injury and Excessive Autophagy in Skeletal Muscles and Kidney. Curr Pharm Des 2020; 25:2077-2085. [PMID: 31538882 DOI: 10.2174/1381612825666190708221539] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Accepted: 06/24/2019] [Indexed: 01/20/2023]
Abstract
BACKGROUND Insulin resistance refers to a pathological state of compromised sensitivity of insulin to promote glucose uptake and utilization, resulting in compensatory excessive insulin secretion and hyperinsulinemia in an effort to maintain glucose homeostasis. Akt2 represents an important member of the Akt family and plays an essential role in the maintenance of insulin signaling. METHODS This study was designed to examine the effects of trehalose on kidney and skeletal muscle (rectus femoris muscle) injury in an Akt2 knockout-induced model of insulin resistance. Akt2 knockout (Akt2-/-) and adult WT mice were treated with trehalose (1 mg/g/d) intraperitoneally for 2 days, followed by providing 2% trehalose in drinking water for 2 months. Intraperitoneal glucose tolerance test (IPGTT), protein carbonyl content and mitochondrial function (aconitase activity) were examined. Apoptosis and autophagy protein markers were monitored using western blot analysis. RESULTS Akt2 ablation impaired glucose tolerance, promoted protein carbonyl formation and decreased aconitase activity in kidney and skeletal muscles, associated with pronounced apoptosis and overt autophagy, the effects of which, with the exception of IPGTT, were greatly ameliorated or negated by trehalose treatment. Moreover, phosphorylation of mTOR was downregulated in both kidney and skeletal muscles from Akt2-/- mice, the effect of which was attenuated by trehalose. Levels of Akt (pan and Akt2) were much lower in Akt2-/- mice, the effect of which was unaffected by trehalose treatment although trehalose itself upregulated Akt levels. CONCLUSION These data suggest that the autophagy inducer trehalose rescued against insulin resistance-induced kidney and skeletal muscle injury, apoptosis and excessive autophagy, possibly in association with restored mTOR phosphorylation without affecting Akt.
Collapse
Affiliation(s)
- Wei Yu
- Department of Pharmacology, School of Pharmacy,Hubei University of Science and Technology, Xianning, Hubei, 437100, China.,Center for Cardiovascular Research and Alternative Medicine, University of Wyoming College of Health Sciences, Laramie, WY 82071, United States
| | - Wenliang Zha
- Center for Cardiovascular Research and Alternative Medicine, University of Wyoming College of Health Sciences, Laramie, WY 82071, United States.,Department of Surgery, Clinic Medical College, Hubei University of Science and Technology, Xianning, Hubei, 437100, China
| | - Hu Peng
- Department of Emergency, Shanghai Tenth People's Hospital, School of Medicine Tongji University, Shanghai, 200072, China
| | - Qiurong Wang
- Center for Cardiovascular Research and Alternative Medicine, University of Wyoming College of Health Sciences, Laramie, WY 82071, United States
| | - Shuning Zhang
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital Fudan University, Shanghai 200032, China
| | - Jun Ren
- Center for Cardiovascular Research and Alternative Medicine, University of Wyoming College of Health Sciences, Laramie, WY 82071, United States.,Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital Fudan University, Shanghai 200032, China
| |
Collapse
|
4
|
Graham ZA, Goldberger A, Azulai D, Conover CF, Ye F, Bauman WA, Cardozo CP, Yarrow JF. Contusion spinal cord injury upregulates p53 protein expression in rat soleus muscle at multiple timepoints but not key senescence cytokines. Physiol Rep 2020; 8:e14357. [PMID: 32026570 PMCID: PMC7002538 DOI: 10.14814/phy2.14357] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Revised: 12/12/2019] [Accepted: 12/27/2019] [Indexed: 01/04/2023] Open
Abstract
To determine whether muscle disuse after a spinal cord injury (SCI) produces elevated markers of cellular senescence and induces markers of the senescence-associated secretory phenotypes (SASPs) in paralyzed skeletal muscle. Four-month-old male Sprague-Dawley rats received a moderate-severe (250 kiloDyne) T-9 contusion SCI or Sham surgery and were monitored over 2 weeks, and 1-, 2-, or 3 months. Animals were sacrificed via isoflurane overdose and terminal exsanguination and the soleus was carefully excised and snap frozen. Protein expression of senescence markers p53, p27, and p16 was determined from whole soleus lysates using Western immunoblotting and RT-qPCR was used to determine the soleus gene expression of IL-1α, IL-1β, IL-6, CXCL1, and TNFα. SCI soleus muscle displayed 2- to 3-fold higher total p53 protein expression at 2 weeks, and at 1 and 2 months when compared with Sham. p27 expression was stable across all groups and timepoints. p16 protein expression was lower at 3 months in SCI versus Sham, but not earlier timepoints. Gene expression was relatively stable between groups at 2 weeks. There were Surgery x Time interaction effects for IL-6 and TNFα mRNA expression but not for IL-1α, IL-1β, or CXCL1. There were no main effects for time or surgery for IL-1α, IL-1β, or CXCL1, but targeted t tests showed reductions in IL-1α and CXCL1 in SCI animals compared to Sham at 3 months and IL-1β was reduced in SCI animals compared to Sham animals at the 2-month timepoint. The elevation in p53 does not appear consistent with the induction of SASP because mRNA expression of cytokines associated with senescence was not uniformly upregulated and, in some instances, was downregulated in the early chronic phase of SCI.
Collapse
Affiliation(s)
- Zachary A. Graham
- Research ServiceBirmingham VA Medical CenterBirminghamALUSA
- Department of Cell, Developmental and Integrative BiologyUniversity of Alabama‐BirminghamBirminghamALUSA
| | - Abigail Goldberger
- Center for the Medical Consequences of Spinal Cord InjuryJames J. Peters VA Medical CenterBronxNYUSA
| | - Daniella Azulai
- Center for the Medical Consequences of Spinal Cord InjuryJames J. Peters VA Medical CenterBronxNYUSA
| | - Christine F. Conover
- Research Service and Brain Rehabilitation Research CenterMalcolm Randall VA Medical CenterNorth Florida/South Georgia Veterans Health SystemGainesvilleFLUSA
| | - Fan Ye
- Research Service and Brain Rehabilitation Research CenterMalcolm Randall VA Medical CenterNorth Florida/South Georgia Veterans Health SystemGainesvilleFLUSA
| | - William A. Bauman
- Center for the Medical Consequences of Spinal Cord InjuryJames J. Peters VA Medical CenterBronxNYUSA
- Icahn School of Medicine at Mount SinaiNew YorkNYUSA
| | - Christopher P. Cardozo
- Center for the Medical Consequences of Spinal Cord InjuryJames J. Peters VA Medical CenterBronxNYUSA
- Icahn School of Medicine at Mount SinaiNew YorkNYUSA
| | - Joshua F. Yarrow
- Research Service and Brain Rehabilitation Research CenterMalcolm Randall VA Medical CenterNorth Florida/South Georgia Veterans Health SystemGainesvilleFLUSA
- Division of Endocrinology, Diabetes, and MetabolismUniversity of Florida College of MedicineGainesvilleFLUSA
| |
Collapse
|
5
|
Bloemberg D, Quadrilatero J. Autophagy, apoptosis, and mitochondria: molecular integration and physiological relevance in skeletal muscle. Am J Physiol Cell Physiol 2019; 317:C111-C130. [PMID: 31017800 DOI: 10.1152/ajpcell.00261.2018] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Apoptosis and autophagy are processes resulting from the integration of cellular stress and death signals. Their individual importance is highlighted by the lethality of various mouse models missing apoptosis or autophagy-related genes. In addition to their independent roles, significant overlap exists with respect to the signals that stimulate these processes as well as their effector consequences. While these cellular systems exemplify the programming redundancies that underlie many fundamental biological mechanisms, their intertwined relationship means that dysfunction can promote pathology. Although both autophagic and apoptotic signaling are active in skeletal muscle during various diseases and atrophy, their specific roles here are somewhat unique. Given our growing understanding of how specific changes at the cellular level impact whole-organism physiology, there is an equally growing interest in pharmacological manipulation of apoptosis and/or autophagy for altering human physiology and health.
Collapse
Affiliation(s)
- Darin Bloemberg
- Department of Kinesiology, University of Waterloo , Waterloo, Ontario , Canada
| | - Joe Quadrilatero
- Department of Kinesiology, University of Waterloo , Waterloo, Ontario , Canada
| |
Collapse
|
6
|
Graham ZA, Harlow L, Bauman WA, Cardozo CP. Alterations in mitochondrial fission, fusion, and mitophagic protein expression in the gastrocnemius of mice after a sciatic nerve transection. Muscle Nerve 2018; 58:592-599. [PMID: 30028528 DOI: 10.1002/mus.26197] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Revised: 06/05/2018] [Accepted: 06/09/2018] [Indexed: 12/13/2022]
Abstract
INTRODUCTION Paralysis and unloading of skeletal muscle leads to a rapid loss in muscle size, function and oxidative capacity. The reduction in metabolic capability after disuse leads to dysregulation and increased breakdown of mitochondria by mitophagy. METHODS Eight-week-old C57BL/6 male mice were given a sham surgery or sciatic nerve transection. Animals were euthanized at 7, 14, 21, or 35 days postsurgery. Whole gastrocnemius muscles were isolated from the animal, weighed and used for Western blotting. RESULTS Markers of mitochondrial fusion were reduced while fission proteins were elevated following a sciatic nerve transection. There were elevations in phosphorylated unc-51-like kinase 1 (ULK1S555 ) and total expression of Beclin1, and of the mitophagy markers PINK1, p62, and microtubule-associated proteins 1A/1B light chain 3b (LC3-II). CONCLUSIONS Paralysis of the gastrocnemius leads to a progressive elevation in expression of mitochondrial fission and mitophagic proteins. Rehabilitative or pharmaceutical interventions to limit excess mitophagy may be effective therapies to protect paralyzed muscle mass and function. Muscle Nerve 58: 592-599, 2018.
Collapse
Affiliation(s)
- Zachary A Graham
- National Center for the Medical Consequences of Spinal Cord Injury, James J. Peters VA Medical Center, 130 West Kingsbridge Road, Bronx, New York, 10468, USA
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Lauren Harlow
- National Center for the Medical Consequences of Spinal Cord Injury, James J. Peters VA Medical Center, 130 West Kingsbridge Road, Bronx, New York, 10468, USA
| | - William A Bauman
- National Center for the Medical Consequences of Spinal Cord Injury, James J. Peters VA Medical Center, 130 West Kingsbridge Road, Bronx, New York, 10468, USA
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Medical Service, James J. Peters VA Medical Center, Bronx, New York, USA
- Department of Rehabilitation Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Christopher P Cardozo
- National Center for the Medical Consequences of Spinal Cord Injury, James J. Peters VA Medical Center, 130 West Kingsbridge Road, Bronx, New York, 10468, USA
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Medical Service, James J. Peters VA Medical Center, Bronx, New York, USA
- Department of Rehabilitation Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| |
Collapse
|
7
|
Cardozo CP. Muscle biology after spinal cord injury: Recent advances and future challenges. Acta Physiol (Oxf) 2018; 223:e13073. [PMID: 29637698 DOI: 10.1111/apha.13073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- C P Cardozo
- Center for the Medical Consequences of Spinal Cord Injury, James J. Peters VA Medical Center, Bronx, NY, USA.,Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
8
|
Lundell LS, Savikj M, Kostovski E, Iversen PO, Zierath JR, Krook A, Chibalin AV, Widegren U. Protein translation, proteolysis and autophagy in human skeletal muscle atrophy after spinal cord injury. Acta Physiol (Oxf) 2018; 223:e13051. [PMID: 29423932 DOI: 10.1111/apha.13051] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2017] [Revised: 01/17/2018] [Accepted: 01/21/2018] [Indexed: 12/14/2022]
Abstract
AIM Spinal cord injury-induced loss of skeletal muscle mass does not progress linearly. In humans, peak muscle loss occurs during the first 6 weeks postinjury, and gradually continues thereafter. The aim of this study was to delineate the regulatory events underlying skeletal muscle atrophy during the first year following spinal cord injury. METHODS Key translational, autophagic and proteolytic proteins were analysed by immunoblotting of human vastus lateralis muscle obtained 1, 3 and 12 months following spinal cord injury. Age-matched able-bodied control subjects were also studied. RESULTS Several downstream targets of Akt signalling decreased after spinal cord injury in skeletal muscle, without changes in resting Akt Ser473 and Akt Thr308 phosphorylation or total Akt protein. Abundance of mTOR protein and mTOR Ser2448 phosphorylation, as well as FOXO1 Ser256 phosphorylation and FOXO3 protein, decreased in response to spinal cord injury, coincident with attenuated protein abundance of E3 ubiquitin ligases, MuRF1 and MAFbx. S6 protein and Ser235/236 phosphorylation, as well as 4E-BP1 Thr37/46 phosphorylation, increased transiently after spinal cord injury, indicating higher levels of protein translation early after injury. Protein abundance of LC3-I and LC3-II decreased 3 months postinjury as compared with 1 month postinjury, but not compared to able-bodied control subjects, indicating lower levels of autophagy. Proteins regulating proteasomal degradation were stably increased in response to spinal cord injury. CONCLUSION Together, these data provide indirect evidence suggesting that protein translation and autophagy transiently increase, while whole proteolysis remains stably higher in skeletal muscle within the first year after spinal cord injury.
Collapse
Affiliation(s)
- L. S. Lundell
- Department of Physiology and Pharmacology; Section for Integrative Physiology; Karolinska Institutet; Stockholm Sweden
| | - M. Savikj
- Department of Physiology and Pharmacology; Section for Integrative Physiology; Karolinska Institutet; Stockholm Sweden
- Faculty of Medicine; University of Oslo; Oslo Norway
- Department of Research; Sunnaas Rehabilitation Hospital; Nesoddtangen Norway
| | - E. Kostovski
- Faculty of Medicine; University of Oslo; Oslo Norway
- Department of Research; Sunnaas Rehabilitation Hospital; Nesoddtangen Norway
| | - P. O. Iversen
- Department of Nutrition; Institute of Basic Medical Sciences; University of Oslo; Oslo Norway
- Department of Hematology; Oslo University Hospital; Oslo Norway
| | - J. R. Zierath
- Department of Physiology and Pharmacology; Section for Integrative Physiology; Karolinska Institutet; Stockholm Sweden
- Department of Molecular Medicine and Surgery; Section for Integrative Physiology; Karolinska Institutet; Stockholm Sweden
| | - A. Krook
- Department of Physiology and Pharmacology; Section for Integrative Physiology; Karolinska Institutet; Stockholm Sweden
| | - A. V. Chibalin
- Department of Molecular Medicine and Surgery; Section for Integrative Physiology; Karolinska Institutet; Stockholm Sweden
| | - U. Widegren
- Department of Molecular Medicine and Surgery; Section for Integrative Physiology; Karolinska Institutet; Stockholm Sweden
| |
Collapse
|
9
|
Pan Y, Yang XH, Guo LL, Gu YH, Qiao QY, Jin HM. Erythropoietin Reduces Insulin Resistance via Regulation of Its Receptor-Mediated Signaling Pathways in db/db Mice Skeletal Muscle. Int J Biol Sci 2017; 13:1329-1340. [PMID: 29104499 PMCID: PMC5666531 DOI: 10.7150/ijbs.19752] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Accepted: 08/08/2017] [Indexed: 01/09/2023] Open
Abstract
Erythropoietin (EPO) can reduce insulin resistance (IR) in adipocytes; however, it is unknown whether EPO can decrease IR in skeletal muscle. Here we investigated whether EPO could reduce IR in type 2 diabetic mouse skeletal muscle and its possible signaling mechanisms of action. Twelve-week-old db/db diabetic mice were employed in this study. Systemic use of EPO improved glucose profiles in type 2 diabetic mice after 4 and 8 weeks treatment. EPO up-regulated EPOR protein expression in skeletal muscle, and subsequently activated downstream signaling molecules such as JAK2, IRS-1, PI3K, AKT, and eNOS. We next constructed lentivirally-delivered shRNAs against EPOR and transfected skeletal muscle cells to knockdown EPOR. EPOR knockdown inhibited EPO induced JAK2, IRS-1, PI3K, AKT, eNOS signaling transduction, autophagy and Glut 4 translocation, as well as promoted apoptosis in skeletal muscle. Thus, EPO reduces skeletal muscle IR in type 2 diabetic mice via its specific receptor, EPOR. Possible mechanisms involved in its action may include increased autophagy and reduced apoptosis in type 2 diabetic skeletal muscles, which provides a new strategy for the treatment of IR.
Collapse
Affiliation(s)
- Yu Pan
- Division of Nephrology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiu Hong Yang
- Division of Nephrology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China
| | - Li Li Guo
- Hemodialysis Center, Baoshan Branch of Shanghai No.1 People's Hospital, Shanghai, China
| | - Yan Hong Gu
- Division of Nephrology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China
| | - Qing Yan Qiao
- Division of Nephrology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China
| | - Hui Min Jin
- Division of Nephrology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China
| |
Collapse
|
10
|
Brocca L, Longa E, Cannavino J, Seynnes O, de Vito G, McPhee J, Narici M, Pellegrino MA, Bottinelli R. Human skeletal muscle fibre contractile properties and proteomic profile: adaptations to 3 weeks of unilateral lower limb suspension and active recovery. J Physiol 2016; 593:5361-85. [PMID: 26369674 DOI: 10.1113/jp271188] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2015] [Accepted: 09/04/2015] [Indexed: 01/08/2023] Open
Abstract
KEY POINTS It is generally assumed that muscle fibres go through atrophy following disuse with a loss of specific force and an increase in unloaded shortening velocity. However, the underlying mechanisms remain to be clarified. Most studies have focused on events taking place during the development of disuse, whereas the subsequent recovery phase, which is equally important, has received little attention. Our findings support the hypotheses that the specific force of muscle fibres decreased following unilateral lower limb suspension (ULLS) and returned to normal after 3 weeks of active recovery as a result of a loss and recovery of myosin and actin content. Furthermore, muscle fibres went through extensive qualitative changes in muscle protein pattern following ULLS, and these were reversed by active recovery. Resistance training was very effective in restoring both muscle mass and qualitative muscle changes, indicating that long-term ULLS did not prevent the positive effect of exercise on human muscle. ABSTRACT Following disuse, muscle fibre function goes through adaptations such as a loss of specific force (PO /CSA) and an increase in unloaded shortening velocity, which could be a result of both quantitative changes (i.e. atrophy) and qualitative changes in protein pattern. The underlying mechanisms remain to be clarified. In addition, little is known about the recovery of muscle mass and strength following disuse. In the present study, we report an extensive dataset describing, in detail,the functional and protein content adaptations of skeletal muscle in response to both disuse and re-training. Eight young healthy subjects were subjected to 3 weeks of unilateral lower limb suspension (ULLS), a widely used human model of disuse skeletal muscle atrophy. Needle biopsies samples were taken from the vastus lateralis muscle Pre-ULLS, Post-ULLS and after 3 weeks of recovery during which heavy resistance training was performed. After disuse, cross-sectional area (CSA), PO /CSA and myosin concentration (MC) decreased in both type 1 and 2A skinned muscle fibres. After recovery, CSA and MC returned to levels comparable to those observed before disuse, whereas Po/CSA and unloaded shortening velocity reached a higher level. Myosin heavy chain isoform composition of muscle samples did not differ among the experimental groups. To study the mechanisms underlying such adaptations, a two-dimensional proteomic analysis was performed. ULLS induced a reduction of myofibrillar, metabolic (glycolytic and oxidative) and anti-oxidant defence system protein content. Resistance training was very effective in counteracting ULLS-induced alterations, indicating that long-term ULLS did not prevent the positive effect of exercise on human muscle.
Collapse
Affiliation(s)
- Lorenza Brocca
- Department of Molecular Medicine, University of Pavia, Pavia, Italy.,Interuniversity Institute of Myology, University of Pavia, Pavia, Italy
| | - Emanuela Longa
- Department of Molecular Medicine, University of Pavia, Pavia, Italy.,Interdipartimental Centre for Biology and Sport Medicine, University of Pavia, Pavia, Italy
| | | | - Olivier Seynnes
- Institute for Biomedical Research into Human Movement and Health, Manchester Metropolitan University, Manchester, UK.,Norwegian School of Sport Sciences, Oslo, Norway
| | - Giuseppe de Vito
- UCD Institute for Sport and Health, University College Dublin, Dublin, Ireland
| | - Jamie McPhee
- Institute for Biomedical Research into Human Movement and Health, Manchester Metropolitan University, Manchester, UK
| | - Marco Narici
- Institute for Biomedical Research into Human Movement and Health, Manchester Metropolitan University, Manchester, UK.,School of Graduate Entry to Medicine and Health, Division of Clinical Physiology, University of Nottingham, Derby, UK
| | - Maria Antonietta Pellegrino
- Department of Molecular Medicine, University of Pavia, Pavia, Italy.,Interdipartimental Centre for Biology and Sport Medicine, University of Pavia, Pavia, Italy.,Interuniversity Institute of Myology, University of Pavia, Pavia, Italy
| | - Roberto Bottinelli
- Department of Molecular Medicine, University of Pavia, Pavia, Italy.,Fondazione Salvatore Maugeri (IRCCS), Scientific Institute of Pavia, Pavia, Italy
| |
Collapse
|
11
|
Konopka AR, Sreekumaran Nair K. Mitochondrial and skeletal muscle health with advancing age. Mol Cell Endocrinol 2013; 379:19-29. [PMID: 23684888 PMCID: PMC3788080 DOI: 10.1016/j.mce.2013.05.008] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2013] [Revised: 04/22/2013] [Accepted: 05/08/2013] [Indexed: 12/21/2022]
Abstract
With increasing age there is a temporal relationship between the decline of mitochondrial and skeletal muscle volume, quality and function (i.e., health). Reduced mitochondrial mRNA expression, protein abundance, and protein synthesis rates appear to promote the decline of mitochondrial protein quality and function. Decreased mitochondrial function is suspected to impede energy demanding processes such as skeletal muscle protein turnover, which is critical for maintaining protein quality and thus skeletal muscle health with advancing age. The focus of this review was to discuss promising human physiological systems underpinning the decline of mitochondrial and skeletal muscle health with advancing age while highlighting therapeutic strategies such as aerobic exercise and caloric restriction for combating age-related functional impairments.
Collapse
Affiliation(s)
- Adam R Konopka
- Endocrine Research Unit, Mayo Clinic College of Medicine, Rochester, Minnesota, United States
| | | |
Collapse
|
12
|
Talbert EE, Smuder AJ, Min K, Kwon OS, Szeto HH, Powers SK. Immobilization-induced activation of key proteolytic systems in skeletal muscles is prevented by a mitochondria-targeted antioxidant. J Appl Physiol (1985) 2013; 115:529-38. [PMID: 23766499 DOI: 10.1152/japplphysiol.00471.2013] [Citation(s) in RCA: 104] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Long periods of skeletal muscle disuse result in muscle fiber atrophy, and mitochondrial production of reactive oxygen species (ROS) appears to be a required signal for the increase in protein degradation that occurs during disuse muscle atrophy. The experiments detailed here demonstrate for the first time in limb muscle that the inactivity-induced increases in E3 ligase expression and autophagy biomarkers result from increases in mitochondrial ROS emission. Treatment of animals with a mitochondrial-targeted antioxidant also prevented the disuse-induced decrease in anabolic signaling (Akt/mammalian target of rapamycin signaling) that is normally associated with prolonged inactivity in skeletal muscles. Additionally, our results confirm previous findings that treatment with a mitochondrial-targeted antioxidant is sufficient to prevent casting-induced skeletal muscle atrophy, mitochondrial dysfunction, and activation of the proteases calpain and caspase-3. Collectively, these data reveal that inactivity-induced increases in mitochondrial ROS emission play a required role in activation of key proteolytic systems and the downregulation of important anabolic signaling molecules in muscle fibers exposed to prolonged inactivity.
Collapse
|