1
|
Palomer X, Salvador JM, Griñán-Ferré C, Barroso E, Pallàs M, Vázquez-Carrera M. GADD45A: With or without you. Med Res Rev 2024; 44:1375-1403. [PMID: 38264852 DOI: 10.1002/med.22015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 12/11/2023] [Accepted: 01/09/2024] [Indexed: 01/25/2024]
Abstract
The growth arrest and DNA damage inducible (GADD)45 family includes three small and ubiquitously distributed proteins (GADD45A, GADD45B, and GADD45G) that regulate numerous cellular processes associated with stress signaling and injury response. Here, we provide a comprehensive review of the current literature investigating GADD45A, the first discovered member of the family. We first depict how its levels are regulated by a myriad of genotoxic and non-genotoxic stressors, and through the combined action of intricate transcriptional, posttranscriptional, and even, posttranslational mechanisms. GADD45A is a recognized tumor suppressor and, for this reason, we next summarize its role in cancer, as well as the different mechanisms by which it regulates cell cycle, DNA repair, and apoptosis. Beyond these most well-known actions, GADD45A may also influence catabolic and anabolic pathways in the liver, adipose tissue and skeletal muscle, among others. Not surprisingly, GADD45A may trigger AMP-activated protein kinase activity, a master regulator of metabolism, and is known to act as a transcriptional coregulator of numerous nuclear receptors. GADD45A has also been reported to display a cytoprotective role by regulating inflammation, fibrosis and oxidative stress in several organs and tissues, and is regarded an important contributor for the development of heart failure. Overall data point to that GADD45A may play an important role in metabolic, neurodegenerative and cardiovascular diseases, and also autoimmune-related disorders. Thus, the potential mechanisms by which dysregulation of GADD45A activity may contribute to the progression of these diseases are also reviewed below.
Collapse
Affiliation(s)
- Xavier Palomer
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Sciences, University of Barcelona, Barcelona, Spain
- Institute of Biomedicine of the University of Barcelona (IBUB), University of Barcelona, Barcelona, Spain
- Spanish Biomedical Research Center in Diabetes and Associated Metabolic Diseases (CIBERDEM)-Instituto de Salud Carlos III, Madrid, Spain
- Pediatric Research Institute-Hospital Sant Joan de Déu, Esplugues de Llobregat, Barcelona, Spain
| | - Jesús M Salvador
- Department of Immunology and Oncology, Centro Nacional de Biotecnología/CSIC, Madrid, Spain
| | - Christian Griñán-Ferré
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Sciences, University of Barcelona, Barcelona, Spain
- Institut de Neurociències, Universitat de Barcelona (NeuroUB), Barcelona, Spain
- Spanish Biomedical Research Center in Neurodegenerative Diseases (CIBERNED)-Instituto de Salud Carlos III, Madrid, Spain
| | - Emma Barroso
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Sciences, University of Barcelona, Barcelona, Spain
- Institute of Biomedicine of the University of Barcelona (IBUB), University of Barcelona, Barcelona, Spain
- Spanish Biomedical Research Center in Diabetes and Associated Metabolic Diseases (CIBERDEM)-Instituto de Salud Carlos III, Madrid, Spain
- Pediatric Research Institute-Hospital Sant Joan de Déu, Esplugues de Llobregat, Barcelona, Spain
| | - Mercè Pallàs
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Sciences, University of Barcelona, Barcelona, Spain
- Institut de Neurociències, Universitat de Barcelona (NeuroUB), Barcelona, Spain
- Spanish Biomedical Research Center in Neurodegenerative Diseases (CIBERNED)-Instituto de Salud Carlos III, Madrid, Spain
| | - Manuel Vázquez-Carrera
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Sciences, University of Barcelona, Barcelona, Spain
- Institute of Biomedicine of the University of Barcelona (IBUB), University of Barcelona, Barcelona, Spain
- Spanish Biomedical Research Center in Diabetes and Associated Metabolic Diseases (CIBERDEM)-Instituto de Salud Carlos III, Madrid, Spain
- Pediatric Research Institute-Hospital Sant Joan de Déu, Esplugues de Llobregat, Barcelona, Spain
| |
Collapse
|
2
|
Harrigan ME, Filous AR, Vadala CP, Webb A, Pietrzak M, Sahenk Z, Prüss H, Reiser PJ, Popovich PG, Arnold WD, Schwab JM. Lesion level-dependent systemic muscle wasting after spinal cord injury is mediated by glucocorticoid signaling in mice. Sci Transl Med 2023; 15:eadh2156. [PMID: 38117902 DOI: 10.1126/scitranslmed.adh2156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Accepted: 11/29/2023] [Indexed: 12/22/2023]
Abstract
An incomplete mechanistic understanding of skeletal muscle wasting early after spinal cord injury (SCI) precludes targeted molecular interventions. Here, we demonstrated systemic wasting that also affected innervated nonparalyzed (supralesional) muscles and emerged within 1 week after experimental SCI in mice. Systemic muscle wasting caused muscle weakness, affected fast type 2 myofibers preferentially, and became exacerbated after high (T3) compared with low (T9) thoracic paraplegia, indicating lesion level-dependent ("neurogenic") mechanisms. The wasting of nonparalyzed muscle and its rapid onset and severity beyond what can be explained by disuse implied unknown systemic drivers. Muscle transcriptome and biochemical analysis revealed a glucocorticoid-mediated catabolic signature early after T3 SCI. SCI-induced systemic muscle wasting was mitigated by (i) endogenous glucocorticoid ablation (adrenalectomy) and (ii) pharmacological glucocorticoid receptor (GR) blockade and was (iii) completely prevented after T3 relative to T9 SCI by genetic muscle-specific GR deletion. These results suggest that neurogenic hypercortisolism contributes to a rapid systemic and functionally relevant muscle wasting syndrome early after paraplegic SCI in mice.
Collapse
Affiliation(s)
- Markus E Harrigan
- Department of Neurology, Spinal Cord Injury Division (Paraplegiology), College of Medicine, Ohio State University, Wexner Medical Center, Columbus, OH 43210, USA
- Medical Scientist Training Program, College of Medicine, Ohio State University, Wexner Medical Center, Columbus, OH 43210, USA
- Neuroscience Research Institute, Ohio State University, Columbus, OH 43210, USA
- Belford Center for Spinal Cord Injury, Ohio State University, Wexner Medical Center, Columbus, OH 43210, USA
| | - Angela R Filous
- Department of Neurology, Spinal Cord Injury Division (Paraplegiology), College of Medicine, Ohio State University, Wexner Medical Center, Columbus, OH 43210, USA
- Neuroscience Research Institute, Ohio State University, Columbus, OH 43210, USA
- Belford Center for Spinal Cord Injury, Ohio State University, Wexner Medical Center, Columbus, OH 43210, USA
| | - Christopher P Vadala
- Department of Neurology, Spinal Cord Injury Division (Paraplegiology), College of Medicine, Ohio State University, Wexner Medical Center, Columbus, OH 43210, USA
- Neuroscience Research Institute, Ohio State University, Columbus, OH 43210, USA
- Belford Center for Spinal Cord Injury, Ohio State University, Wexner Medical Center, Columbus, OH 43210, USA
| | - Amy Webb
- Department of Biomedical Informatics, College of Medicine, Ohio State University, Wexner Medical Center, Columbus, OH 43210, USA
| | - Maciej Pietrzak
- Department of Biomedical Informatics, College of Medicine, Ohio State University, Wexner Medical Center, Columbus, OH 43210, USA
| | - Zarife Sahenk
- Center for Gene Therapy, Research Institute at Nationwide Children's Hospital, Columbus, OH 43205, USA
- Department of Pediatrics and Neurology, Nationwide Children's Hospital and Ohio State University, Columbus, OH 43205, USA
- Department of Pathology and Laboratory Medicine, Nationwide Children's Hospital, Columbus, OH 43205, USA
| | - Harald Prüss
- Department of Neurology and Experimental Neurology, Charité-Universitätsmedizin, 10117 Berlin, Germany
- German Center for Neurodegenerative Diseases (DZNE), 10117 Berlin, Germany
| | - Peter J Reiser
- Division of Biosciences, College of Dentistry, Ohio State University, Columbus, OH 43210, USA
| | - Phillip G Popovich
- Neuroscience Research Institute, Ohio State University, Columbus, OH 43210, USA
- Belford Center for Spinal Cord Injury, Ohio State University, Wexner Medical Center, Columbus, OH 43210, USA
- Department of Neuroscience, Ohio State University, Wexner Medical Center, Columbus, OH 43210, USA
| | - W David Arnold
- NextGen Precision Health, University of Missouri, Columbia, MO 65211, USA
- Department of Physical Medicine and Rehabilitation, University of Missouri, Columbia, MO 65212, USA
| | - Jan M Schwab
- Department of Neurology, Spinal Cord Injury Division (Paraplegiology), College of Medicine, Ohio State University, Wexner Medical Center, Columbus, OH 43210, USA
- Neuroscience Research Institute, Ohio State University, Columbus, OH 43210, USA
- Belford Center for Spinal Cord Injury, Ohio State University, Wexner Medical Center, Columbus, OH 43210, USA
- Department of Neuroscience, Ohio State University, Wexner Medical Center, Columbus, OH 43210, USA
- Department of Physical Medicine and Rehabilitation, Ohio State University, Wexner Medical Center, Columbus, OH 43210, USA
| |
Collapse
|
3
|
Xu X, Du HY, Talifu Z, Zhang CJ, Li ZH, Liu WB, Liang YX, Xu XL, Zhang JM, Yang DG, Gao F, Du LJ, Yu Y, Jing YL, Li JJ. Glycine and N-Acetylcysteine (GlyNAC) Combined with Body Weight Support Treadmill Training Improved Spinal Cord and Skeletal Muscle Structure and Function in Rats with Spinal Cord Injury. Nutrients 2023; 15:4578. [PMID: 37960231 PMCID: PMC10649910 DOI: 10.3390/nu15214578] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 10/13/2023] [Accepted: 10/25/2023] [Indexed: 11/15/2023] Open
Abstract
Skeletal muscle atrophy is a frequent complication after spinal cord injury (SCI) and can influence the recovery of motor function and metabolism in affected patients. Delaying skeletal muscle atrophy can promote functional recovery in SCI rats. In the present study, we investigated whether a combination of body weight support treadmill training (BWSTT) and glycine and N-acetylcysteine (GlyNAC) could exert neuroprotective effects, promote motor function recovery, and delay skeletal muscle atrophy in rats with SCI, and we assessed the therapeutic effects of the double intervention from both a structural and functional viewpoint. We found that, after SCI, rats given GlyNAC alone showed an improvement in Basso-Beattie-Bresnahan (BBB) scores, gait symmetry, and results in the open field test, indicative of improved motor function, while GlyNAC combined with BWSTT was more effective than either treatment alone at ameliorating voluntary motor function in injured rats. Meanwhile, the results of the skeletal muscle myofiber cross-sectional area (CSA), hindlimb grip strength, and acetylcholinesterase (AChE) immunostaining analysis demonstrated that GlyNAC improved the structure and function of the skeletal muscle in rats with SCI and delayed the atrophication of skeletal muscle.
Collapse
Affiliation(s)
- Xin Xu
- School of Rehabilitation, Capital Medical University, Beijing 100069, China; (X.X.)
| | - Hua-Yong Du
- School of Rehabilitation, Capital Medical University, Beijing 100069, China; (X.X.)
| | - Zuliyaer Talifu
- School of Population Medicine and Public Health, Chinese Academy of Medical Sciences/Peking Union Medical College, Beijing 100730, China
| | - Chun-Jia Zhang
- School of Rehabilitation, Capital Medical University, Beijing 100069, China; (X.X.)
| | - Ze-Hui Li
- School of Rehabilitation, Capital Medical University, Beijing 100069, China; (X.X.)
| | - Wu-Bo Liu
- Department of Orthopedics, Qilu Hospital of Shandong University, Jinan 250100, China
| | - Yi-Xiong Liang
- School of Rehabilitation, Capital Medical University, Beijing 100069, China; (X.X.)
| | - Xu-Luan Xu
- School of Rehabilitation, Capital Medical University, Beijing 100069, China; (X.X.)
| | - Jin-Ming Zhang
- School of Rehabilitation, Capital Medical University, Beijing 100069, China; (X.X.)
| | - De-Gang Yang
- School of Rehabilitation, Capital Medical University, Beijing 100069, China; (X.X.)
| | - Feng Gao
- School of Rehabilitation, Capital Medical University, Beijing 100069, China; (X.X.)
| | - Liang-Jie Du
- School of Rehabilitation, Capital Medical University, Beijing 100069, China; (X.X.)
| | - Yan Yu
- School of Rehabilitation, Capital Medical University, Beijing 100069, China; (X.X.)
| | - Ying-Li Jing
- School of Rehabilitation, Capital Medical University, Beijing 100069, China; (X.X.)
| | - Jian-Jun Li
- School of Rehabilitation, Capital Medical University, Beijing 100069, China; (X.X.)
| |
Collapse
|
4
|
Martín AI, Moreno-Rupérez Á, Nebot E, Granado M, Jaque D, Nieto-Bona MP, López-Calderón A, Priego T. Time-Dependent Changes in Muscle IGF1-IGFBP5-PAPP System after Sciatic Denervation. Int J Mol Sci 2023; 24:14112. [PMID: 37762414 PMCID: PMC10531309 DOI: 10.3390/ijms241814112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 09/08/2023] [Accepted: 09/11/2023] [Indexed: 09/29/2023] Open
Abstract
Denervation-induced muscle atrophy is a frequent cause of skeletal muscle diseases. However, the role of the most important muscle growth factor, insulin-like growth factor (IGF-1), in this process is poorly understood. IGF-1 activity is controlled by six IGF-1 binding proteins (IGFBPs). In skeletal muscle, IGFBP-5 seems to have an important role in atrophic processes. Furthermore, pappalysins (PAPP-A) modulate muscle growth by increasing IGF-1 bioavailability through IGFBP cleavage. We aimed to study the time-dependent changes in the IGF1-IGFBP5-PAPP system and its regulators in gastrocnemius muscle after sciatic denervation. Gastrocnemius atrophy and overexpression of IGF-1 was observed from day 3 post-denervation. The proteolytic factors measured were elevated from day 1 post-denervation onwards. Expression of both IGFBP-5 and pappalysins were increased on days 1 and 3. Subsequently, on days 7 to 14 pappalysins returned to control levels while IGFBP-5 remained elevated. The ratio IGFBP-5/PAPP-A was correlated with the main proteolytic markers. All data suggest that the initial increase of pappalysins could facilitate the IGF-1 action on muscle growth, whereas their subsequent decrease could lead to further muscle wasting.
Collapse
Affiliation(s)
- Ana Isabel Martín
- Departamento de Fisiología, Facultad de Medicina, Universidad Complutense de Madrid, Plaza de Ramón y Cajal sn, 28040 Madrid, Spain; (A.I.M.); (Á.M.-R.); (E.N.); (A.L.-C.)
| | - Álvaro Moreno-Rupérez
- Departamento de Fisiología, Facultad de Medicina, Universidad Complutense de Madrid, Plaza de Ramón y Cajal sn, 28040 Madrid, Spain; (A.I.M.); (Á.M.-R.); (E.N.); (A.L.-C.)
| | - Elena Nebot
- Departamento de Fisiología, Facultad de Medicina, Universidad Complutense de Madrid, Plaza de Ramón y Cajal sn, 28040 Madrid, Spain; (A.I.M.); (Á.M.-R.); (E.N.); (A.L.-C.)
| | - Miriam Granado
- Departamento de Fisiología, Facultad de Medicina, Universidad Autónoma de Madrid, Calle Arzobispo Morcillo 2, 28029 Madrid, Spain;
| | - Daniel Jaque
- Nanomaterials for Bioimaging Group (NanoBIG), Departamento de Física de Materiales, Facultad de Ciencias, Universidad Autónoma de Madrid, Avenida Francisco Tomas y Valiente, 28049 Madrid, Spain;
| | - M. Paz Nieto-Bona
- Departamento de Ciencias Básicas de la Salud, Facultad CC Salud, Universidad Rey Juan Carlos, Avenida de Atenas sn, 20922 Madrid, Spain;
| | - Asunción López-Calderón
- Departamento de Fisiología, Facultad de Medicina, Universidad Complutense de Madrid, Plaza de Ramón y Cajal sn, 28040 Madrid, Spain; (A.I.M.); (Á.M.-R.); (E.N.); (A.L.-C.)
| | - Teresa Priego
- Departamento de Fisiología, Facultad de Enfermería, Fisioterapia y Podología, Universidad Complutense de Madrid, Plaza de Ramón y Cajal sn, 28040 Madrid, Spain
| |
Collapse
|
5
|
Colasuonno F, Price R, Moreno S. Upper and Lower Motor Neurons and the Skeletal Muscle: Implication for Amyotrophic Lateral Sclerosis (ALS). ADVANCES IN ANATOMY, EMBRYOLOGY, AND CELL BIOLOGY 2023; 236:111-129. [PMID: 37955773 DOI: 10.1007/978-3-031-38215-4_5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/14/2023]
Abstract
The relationships between motor neurons and the skeletal muscle during development and in pathologic contexts are addressed in this Chapter.We discuss the developmental interplay of muscle and nervous tissue, through neurotrophins and the activation of differentiation and survival pathways. After a brief overview on muscular regulatory factors, we focus on the contribution of muscle to early and late neurodevelopment. Such a role seems especially intriguing in relation to the epigenetic shaping of developing motor neuron fate choices. In this context, emphasis is attributed to factors regulating energy metabolism, which may concomitantly act in muscle and neural cells, being involved in common pathways.We then review the main features of motor neuron diseases, addressing the cellular processes underlying clinical symptoms. The involvement of different muscle-associated neurotrophic factors for survival of lateral motor column neurons, innervating MyoD-dependent limb muscles, and of medial motor column neurons, innervating Myf5-dependent back musculature is discussed. Among the pathogenic mechanisms, we focus on oxidative stress, that represents a common and early trait in several neurodegenerative disorders. The role of organelles primarily involved in reactive oxygen species scavenging and, more generally, in energy metabolism-namely mitochondria and peroxisomes-is discussed in the frame of motor neuron degeneration.We finally address muscular involvement in amyotrophic lateral sclerosis (ALS), a multifactorial degenerative disorder, hallmarked by severe weight loss, caused by imbalanced lipid metabolism. Even though multiple mechanisms have been recognized to play a role in the disease, current literature generally assumes that the primum movens is neuronal degeneration and that muscle atrophy is only a consequence of such pathogenic event. However, several lines of evidence point to the muscle as primarily involved in the disease, mainly through its role in energy homeostasis. Data from different ALS mouse models strongly argue for an early mitochondrial dysfunction in muscle tissue, possibly leading to motor neuron disturbances. Detailed understanding of skeletal muscle contribution to ALS pathogenesis will likely lead to the identification of novel therapeutic strategies.
Collapse
Affiliation(s)
- Fiorella Colasuonno
- Department of Experimental Medicine , University of Rome "Tor Vergata", Rome, Italy
- Department of Science, LIME, University Roma Tre, Rome, Italy
| | - Rachel Price
- Department of Science, LIME, University Roma Tre, Rome, Italy
- Laboratory of Neurodevelopmental Biology, Neurogenetics and Molecular Neurobiology, IRCCS Fondazione Santa Lucia, Rome, Italy
| | - Sandra Moreno
- Department of Science, LIME, University Roma Tre, Rome, Italy.
- Laboratory of Neurodevelopmental Biology, Neurogenetics and Molecular Neurobiology, IRCCS Fondazione Santa Lucia, Rome, Italy.
| |
Collapse
|
6
|
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: 4.0] [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
|
7
|
Li H, Yuan W, Chen Y, Lin B, Wang S, Deng Z, Zheng Q, Li Q. Transcription and proteome changes involved in re-innervation muscle following nerve crush in rats. BMC Genomics 2022; 23:666. [PMID: 36131238 PMCID: PMC9494802 DOI: 10.1186/s12864-022-08895-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Accepted: 09/16/2022] [Indexed: 02/05/2023] Open
Abstract
Severe peripheral nerve injury leads to the irreparable disruption of nerve fibers. This leads to disruption of synapses with the designated muscle, which consequently go through progressive atrophy and damage of muscle function. The molecular mechanism that underlies the re-innervation process has yet to be evaluated using proteomics or transcriptomics. In the present study, multi-dimensional data were therefore integrated with transcriptome and proteome profiles in order to investigate the mechanism of re-innervation in muscles. Two simulated nerve injury muscle models in the rat tibial nerve were compared: the nerve was either cut (denervated, DN group) or crushed but with the nerve sheath intact (re-innervated, RN group). The control group had a preserved and intact tibial nerve. At 4 weeks, the RN group showed better tibial nerve function and recovery of muscle atrophy compared to the DN group. As the high expression of Myh3, Postn, Col6a1 and Cfi, the RN group demonstrated superior re-innervation as well. Both differentially expressed genes (DEGs) and proteins (DEPs) were enriched in the peroxisome proliferator-activated receptors (PPARs) signaling pathway, as well as the energy metabolism. This study provides basic information regarding DEGs and DEPs during re-innervation-induced muscle atrophy. Furthermore, the crucial genes and proteins can be detected as possible treatment targets in the future.
Collapse
Affiliation(s)
- Haotao Li
- Department of Orthopedics, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, 106, Zhongshan Road, Yuexiu District, Guangzhou, People's Republic of China
- Shantou University Medical College, Shantou, People's Republic of China
| | - Wanqiong Yuan
- Department of Orthopedics, Peking University Third Hospital, Beijing, People's Republic of China
- Beijing Key Laboratory of Spinal Disease, Beijing, People's Republic of China
- Engineering Research Center of Bone and Joint Precision Medicine, Beijing, People's Republic of China
| | - Yijian Chen
- Department of Orthopedics, Shantou Central Hospital, Shantou, Guangdong, People's Republic of China
| | - Bofu Lin
- Department of Orthopedics, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, 106, Zhongshan Road, Yuexiu District, Guangzhou, People's Republic of China
- Shantou University Medical College, Shantou, People's Republic of China
| | - Shuai Wang
- Department of Orthopedics, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, 106, Zhongshan Road, Yuexiu District, Guangzhou, People's Republic of China
| | - Zhantao Deng
- Department of Orthopedics, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, 106, Zhongshan Road, Yuexiu District, Guangzhou, People's Republic of China
| | - Qiujian Zheng
- Department of Orthopedics, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, 106, Zhongshan Road, Yuexiu District, Guangzhou, People's Republic of China
| | - Qingtian Li
- Department of Orthopedics, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, 106, Zhongshan Road, Yuexiu District, Guangzhou, People's Republic of China.
| |
Collapse
|
8
|
Laskin GR, Gordon BS. The influence of nutrients on mechanical overload-induced changes to skeletal muscle mRNA content. Physiol Genomics 2022; 54:360-369. [PMID: 35848636 DOI: 10.1152/physiolgenomics.00075.2022] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Mechanical overload and nutrients influence skeletal muscle phenotype, with the combination sometimes having a synergistic effect. Muscle phenotypes influenced by these stimuli are mediated in part by changes to the muscle mRNA signature. However, the mechanical overload-sensitive gene programs that are influenced by nutrients remain unclear. The purpose of this study was to identify mechanical overload-sensitive gene programs that are influenced by nutrients and identify potential transcription factors that may differentiate the change in mRNA in response to mechanical overload versus nutrients. Nutrient deprived 12-week-old male mice were randomized to remain fasted or allowed access to food. All mice underwent a single bout of unilateral high force contractions of the tibialis anterior (TA). Four hours post-contractions TA muscles were extracted and content of 12 contraction-sensitive mRNAs were analyzed. The mRNA content of genes associated with Transcription, PI3K-Akt Signaling Pathway, Z-Disc, Intracellular Signal Transduction, Cell Cycle, and Amino Acid Transport was altered by contractions without influence of nutrient consumption. Conversely, the mRNA content of genes associated with Transcription, Cell Cycle, FoxO Signaling Pathway, and Amino Acid Transport was altered by contractions with nutrition consumption influencing the change. We identified Signal transducer and activator of transcription 3 (STAT3) and Activator protein 1 (AP-1) as transcription factors common amongst mRNAs that were primarily altered by mechanical overload regardless of feeding. Overall, these data provide a deeper molecular basis for the specific muscle phenotypes exclusive to mechanical overload versus those regulated by the addition of nutrients.
Collapse
Affiliation(s)
- Grant R Laskin
- Department of Nutrition and Integrative Physiology, Florida State University, Tallahassee, FL, United States
| | - Bradley S Gordon
- Department of Nutrition and Integrative Physiology, Florida State University, Tallahassee, FL, United States.,Institute of Sports Sciences and Medicine, Florida State University, Tallahassee, FL, United States
| |
Collapse
|
9
|
Sultan FA, Sawaya BE. Gadd45 in Neuronal Development, Function, and Injury. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1360:117-148. [PMID: 35505167 DOI: 10.1007/978-3-030-94804-7_9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
The growth arrest and DNA damage-inducible (Gadd) 45 proteins have been associated with numerous cellular mechanisms including cell cycle control, DNA damage sensation and repair, genotoxic stress, neoplasia, and molecular epigenetics. The genes were originally identified in in vitro screens of irradiation- and interleukin-induced transcription and have since been implicated in a host of normal and aberrant central nervous system processes. These include early and postnatal development, injury, cancer, memory, aging, and neurodegenerative and psychiatric disease states. The proteins act through a variety of molecular signaling cascades including the MAPK cascade, cell cycle control mechanisms, histone regulation, and epigenetic DNA demethylation. In this review, we provide a comprehensive discussion of the literature implicating each of the three members of the Gadd45 family in these processes.
Collapse
Affiliation(s)
- Faraz A Sultan
- Department of Psychiatry, Rush University, Chicago, IL, USA.
| | - Bassel E Sawaya
- Molecular Studies of Neurodegenerative Diseases Lab, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA.,FELS Cancer Institute for Personalized Medicine Institute, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA.,Departments of Neurology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA.,Cancer and Cell Biology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA.,Neural Sciences, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| |
Collapse
|
10
|
Hook MA, Falck A, Dundumulla R, Terminel M, Cunningham R, Sefiani A, Callaway K, Gaddy D, Geoffroy CG. Osteopenia in a Mouse Model of Spinal Cord Injury: Effects of Age, Sex and Motor Function. BIOLOGY 2022; 11:biology11020189. [PMID: 35205056 PMCID: PMC8869334 DOI: 10.3390/biology11020189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 01/20/2022] [Accepted: 01/22/2022] [Indexed: 11/16/2022]
Abstract
Simple Summary In the first two years following spinal cord injury, people lose up to 50% of bone below the injury. This injury-induced bone loss significantly affects rehabilitation and leaves people vulnerable to fractures and post-fracture complications, including lung and urinary tract infections, blood clots in the veins, and depression. Unfortunately, little is known about the factors driving this bone loss. In fact, even though we know that injury, age, and sex independently increase bone loss, there have been no studies looking at the cumulative effects of these variables. People with spinal injury are aging, and the age at which injuries occur is increasing. It is essential to know whether these factors together will further compromise bone. To examine this, we assessed bone loss in young and old, male and female mice after spinal injury. As expected, we found that aging alone decreased motor activity and bone volume. Spinal injury also reduced bone volume, but it did not worsen the effects of age. Instead, injury effects appeared related to reduced rearing activity. The data suggest that although partial weight-bearing does not reduce bone loss after spinal cord injury, therapies that put full weight on the legs may be clinically effective. Abstract After spinal cord injury (SCI), 80% of individuals are diagnosed with osteopenia or osteoporosis. The dramatic loss of bone after SCI increases the potential for fractures 100-fold, with post-fracture complications occurring in 54% of cases. With the age of new SCI injuries increasing, we hypothesized that a SCI-induced reduction in weight bearing could further exacerbate age-induced bone loss. To test this, young (2–3 months) and old (20–30 months) male and female mice were given a moderate spinal contusion injury (T9–T10), and recovery was assessed for 28 days (BMS, rearing counts, distance traveled). Tibial trabecular bone volume was measured after 28 days with ex vivo microCT. While BMS scores did not differ across groups, older subjects travelled less in the open field and there was a decrease in rearing with age and SCI. As expected, aging decreased trabecular bone volume and cortical thickness in both old male and female mice. SCI alone also reduced trabecular bone volume in young mice, but did not have an additional effect beyond the age-dependent decrease in trabecular and cortical bone volume seen in both sexes. Interestingly, both rearing and total activity correlated with decreased bone volume. These data underscore the importance of load and use on bone mass. While partial weight-bearing does not stabilize/reverse bone loss in humans, our data suggest that therapies that simulate complete loading may be effective after SCI.
Collapse
Affiliation(s)
- Michelle A. Hook
- Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M Health Science Center, Bryan, TX 77807, USA; (R.D.); (M.T.); (R.C.); (A.S.); (K.C.); (C.G.G.)
- Correspondence: ; Tel.: +1-979-436-0568
| | - Alyssa Falck
- Veterinary Integrative Biosciences, College of Veterinary Medicine and Biomedical Sciences, College Station, TX 77843, USA; (A.F.); (D.G.)
| | - Ravali Dundumulla
- Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M Health Science Center, Bryan, TX 77807, USA; (R.D.); (M.T.); (R.C.); (A.S.); (K.C.); (C.G.G.)
| | - Mabel Terminel
- Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M Health Science Center, Bryan, TX 77807, USA; (R.D.); (M.T.); (R.C.); (A.S.); (K.C.); (C.G.G.)
| | - Rachel Cunningham
- Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M Health Science Center, Bryan, TX 77807, USA; (R.D.); (M.T.); (R.C.); (A.S.); (K.C.); (C.G.G.)
| | - Arthur Sefiani
- Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M Health Science Center, Bryan, TX 77807, USA; (R.D.); (M.T.); (R.C.); (A.S.); (K.C.); (C.G.G.)
| | - Kayla Callaway
- Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M Health Science Center, Bryan, TX 77807, USA; (R.D.); (M.T.); (R.C.); (A.S.); (K.C.); (C.G.G.)
| | - Dana Gaddy
- Veterinary Integrative Biosciences, College of Veterinary Medicine and Biomedical Sciences, College Station, TX 77843, USA; (A.F.); (D.G.)
| | - Cédric G. Geoffroy
- Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M Health Science Center, Bryan, TX 77807, USA; (R.D.); (M.T.); (R.C.); (A.S.); (K.C.); (C.G.G.)
| |
Collapse
|
11
|
Bigford GE, Donovan A, Webster MT, Dietrich WD, Nash MS. Selective Myostatin Inhibition Spares Sublesional Muscle Mass and Myopenia-Related Dysfunction after Severe Spinal Cord Contusion in Mice. J Neurotrauma 2021; 38:3440-3455. [PMID: 34714134 DOI: 10.1089/neu.2021.0061] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Clinically relevant myopenia accompanies spinal cord injury (SCI), and compromises function, metabolism, body composition, and health. Myostatin, a transforming growth factor (TGF)β family member, is a key negative regulator of skeletal muscle mass. We investigated inhibition of myostatin signaling using systemic delivery of a highly selective monoclonal antibody - muSRK-015P (40 mg/kg) - that blocks release of active growth factor from the latent form of myostatin. Adult female mice (C57BL/6) were subjected to a severe SCI (65 kdyn) at T9 and were then immediately and 1 week later administered test articles: muSRK-015P (40 mg/kg) or control (vehicle or IgG). A sham control group (laminectomy only) was included. At euthanasia, (2 weeks post-SCI) muSRK-015P preserved whole body lean mass and sublesional gastrocnemius and soleus mass. muSRK-015P-treated mice with SCI also had significantly attenuated myofiber atrophy, lipid infiltration, and loss of slow-oxidative phenotype in soleus muscle. These outcomes were accompanied by significantly improved sublesional motor function and muscle force production at 1 and 2 weeks post-SCI. At 2 weeks post-SCI, lean mass was significantly decreased in SCI-IgG mice, but was not different in SCI-muSRK-015P mice than in sham controls. Total energy expenditure (kCal/day) at 2 weeks post-SCI was lower in SCI-immunoglobulin (Ig)G mice, but not different in SCI-muSRK-015P mice than in sham controls. We conclude that in a randomized, blinded, and controlled study in mice, myostatin inhibition using muSRK-015P had broad effects on physical, metabolic, and functional outcomes when compared with IgG control treated SCI animals. These findings may identify a useful, targeted therapeutic strategy for treating post-SCI myopenia and related sequelae in humans.
Collapse
Affiliation(s)
- Gregory E Bigford
- Department of Neurological Surgery and the Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, Florida, USA
| | | | | | - W Dalton Dietrich
- Department of Neurological Surgery and the Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Mark S Nash
- Department of Neurological Surgery and the Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, Florida, USA.,Department of Physical Medicine and Rehabilitation, University of Miami Miller School of Medicine, Miami, Florida, USA.,Department of Physical Therapy, University of Miami, Miami, Florida, USA
| |
Collapse
|
12
|
Pharmacologic approaches to prevent skeletal muscle atrophy after spinal cord injury. Curr Opin Pharmacol 2021; 60:193-199. [PMID: 34461564 PMCID: PMC9190029 DOI: 10.1016/j.coph.2021.07.023] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 07/26/2021] [Accepted: 07/28/2021] [Indexed: 11/20/2022]
Abstract
Skeletal muscle atrophy is a hallmark of severe spinal cord injury (SCI) that is precipitated by the neural insult and paralysis. Additionally, other factors may influence muscle loss, including systemic inflammation, low testosterone, low insulin-like growth factor (IGF)-1, and high-dose glucocorticoid treatment. The signaling cascades that drive SCI-induced muscle loss are common among most forms of disuse atrophy and include ubiquitin-proteasome signaling and others. However, differing magnitudes and patterns of atrophic signals exist after SCI versus other disuse conditions and are accompanied by endogenous inhibition of IGF-1/PI3K/Akt signaling, which combine to produce exceedingly rapid atrophy. Several well-established anabolic agents, including androgens and myostatin inhibitors, display diminished ability to prevent SCI-induced atrophy, while ursolic acid and β2-agonists more effectively attenuate muscle loss. Strategies combining physical rehabilitation regimens to reload the paralyzed limbs with drugs targeting the underlying molecular pathways hold the greatest potential to improve muscle recovery after severe SCI.
Collapse
|
13
|
Fujita H, Horie M, Shimizu K, Nagamori E. Microarray profiling of gene expression in C2C12 myotubes trained by electric pulse stimulation. J Biosci Bioeng 2021; 132:417-422. [PMID: 34348874 DOI: 10.1016/j.jbiosc.2021.06.016] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 06/16/2021] [Accepted: 06/30/2021] [Indexed: 10/20/2022]
Abstract
Electric pulse-stimulated C2C12 myotubes are gaining interest in the field of muscle physiology and biotechnology because electric pulse stimulation (EPS) enhances sarcomere structure development and active tension generation capability. Recently, we found that termination of EPS results in the rapid loss of active tension generation accompanied by disassembly of the sarcomere structure, which may represent an in vitro muscle atrophy model. To elucidate the molecular mechanism underlying this rapid loss of active tension generation and sarcomere structure disassembly after termination of EPS, we performed transcriptomic analysis using microarray. After termination of EPS, 74 genes were upregulated and 120 genes were downregulated after 30 min; however, atrophy-related genes were not found among these genes. To further assess the effect of EPS on gene expression, we re-applied EPS after its termination for 8 h and searched for genes whose expression was reversed. Four genes were upregulated by termination of EPS and downregulated by the re-application of EPS, whereas two genes were downregulated by termination of EPS and upregulated by the re-application of EPS. Although none of these genes were atrophy- or hypertrophy-related, the results presented in this study will contribute to the understanding of gene expression changes that mediate rapid loss of active tension generation and sarcomere structure disassembly following termination of EPS in C2C12 myotubes.
Collapse
Affiliation(s)
- Hideaki Fujita
- Department of Stem Cell Biology, Research Institute for Radiation Biology and Medicine, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima 734-8553, Japan
| | - Masanobu Horie
- Division of Biochemical Engineering, Radioisotope Research Center, Kyoto University, Yoshida-Konoe-Cho, Sakyo-ku, Kyoto 606-8507, Japan
| | - Kazunori Shimizu
- Department of Biomolecular Engineering, Graduate School of Engineering, Nagoya University, Nagoya 464-8603, Japan
| | - Eiji Nagamori
- Department of Biomedical Engineering, Osaka Institute of Technology, 5-16-1 Omiya, Asahi-ku, Osaka 535-8585, Japan.
| |
Collapse
|
14
|
Ehmsen JT, Kawaguchi R, Kaval D, Johnson AE, Nachun D, Coppola G, Höke A. GADD45A is a protective modifier of neurogenic skeletal muscle atrophy. JCI Insight 2021; 6:e149381. [PMID: 34128833 PMCID: PMC8410074 DOI: 10.1172/jci.insight.149381] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Accepted: 05/26/2021] [Indexed: 12/14/2022] Open
Abstract
Neurogenic muscle atrophy is the loss of skeletal muscle mass and function that occurs with nerve injury and in denervating diseases, such as amyotrophic lateral sclerosis. Aside from prompt restoration of innervation and exercise where feasible, there are currently no effective strategies for maintaining skeletal muscle mass in the setting of denervation. We conducted a longitudinal analysis of gene expression changes occurring in atrophying skeletal muscle and identified growth arrest and DNA damage-inducible A (Gadd45a) as a gene that shows one of the earliest and most sustained increases in expression in skeletal muscle after denervation. We evaluated the role of this induction using genetic mouse models and found that mice lacking GADD45A showed accelerated and exacerbated neurogenic muscle atrophy, as well as loss of fiber type identity. Our genetic analyses demonstrate that, rather than directly contributing to muscle atrophy as proposed in earlier studies, GADD45A induction likely represents a protective negative feedback response to denervation. Establishing the downstream effectors that mediate this protective effect and the pathways they participate in may yield new opportunities to modify the course of muscle atrophy.
Collapse
Affiliation(s)
- Jeffrey T Ehmsen
- Neuromuscular Division, Department of Neurology, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Riki Kawaguchi
- Department of Neurology and Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California, USA
| | - Damlanur Kaval
- Neuromuscular Division, Department of Neurology, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Anna E Johnson
- Neuromuscular Division, Department of Neurology, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Daniel Nachun
- Department of Neurology and Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California, USA
| | - Giovanni Coppola
- Department of Neurology and Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California, USA
| | - Ahmet Höke
- Neuromuscular Division, Department of Neurology, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
15
|
Zhao W, Peng Y, Hu Y, Guo XE, Li J, Cao J, Pan J, Feng JQ, Cardozo C, Jarvis J, Bauman WA, Qin W. Electrical stimulation of hindlimb skeletal muscle has beneficial effects on sublesional bone in a rat model of spinal cord injury. Bone 2021; 144:115825. [PMID: 33348128 PMCID: PMC7868091 DOI: 10.1016/j.bone.2020.115825] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 12/15/2020] [Accepted: 12/16/2020] [Indexed: 12/16/2022]
Abstract
Spinal cord injury (SCI) results in marked atrophy of sublesional skeletal muscle and substantial loss of bone. In this study, the effects of prolonged electrical stimulation (ES) and/or testosterone enanthate (TE) on muscle mass and bone formation in a rat model of SCI were tested. Compared to sham-transected animals, a significant reduction of the mass of soleus, plantaris and extensor digitorum longus (EDL) muscles was observed in animals 6 weeks post-SCI. Notably, ES or ES + TE resulted in the increased mass of the EDL muscles. ES or ES + TE significantly decreased mRNA levels of muscle atrophy markers (e.g., MAFbx and MurF1) in the EDL. Significant decreases in bone mineral density (BMD) (-27%) and trabecular bone volume (-49.3%) at the distal femur were observed in animals 6 weeks post injury. TE, ES and ES + TE treatment significantly increased BMD by +6.4%, +5.4%, +8.5% and bone volume by +22.2%, and +56.2% and+ 60.2%, respectively. Notably, ES alone or ES + TE resulted in almost complete restoration of cortical stiffness estimated by finite element analysis in SCI animals. Osteoblastogenesis was evaluated by colony-forming unit-fibroblastic (CFU-F) staining using bone marrow mesenchymal stem cells obtained from the femur. SCI decreased the CFU-F+ cells by -56.8% compared to sham animals. TE or ES + TE treatment after SCI increased osteoblastogenesis by +74.6% and +67.2%, respectively. An osteoclastogenesis assay revealed significantly increased TRAP+ multinucleated cells (+34.8%) in SCI animals compared to sham animals. TE, ES and TE + ES treatment following SCI markedly decreased TRAP+ cells by -51.3%, -40.3% and -46.9%, respectively. Each intervention greatly reduced the ratio of RANKL to OPG mRNA of sublesional long bone. Collectively, our findings demonstrate that after neurologically complete paralysis, dynamic muscle resistance exercise by ES reduced muscle atrophy, downregulated genes involved in muscle wasting, and restored mechanical loading to sublesional bone to a degree that allowed for the preservation of bone by inhibition of bone resorption and/or by facilitating bone formation.
Collapse
Affiliation(s)
- Wei Zhao
- National Center for the Medical Consequences of SCI, James J. Peters VA Medical Center, Bronx, NY, USA; Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Yuanzhen Peng
- National Center for the Medical Consequences of SCI, James J. Peters VA Medical Center, Bronx, NY, USA
| | - Yizhong Hu
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | - X Edward Guo
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | - Jiliang Li
- Indiana University Purdue University Indianapolis, Indianapolis, IN, USA
| | - Jay Cao
- United States Department of Agriculture Agricultural Research Service Human Nutrition Research Center, Grand Forks, ND, USA
| | - Jiangping Pan
- National Center for the Medical Consequences of SCI, James J. Peters VA Medical Center, Bronx, NY, USA
| | - Jian Q Feng
- Baylor College of Dentistry, TX A&M, Dallas, TX, USA
| | - Christopher Cardozo
- National Center for the Medical Consequences of SCI, James J. Peters VA Medical Center, Bronx, NY, USA; Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Rehabilitation Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jonathan Jarvis
- School of Sport and Exercise Sciences, Liverpool John Moores University, Liverpool L3 3AF, United Kingdom
| | - William A Bauman
- National Center for the Medical Consequences of SCI, James J. Peters VA Medical Center, Bronx, NY, USA; Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Rehabilitation Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Weiping Qin
- National Center for the Medical Consequences of SCI, James J. Peters VA Medical Center, Bronx, NY, USA; Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
16
|
The neuromuscular junction is a focal point of mTORC1 signaling in sarcopenia. Nat Commun 2020; 11:4510. [PMID: 32908143 PMCID: PMC7481251 DOI: 10.1038/s41467-020-18140-1] [Citation(s) in RCA: 87] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Accepted: 08/05/2020] [Indexed: 12/21/2022] Open
Abstract
With human median lifespan extending into the 80s in many developed countries, the societal burden of age-related muscle loss (sarcopenia) is increasing. mTORC1 promotes skeletal muscle hypertrophy, but also drives organismal aging. Here, we address the question of whether mTORC1 activation or suppression is beneficial for skeletal muscle aging. We demonstrate that chronic mTORC1 inhibition with rapamycin is overwhelmingly, but not entirely, positive for aging mouse skeletal muscle, while genetic, muscle fiber-specific activation of mTORC1 is sufficient to induce molecular signatures of sarcopenia. Through integration of comprehensive physiological and extensive gene expression profiling in young and old mice, and following genetic activation or pharmacological inhibition of mTORC1, we establish the phenotypically-backed, mTORC1-focused, multi-muscle gene expression atlas, SarcoAtlas (https://sarcoatlas.scicore.unibas.ch/), as a user-friendly gene discovery tool. We uncover inter-muscle divergence in the primary drivers of sarcopenia and identify the neuromuscular junction as a focal point of mTORC1-driven muscle aging.
Collapse
|
17
|
Chandrasekaran S, Davis J, Bersch I, Goldberg G, Gorgey AS. Electrical stimulation and denervated muscles after spinal cord injury. Neural Regen Res 2020; 15:1397-1407. [PMID: 31997798 PMCID: PMC7059583 DOI: 10.4103/1673-5374.274326] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Spinal cord injury (SCI) population with injury below T10 or injury to the cauda equina region is characterized by denervated muscles, extensive muscle atrophy, infiltration of intramuscular fat and formation of fibrous tissue. These morphological changes may put individuals with SCI at higher risk for developing other diseases such as various cardiovascular diseases, diabetes, obesity and osteoporosis. Currently, there is no available rehabilitation intervention to rescue the muscles or restore muscle size in SCI individuals with lower motor neuron denervation. We, hereby, performed a review of the available evidence that supports the use of electrical stimulation in restoration of denervated muscle following SCI. Long pulse width stimulation (LPWS) technique is an upcoming method of stimulating denervated muscles. Our primary objective is to explore the best stimulation paradigms (stimulation parameters, stimulation technique and stimulation wave) to achieve restoration of the denervated muscle. Stimulation parameters, such as the pulse duration, need to be 100–1000 times longer than in innervated muscles to achieve desirable excitability and contraction. The use of electrical stimulation in animal and human models induces muscle hypertrophy. Findings in animal models indicate that electrical stimulation, with a combination of exercise and pharmacological interventions, have proven to be effective in improving various aspects like relative muscle weight, muscle cross sectional area, number of myelinated regenerated fibers, and restoring some level of muscle function. Human studies have shown similar outcomes, identifying the use of LPWS as an effective strategy in increasing muscle cross sectional area, the size of muscle fibers, and improving muscle function. Therefore, displaying promise is an effective future stimulation intervention. In summary, LPWS is a novel stimulation technique for denervated muscles in humans with SCI. Successful studies on LPWS of denervated muscles will help in translating this stimulation technique to the clinical level as a rehabilitation intervention after SCI.
Collapse
Affiliation(s)
| | - John Davis
- Spinal Cord Injury and Disorders, Hunter Holmes McGuire VA Medical Center, Richmond, VA, USA
| | - Ines Bersch
- Swiss Paraplegic Centre, Nottwil, Switzerland; Institute of Clinical Sciences, Department of Orthopedics at the University of Gothenburg, Gothenburg, Sweden
| | - Gary Goldberg
- Department of Physical Medicine and Rehabilitation, Virginia Commonwealth University; Electrodiagnostic Center, Hunter Holmes McGuire VA Medical Center, Richmond, VA, USA
| | - Ashraf S Gorgey
- Spinal Cord Injury and Disorders, Hunter Holmes McGuire VA Medical Center; Department of Physical Medicine and Rehabilitation, Virginia Commonwealth University, Richmond, VA, USA
| |
Collapse
|
18
|
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
|
19
|
A transient protective effect of low-level laser irradiation against disuse-induced atrophy of rats. Lasers Med Sci 2019; 34:1829-1839. [PMID: 30949786 DOI: 10.1007/s10103-019-02778-5] [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: 11/30/2018] [Accepted: 03/18/2019] [Indexed: 12/25/2022]
Abstract
Satellite cells, a population of skeletal muscular stem cells, are generally recognized as the main and, possibly, the sole source of postnatal muscle regeneration. Previous studies have revealed the potential of low-level laser (LLL) irradiation in promoting satellite cell proliferation, which, thereby, boosts the recovery of skeletal muscle from atrophy. The purpose of this study is to investigate the beneficial effect of LLL on disuse-induced atrophy. The optimal irradiation condition of LLL (808 nm) enhancing the proliferation of Pax7+ve cells, isolated from tibialis anterior (TA) muscle, was examined and applied on TA muscle of disuse-induced atrophy model of the rats accordingly. Healthy rats were used as the control. On one hand, transiently, LLL was able to postpone the progression of atrophy for 1 week through a reduction of apoptosis in Pax7-veMyoD+ve (myocyte) population. Simultaneously, a significant enhancement was observed in Pax7+veMyoD+ve population; however, most of the increased cells underwent apoptosis since the second week, which suggested an impaired maturation of the population. On the other hand, in normal control rats with LLL irradiation, a significant increase in Pax7+veMyoD+ve cells and a significant decrease of apoptosis were observed. As a result, a strengthened muscle contraction was observed. Our data showed the capability of LLL in postponing the progression of disuse-induced atrophy for the first time. Furthermore, the result of normal rats with LLL irradiation showed the effectiveness of LLL to strengthen muscle contraction in healthy control.
Collapse
|
20
|
Rodrigues ACZ, Messi ML, Wang ZM, Abba MC, Pereyra A, Birbrair A, Zhang T, O’Meara M, Kwan P, Lopez EIS, Willis MS, Mintz A, Files DC, Furdui C, Oppenheim RW, Delbono O. The sympathetic nervous system regulates skeletal muscle motor innervation and acetylcholine receptor stability. Acta Physiol (Oxf) 2019; 225:e13195. [PMID: 30269419 PMCID: PMC7224611 DOI: 10.1111/apha.13195] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Revised: 09/23/2018] [Accepted: 09/26/2018] [Indexed: 12/19/2022]
Abstract
AIM Symptoms of autonomic failure are frequently the presentation of advanced age and neurodegenerative diseases that impair adaptation to common physiologic stressors. The aim of this work was to examine the interaction between the sympathetic and motor nervous system, the involvement of the sympathetic nervous system (SNS) in neuromuscular junction (NMJ) presynaptic motor function, the stability of postsynaptic molecular organization, and the skeletal muscle composition and function. METHODS Since muscle weakness is a symptom of diseases characterized by autonomic dysfunction, we studied the impact of regional sympathetic ablation on muscle motor innervation by using transcriptome analysis, retrograde tracing of the sympathetic outflow to the skeletal muscle, confocal and electron microscopy, NMJ transmission by electrophysiological methods, protein analysis, and state of the art microsurgical techniques, in C57BL6, MuRF1KO and Thy-1 mice. RESULTS We found that the SNS regulates motor nerve synaptic vesicle release, skeletal muscle transcriptome, muscle force generated by motor nerve activity, axonal neurofilament phosphorylation, myelin thickness, and myofibre subtype composition and CSA. The SNS also modulates the levels of postsynaptic membrane acetylcholine receptor by regulating the Gαi2 -Hdac4-Myogenin-MuRF1pathway, which is prevented by the overexpression of the guanine nucleotide-binding protein Gαi2 (Q205L), a constitutively active mutant G protein subunit. CONCLUSION The SNS regulates NMJ transmission, maintains optimal Gαi2 expression, and prevents any increase in Hdac4, myogenin, MuRF1, and miR-206. SNS ablation leads to upregulation of MuRF1, muscle atrophy, and downregulation of postsynaptic AChR. Our findings are relevant to clinical conditions characterized by progressive decline of sympathetic innervation, such as neurodegenerative diseases and aging.
Collapse
Affiliation(s)
- Anna C. Z. Rodrigues
- Department of Internal Medicine, Gerontology, Wake Forest School of Medicine, Winston-Salem, North Carolina
- Neuroscience Program, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Maria L. Messi
- Department of Internal Medicine, Gerontology, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Zhong-Min Wang
- Department of Internal Medicine, Gerontology, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Martin C. Abba
- Basic and Applied Immunological Research Center (CINIBA), School of Medicine, National University of La Plata, Buenos Aires, Argentina
| | - Andrea Pereyra
- Department of Internal Medicine, Gerontology, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Alexander Birbrair
- Department of Internal Medicine, Gerontology, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Tan Zhang
- Department of Internal Medicine, Gerontology, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Meaghan O’Meara
- Department of Internal Medicine, Gerontology, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Ping Kwan
- Department of Internal Medicine, Gerontology, Wake Forest School of Medicine, Winston-Salem, North Carolina
- Neuroscience Program, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Elsa I. S. Lopez
- Department of Internal Medicine, Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Monte S. Willis
- Department of Pathology, McAllister Heart Institute, University of North Carolina-Chapel Hill, Chapel Hill, North Carolina
| | - Akiva Mintz
- Department of Radiology, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - D. Clark Files
- Department of Internal Medicine, Gerontology, Wake Forest School of Medicine, Winston-Salem, North Carolina
- Department of Internal Medicine, Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina
- Department of Internal Medicine, Pulmonary, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Cristina Furdui
- Department of Internal Medicine, Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Ronald W. Oppenheim
- Department of Neurobiology and Anatomy, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Osvaldo Delbono
- Department of Internal Medicine, Gerontology, Wake Forest School of Medicine, Winston-Salem, North Carolina
- Neuroscience Program, Wake Forest School of Medicine, Winston-Salem, North Carolina
| |
Collapse
|
21
|
Gorgey AS, Witt O, O’Brien L, Cardozo C, Chen Q, Lesnefsky EJ, Graham ZA. Mitochondrial health and muscle plasticity after spinal cord injury. Eur J Appl Physiol 2018; 119:315-331. [DOI: 10.1007/s00421-018-4039-0] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2018] [Accepted: 11/22/2018] [Indexed: 01/15/2023]
|
22
|
Locomotor Training Promotes Time-dependent Functional Recovery after Experimental Spinal Cord Contusion. Neuroscience 2018; 392:258-269. [DOI: 10.1016/j.neuroscience.2018.08.033] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Revised: 08/28/2018] [Accepted: 08/29/2018] [Indexed: 12/13/2022]
|
23
|
Bigford GE, Darr AJ, Bracchi-Ricard VC, Gao H, Nash MS, Bethea JR. Effects of ursolic acid on sub-lesional muscle pathology in a contusion model of spinal cord injury. PLoS One 2018; 13:e0203042. [PMID: 30157245 PMCID: PMC6114926 DOI: 10.1371/journal.pone.0203042] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2018] [Accepted: 08/14/2018] [Indexed: 12/25/2022] Open
Abstract
Spinal Cord Injury (SCI) results in severe sub-lesional muscle atrophy and fiber type transformation from slow oxidative to fast glycolytic, both contributing to functional deficits and maladaptive metabolic profiles. Therapeutic countermeasures have had limited success and muscle-related pathology remains a clinical priority. mTOR signaling is known to play a critical role in skeletal muscle growth and metabolism, and signal integration of anabolic and catabolic pathways. Recent studies show that the natural compound ursolic acid (UA) enhances mTOR signaling intermediates, independently inhibiting atrophy and inducing hypertrophy. Here, we examine the effects of UA treatment on sub-lesional muscle mTOR signaling, catabolic genes, and functional deficits following severe SCI in mice. We observe that UA treatment significantly attenuates SCI induced decreases in activated forms of mTOR, and signaling intermediates PI3K, AKT, and S6K, and the upregulation of catabolic genes including FOXO1, MAFbx, MURF-1, and PSMD11. In addition, UA treatment improves SCI induced deficits in body and sub-lesional muscle mass, as well as functional outcomes related to muscle function, motor coordination, and strength. These findings provide evidence that UA treatment may be a potential therapeutic strategy to improve muscle-specific pathological consequences of SCI.
Collapse
Affiliation(s)
- Gregory E. Bigford
- The Miami Project to Cure Paralysis, Miami, Florida, United States of America
- Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, Florida, United States of America
| | - Andrew J. Darr
- Department of Health Sciences Education, University of Illinois College of Medicine at Peoria, Peoria, Illinois, United States of America
| | | | - Han Gao
- The Miami Project to Cure Paralysis, Miami, Florida, United States of America
- Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, Florida, United States of America
| | - Mark S. Nash
- The Miami Project to Cure Paralysis, Miami, Florida, United States of America
- Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, Florida, United States of America
- Department of Rehabilitation Medicine, University of Miami Miller School of Medicine, Miami, Florida, United States of America
| | - John R. Bethea
- Department of Biology, Drexel University, Philadelphia, Pennsylvania, United States of America
| |
Collapse
|
24
|
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
|
25
|
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: 2.0] [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
|
26
|
Otzel DM, Lee J, Ye F, Borst SE, Yarrow JF. Activity-Based Physical Rehabilitation with Adjuvant Testosterone to Promote Neuromuscular Recovery after Spinal Cord Injury. Int J Mol Sci 2018; 19:ijms19061701. [PMID: 29880749 PMCID: PMC6032131 DOI: 10.3390/ijms19061701] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2018] [Revised: 05/31/2018] [Accepted: 06/01/2018] [Indexed: 12/22/2022] Open
Abstract
Neuromuscular impairment and reduced musculoskeletal integrity are hallmarks of spinal cord injury (SCI) that hinder locomotor recovery. These impairments are precipitated by the neurological insult and resulting disuse, which has stimulated interest in activity-based physical rehabilitation therapies (ABTs) that promote neuromuscular plasticity after SCI. However, ABT efficacy declines as SCI severity increases. Additionally, many men with SCI exhibit low testosterone, which may exacerbate neuromusculoskeletal impairment. Incorporating testosterone adjuvant to ABTs may improve musculoskeletal recovery and neuroplasticity because androgens attenuate muscle loss and the slow-to-fast muscle fiber-type transition after SCI, in a manner independent from mechanical strain, and promote motoneuron survival. These neuromusculoskeletal benefits are promising, although testosterone alone produces only limited functional improvement in rodent SCI models. In this review, we discuss the (1) molecular deficits underlying muscle loss after SCI; (2) independent influences of testosterone and locomotor training on neuromuscular function and musculoskeletal integrity post-SCI; (3) hormonal and molecular mechanisms underlying the therapeutic efficacy of these strategies; and (4) evidence supporting a multimodal strategy involving ABT with adjuvant testosterone, as a potential means to promote more comprehensive neuromusculoskeletal recovery than either strategy alone.
Collapse
Affiliation(s)
- Dana M Otzel
- Brain Rehabilitation Research Center, Malcom Randall Veterans Affairs Medical Center, North Florida/South Georgia Veterans Health System, Gainesville, FL 32608, USA.
| | - Jimmy Lee
- Research Service, Malcom Randall Veterans Affairs Medical Center, North Florida/South Georgia Veterans Health System, Gainesville, FL 32608, USA.
| | - Fan Ye
- Research Service, Malcom Randall Veterans Affairs Medical Center, North Florida/South Georgia Veterans Health System, Gainesville, FL 32608, USA.
| | - Stephen E Borst
- Department of Applied Physiology, Kinesiology and University of Florida College of Health and Human Performance, Gainesville, FL 32603, USA.
| | - Joshua F Yarrow
- Research Service, Malcom Randall Veterans Affairs Medical Center, North Florida/South Georgia Veterans Health System, Gainesville, FL 32608, USA.
- Division of Endocrinology, Diabetes and Metabolism, University of Florida College of Medicine, Gainesville, FL 32610, USA.
| |
Collapse
|
27
|
The Role of IGF-1 Signaling in Skeletal Muscle Atrophy. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1088:109-137. [PMID: 30390250 DOI: 10.1007/978-981-13-1435-3_6] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Insulin-like growth factor 1 (IGF-1) is a key anabolic growth factor stimulating phosphatidylinositol 3-kinase (PI3K)/Akt signaling which is well known for regulating muscle hypertrophy. However, the role of IGF-1 in muscle atrophy is less clear. This review provides an overview of the mechanisms via which IGF-1 signaling is implicated in several conditions of muscle atrophy and via which mechanisms protein turnover is altered. IGF-1/PI3K/Akt signaling stimulates the rate of protein synthesis via p70S6Kinase and p90 ribosomal S6 kinase and negatively regulates protein degradation, predominantly by its inhibiting effect on proteasomal and lysosomal protein degradation. Caspase-dependent protein degradation is also attenuated by IGF/PI3K/Akt signaling, whereas evidence for an effect on calpain-dependent protein degradation is inconclusive. IGF-1/PI3K/Akt signaling reduces during denervation-, unloading-, and joint immobilization-induced muscle atrophy, whereas IGF-1/PI3K/Akt signaling seems unaltered during aging-associated muscle atrophy. During denervation and aging, IGF-1 overexpression or injection counteracts denervation- and aging-associated muscle atrophy, despite enhanced anabolic resistance with regard to IGF-1 signaling with aging. It remains unclear whether pharmacological stimulation of IGF-1/PI3K/Akt signaling attenuates immobilization- or unloading-induced muscle atrophy. Exploration of the possibilities to interfere with IGF-1/PI3K/Akt signaling reveals that microRNAs targeting IGF-1 signaling components are promising targets to counterbalance muscle atrophy. Overall, the findings summarized in this review show that in disuse conditions, but not with aging, IGF-1/PI3K/Akt signaling is attenuated and that in some conditions stimulation of this pathway may alleviate skeletal muscle atrophy.
Collapse
|
28
|
De Gasperi R, Graham ZA, Harlow LM, Bauman WA, Qin W, Cardozo CP. The Signature of MicroRNA Dysregulation in Muscle Paralyzed by Spinal Cord Injury Includes Downregulation of MicroRNAs that Target Myostatin Signaling. PLoS One 2016; 11:e0166189. [PMID: 27907012 PMCID: PMC5132212 DOI: 10.1371/journal.pone.0166189] [Citation(s) in RCA: 16] [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: 08/05/2016] [Accepted: 10/24/2016] [Indexed: 12/25/2022] Open
Abstract
Spinal cord injury (SCI) results in muscle atrophy, reduced force generation and an oxidative-to-glycolytic fiber type shift. The mechanisms responsible for these alterations remain incompletely understood. To gain new insights regarding mechanisms involved in deterioration of muscle after SCI, global expression profiles of miRs in paralyzed gastrocnemius muscle were compared between sham-operated (Sham) and spinal cord-transected (SCI) rats. Ingenuity Pathways Analysis of the altered miRs identified signaling via insulin, IGF-1, integrins and TGF-β as being significantly enriched for target genes. By qPCR, miRs 23a, 23b, 27b, 145, and 206, were downregulated in skeletal muscle 56 days after SCI. Using FISH, miR-145, a miR not previously implicated in the function of skeletal muscle, was found to be localized to skeletal muscle fibers. One predicted target of miR-145 was Cited2, a transcriptional regulator that modulates signaling through NF-κB, Smad3 and other transcription factors. The 3’ UTR of Cited2 mRNA contained a highly conserved miR-145 seed sequence. Luciferase reporter assays confirmed that miR-145 interacts with this seed sequence. However, Cited2 protein levels were similar between Sham and SCI groups, indicating a biochemical interaction that was not involved in the context of adaptations after SCI. Taken together, the findings indicate dysregulation of several highly expressed miRs in skeletal muscle after SCI and suggest that reduced expression of miR-23a, 145 and 206 may have roles in alteration in skeletal muscle mass and insulin responsiveness in muscle paralyzed by upper motor neuron injuries.
Collapse
Affiliation(s)
- Rita De Gasperi
- VA RR&D Service National Center for the Medical Consequences of Spinal Cord Injury, James J. Peters Medical Center, Bronx, New York
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, New York
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Zachary A. Graham
- VA RR&D Service National Center for the Medical Consequences of Spinal Cord Injury, James J. Peters Medical Center, Bronx, New York
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Lauren M. Harlow
- VA RR&D Service National Center for the Medical Consequences of Spinal Cord Injury, James J. Peters Medical Center, Bronx, New York
| | - William A. Bauman
- VA RR&D Service National Center for the Medical Consequences of Spinal Cord Injury, James J. Peters Medical Center, Bronx, New York
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
- Department of Rehabilitation Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Weiping Qin
- VA RR&D Service National Center for the Medical Consequences of Spinal Cord Injury, James J. Peters Medical Center, Bronx, New York
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Christopher P. Cardozo
- VA RR&D Service National Center for the Medical Consequences of Spinal Cord Injury, James J. Peters Medical Center, Bronx, New York
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
- Department of Rehabilitation Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
- Department of Pharmacologic Science Icahn School of Medicine at Mount Sinai, New York, New York
- * E-mail:
| |
Collapse
|
29
|
Güth R, Chaidez A, Samanta MP, Unguez GA. Properties of skeletal muscle in the teleost Sternopygus macrurus are unaffected by short-term electrical inactivity. Physiol Genomics 2016; 48:699-710. [PMID: 27449658 DOI: 10.1152/physiolgenomics.00068.2016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Accepted: 07/18/2016] [Indexed: 11/22/2022] Open
Abstract
Skeletal muscle is distinguished from other tissues on the basis of its shape, biochemistry, and physiological function. Based on mammalian studies, fiber size, fiber types, and gene expression profiles are regulated, in part, by the electrical activity exerted by the nervous system. To address whether similar adaptations to changes in electrical activity in skeletal muscle occur in teleosts, we studied these phenotypic properties of ventral muscle in the electric fish Sternopygus macrurus following 2 and 5 days of electrical inactivation by spinal transection. Our data show that morphological and biochemical properties of skeletal muscle remained largely unchanged after these treatments. Specifically, the distribution of type I and type II muscle fibers and the cross-sectional areas of these fiber types observed in control fish remained unaltered after each spinal transection survival period. This response to electrical inactivation was generally reflected at the transcript level in real-time PCR and RNA-seq data by showing little effect on the transcript levels of genes associated with muscle fiber type differentiation and plasticity, the sarcomere complex, and pathways implicated in the regulation of muscle fiber size. Data from this first study characterizing the acute influence of neural activity on muscle mass and sarcomere gene expression in a teleost are discussed in the context of comparative studies in mammalian model systems and vertebrate species from different lineages.
Collapse
Affiliation(s)
- Robert Güth
- Department of Biology, New Mexico State University, Las Cruces, New Mexico; and
| | - Alexander Chaidez
- Department of Biology, New Mexico State University, Las Cruces, New Mexico; and
| | | | - Graciela A Unguez
- Department of Biology, New Mexico State University, Las Cruces, New Mexico; and
| |
Collapse
|
30
|
Bullard SA, Seo S, Schilling B, Dyle MC, Dierdorff JM, Ebert SM, DeLau AD, Gibson BW, Adams CM. Gadd45a Protein Promotes Skeletal Muscle Atrophy by Forming a Complex with the Protein Kinase MEKK4. J Biol Chem 2016; 291:17496-17509. [PMID: 27358404 PMCID: PMC5016147 DOI: 10.1074/jbc.m116.740308] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2016] [Indexed: 12/13/2022] Open
Abstract
Skeletal muscle atrophy is a serious and highly prevalent condition that remains poorly understood at the molecular level. Previous work found that skeletal muscle atrophy involves an increase in skeletal muscle Gadd45a expression, which is necessary and sufficient for skeletal muscle fiber atrophy. However, the direct mechanism by which Gadd45a promotes skeletal muscle atrophy was unknown. To address this question, we biochemically isolated skeletal muscle proteins that associate with Gadd45a as it induces atrophy in mouse skeletal muscle fibers in vivo. We found that Gadd45a interacts with multiple proteins in skeletal muscle fibers, including, most prominently, MEKK4, a mitogen-activated protein kinase kinase kinase that was not previously known to play a role in skeletal muscle atrophy. Furthermore, we found that, by forming a complex with MEKK4 in skeletal muscle fibers, Gadd45a increases MEKK4 protein kinase activity, which is both sufficient to induce skeletal muscle fiber atrophy and required for Gadd45a-mediated skeletal muscle fiber atrophy. Together, these results identify a direct biochemical mechanism by which Gadd45a induces skeletal muscle atrophy and provide new insight into the way that skeletal muscle atrophy occurs at the molecular level.
Collapse
Affiliation(s)
- Steven A Bullard
- From the Department of Internal Medicine.,Fraternal Order of Eagles Diabetes Research Center, and.,Departments of Molecular Physiology and Biophysics and.,the Iowa City Veterans Affairs Medical Center, Iowa City, Iowa 52246
| | - Seongjin Seo
- Ophthalmology and Visual Sciences, University of Iowa, Iowa City, Iowa 52242
| | - Birgit Schilling
- the Buck Institute for Research on Aging, Novato, California 94945, and
| | - Michael C Dyle
- From the Department of Internal Medicine.,Fraternal Order of Eagles Diabetes Research Center, and.,Departments of Molecular Physiology and Biophysics and.,the Iowa City Veterans Affairs Medical Center, Iowa City, Iowa 52246.,Emmyon, Inc., Coralville, Iowa 52241
| | - Jason M Dierdorff
- From the Department of Internal Medicine.,Fraternal Order of Eagles Diabetes Research Center, and.,Departments of Molecular Physiology and Biophysics and.,the Iowa City Veterans Affairs Medical Center, Iowa City, Iowa 52246
| | - Scott M Ebert
- From the Department of Internal Medicine.,Fraternal Order of Eagles Diabetes Research Center, and.,Departments of Molecular Physiology and Biophysics and.,the Iowa City Veterans Affairs Medical Center, Iowa City, Iowa 52246.,Emmyon, Inc., Coralville, Iowa 52241
| | - Austin D DeLau
- From the Department of Internal Medicine.,Fraternal Order of Eagles Diabetes Research Center, and.,Departments of Molecular Physiology and Biophysics and.,the Iowa City Veterans Affairs Medical Center, Iowa City, Iowa 52246
| | - Bradford W Gibson
- the Buck Institute for Research on Aging, Novato, California 94945, and.,the Department of Pharmaceutical Chemistry, University of California at San Francisco, San Francisco, California 94143
| | - Christopher M Adams
- From the Department of Internal Medicine, .,Fraternal Order of Eagles Diabetes Research Center, and.,Departments of Molecular Physiology and Biophysics and.,the Iowa City Veterans Affairs Medical Center, Iowa City, Iowa 52246.,Emmyon, Inc., Coralville, Iowa 52241
| |
Collapse
|
31
|
Silva KOGD, Pereira SDC, Portovedo M, Milanski M, Galindo LCM, Guzmán‐Quevedo O, Manhães‐de‐Castro R, Toscano AE. Effects of maternal low‐protein diet on parameters of locomotor activity in a rat model of cerebral palsy. Int J Dev Neurosci 2016; 52:38-45. [DOI: 10.1016/j.ijdevneu.2016.05.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Revised: 05/10/2016] [Accepted: 05/11/2016] [Indexed: 01/07/2023] Open
Affiliation(s)
| | | | - Mariana Portovedo
- Faculty of Applied SciencesUniversity of Campinas13084‐970CampinasBrazil
| | - Marciane Milanski
- Faculty of Applied SciencesUniversity of Campinas13084‐970CampinasBrazil
| | | | | | | | - Ana Elisa Toscano
- Department of Nursing, CAVFederal University of Pernambuco55608‐680Vitória de Santo AntãoBrazil
| |
Collapse
|
32
|
Su Z, Hu L, Cheng J, Klein JD, Hassounah F, Cai H, Li M, Wang H, Wang XH. Acupuncture plus low-frequency electrical stimulation (Acu-LFES) attenuates denervation-induced muscle atrophy. J Appl Physiol (1985) 2016; 120:426-36. [PMID: 26679610 PMCID: PMC4754622 DOI: 10.1152/japplphysiol.00175.2015] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2015] [Accepted: 12/16/2015] [Indexed: 12/20/2022] Open
Abstract
Muscle wasting occurs in a variety of clinical situations, including denervation. There is no effective pharmacological treatment for muscle wasting. In this study, we used a tibial nerve denervation model to test acupuncture plus low-frequency electric stimulation (Acu-LFES) as a therapeutic strategy for muscle atrophy. Acupuncture needles were connected to an SDZ-II electronic acupuncture device delivering pulses at 20 Hz and 1 mA; the treatment was 15 min daily for 2 wk. Acu-LFES prevented soleus and plantaris muscle weight loss and increased muscle cross-sectional area in denervated mice. The abundances of Pax7, MyoD, myogenin, and embryonic myosin heavy chain were significantly increased by Acu-LFES in both normal and denervated muscle. The number of central nuclei was increased in Acu-LFES-treated muscle fibers. Phosphorylation of Akt was downregulated by denervation leading to a decline in muscle mass; however, Acu-LFES prevented the denervation-induced decline largely by upregulation of the IGF-1 signaling pathway. Acu-LFES reduced the abundance of muscle catabolic proteins forkhead O transcription factor and myostatin, contributing to the attenuated muscle atrophy. Acu-LFES stimulated the expression of macrophage markers (F4/80, IL-1b, and arginase-1) and inflammatory cytokines (IL-6, IFNγ, and TNFα) in normal and denervated muscle. Acu-LFES also stimulated production of the muscle-specific microRNAs miR-1 and miR-206. We conclude that Acu-LFES is effective in counteracting denervation-induced skeletal muscle atrophy and increasing muscle regeneration. Upregulation of IGF-1, downregulation of myostatin, and alteration of microRNAs contribute to the attenuation of muscle atrophy in denervated mice.
Collapse
Affiliation(s)
- Zhen Su
- Department of Nephrology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China; Renal Division, Department of Medicine, Emory University, Atlanta, Georgia
| | - Li Hu
- Acumox and Tuina Research Section, College of Acumox and Tuina, Shanghai University of Traditional Chinese Medicine, Shanghai, China; Renal Division, Department of Medicine, Emory University, Atlanta, Georgia
| | - Jinzhong Cheng
- Nephrology, Department of Medicine, Baylor College of Medicine, Houston, Texas; and
| | - Janet D Klein
- Renal Division, Department of Medicine, Emory University, Atlanta, Georgia
| | - Faten Hassounah
- Renal Division, Department of Medicine, Emory University, Atlanta, Georgia
| | - Hui Cai
- Renal Division, Department of Medicine, Emory University, Atlanta, Georgia
| | - Min Li
- Renal Division, Department of Medicine, Emory University, Atlanta, Georgia
| | - Haidong Wang
- Renal Division, Department of Medicine, Emory University, Atlanta, Georgia
| | - Xiaonan H Wang
- Renal Division, Department of Medicine, Emory University, Atlanta, Georgia
| |
Collapse
|
33
|
Graham ZA, Collier L, Peng Y, Saéz JC, Bauman WA, Qin W, Cardozo CP. A Soluble Activin Receptor IIB Fails to Prevent Muscle Atrophy in a Mouse Model of Spinal Cord Injury. J Neurotrauma 2016; 33:1128-35. [PMID: 26529111 DOI: 10.1089/neu.2015.4058] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Myostatin (MST) is a potent regulator of muscle growth and size. Spinal cord injury (SCI) results in marked atrophy of muscle below the level of injury. Currently, there is no effective pharmaceutical treatment available to prevent sublesional muscle atrophy post-SCI. To determine whether inhibition of MST with a soluble activin IIB receptor (RAP-031) prevents sublesional SCI-induced muscle atrophy, mice were randomly assigned to the following groups: Sham-SCI; SCI+Vehicle group (SCI-VEH); and SCI+RAP-031 (SCI-RAP-031). SCI was induced by complete transection at thoracic level 10. Animals were euthanized at 56 days post-surgery. RAP-031 reduced, but did not prevent, body weight loss post-SCI. RAP-031 increased total lean tissue mass compared to SCI-VEH (14.8%). RAP-031 increased forelimb muscle mass post-SCI by 38% and 19% for biceps and triceps, respectively (p < 0.001). There were no differences in hindlimb muscle weights between the RAP-031 and SCI-VEH groups. In the gastrocnemius, messenger RNA (mRNA) expression was elevated for interleukin (IL)-6 (8-fold), IL-1β (3-fold), and tumor necrosis factor alpha (8-fold) in the SCI-VEH, compared to the Sham group. Muscle RING finger protein 1 mRNA was 2-fold greater in the RAP-031 group, compared to Sham-SCI. RAP-031 did not influence cytokine expression. Bone mineral density of the distal femur and proximal tibia were decreased post-SCI (-26% and -28%, respectively) and were not altered by RAP-031. In conclusion, MST inhibition increased supralesional muscle mass, but did not prevent sublesional muscle or bone loss, or the inflammation in paralyzed muscle.
Collapse
Affiliation(s)
- Zachary A Graham
- 1 National Center of Excellence for the Medical Consequences of Spinal Cord Injury, James J. Peters VA Medical Center , Bronx, New York.,5 Department of Medicine, Icahn School of Medicine at Mount Sinai , New York, New York
| | - Lauren Collier
- 1 National Center of Excellence for the Medical Consequences of Spinal Cord Injury, James J. Peters VA Medical Center , Bronx, New York
| | - Yuanzhen Peng
- 1 National Center of Excellence for the Medical Consequences of Spinal Cord Injury, James J. Peters VA Medical Center , Bronx, New York
| | - Juan C Saéz
- 3 Department of Physiology, Pontificia Universidad Católica , Santiago, Chile .,4 Centro Interdisciplinario de Neurociencias de Valparaiso , Valparaiso, Chile
| | - William A Bauman
- 1 National Center of Excellence for the Medical Consequences of Spinal Cord Injury, James J. Peters VA Medical Center , Bronx, New York.,2 Medical Service, James J. Peters VA Medical Center , Bronx, New York.,5 Department of Medicine, Icahn School of Medicine at Mount Sinai , New York, New York.,6 Department of Rehabilitation Medicine, Icahn School of Medicine at Mount Sinai , New York, New York
| | - Weiping Qin
- 1 National Center of Excellence for the Medical Consequences of Spinal Cord Injury, James J. Peters VA Medical Center , Bronx, New York.,5 Department of Medicine, Icahn School of Medicine at Mount Sinai , New York, New York
| | - Christopher P Cardozo
- 1 National Center of Excellence for the Medical Consequences of Spinal Cord Injury, James J. Peters VA Medical Center , Bronx, New York.,2 Medical Service, James J. Peters VA Medical Center , Bronx, New York.,5 Department of Medicine, Icahn School of Medicine at Mount Sinai , New York, New York.,6 Department of Rehabilitation Medicine, Icahn School of Medicine at Mount Sinai , New York, New York.,7 Pharmacology and Systems Therapeutics, Icahn School of Medicine at Mount Sinai , New York, New York
| |
Collapse
|
34
|
Nakao R, Yamamoto S, Yasumoto Y, Kadota K, Oishi K. Impact of denervation-induced muscle atrophy on housekeeping gene expression in mice. Muscle Nerve 2014; 51:276-81. [DOI: 10.1002/mus.24310] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/03/2014] [Indexed: 01/30/2023]
Affiliation(s)
- Reiko Nakao
- Biological Clock Research Group, Biomedical Research Institute; National Institute of Advanced Industrial Science and Technology; Central 6, 1-1-1 Higashi Tsukuba Ibaraki 305-8566 Japan
| | - Saori Yamamoto
- Biological Clock Research Group, Biomedical Research Institute; National Institute of Advanced Industrial Science and Technology; Central 6, 1-1-1 Higashi Tsukuba Ibaraki 305-8566 Japan
| | - Yuki Yasumoto
- Biological Clock Research Group, Biomedical Research Institute; National Institute of Advanced Industrial Science and Technology; Central 6, 1-1-1 Higashi Tsukuba Ibaraki 305-8566 Japan
- Department of Applied Biological Science, Graduate School of Science and Technology; Tokyo University of Science; Chiba Japan
| | - Koji Kadota
- Agricultural Bioinformatics Research Unit, Graduate School of Agricultural and Life Sciences; University of Tokyo; Tokyo Japan
| | - Katsutaka Oishi
- Biological Clock Research Group, Biomedical Research Institute; National Institute of Advanced Industrial Science and Technology; Central 6, 1-1-1 Higashi Tsukuba Ibaraki 305-8566 Japan
- Department of Applied Biological Science, Graduate School of Science and Technology; Tokyo University of Science; Chiba Japan
- Department of Medical Genome Sciences, Graduate School of Frontier Sciences; University of Tokyo; Kashiwa Chiba Japan
| |
Collapse
|
35
|
Cisterna BA, Cardozo C, Sáez JC. Neuronal involvement in muscular atrophy. Front Cell Neurosci 2014; 8:405. [PMID: 25540609 PMCID: PMC4261799 DOI: 10.3389/fncel.2014.00405] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2014] [Accepted: 11/10/2014] [Indexed: 12/18/2022] Open
Abstract
The innervation of skeletal myofibers exerts a crucial influence on the maintenance of muscle tone and normal operation. Consequently, denervated myofibers manifest atrophy, which is preceded by an increase in sarcolemma permeability. Recently, de novo expression of hemichannels (HCs) formed by connexins (Cxs) and other none selective channels, including P2X7 receptors (P2X7Rs), and transient receptor potential, sub-family V, member 2 (TRPV2) channels was demonstrated in denervated fast skeletal muscles. The denervation-induced atrophy was drastically reduced in denervated muscles deficient in Cxs 43 and 45. Nonetheless, the transduction mechanism by which the nerve represses the expression of the above mentioned non-selective channels remains unknown. The paracrine action of extracellular signaling molecules including ATP, neurotrophic factors (i.e., brain-derived neurotrophic factor (BDNF)), agrin/LDL receptor-related protein 4 (Lrp4)/muscle-specific receptor kinase (MuSK) and acetylcholine (Ach) are among the possible signals for repression for connexin expression. This review discusses the possible role of relevant factors in maintaining the normal functioning of fast skeletal muscles and suppression of connexin hemichannel expression.
Collapse
Affiliation(s)
- Bruno A. Cisterna
- Departamento de Fisiología, Pontificia Universidad Católica de ChileSantiago, Chile
| | - Christopher Cardozo
- Center of Excellence for the Medical Consequences of Spinal Cord Injury, James J. Peters Veterans Affairs Medical CenterBronx, NY, USA
- Departments of Medicine and Rehabilitation Medicine, Icahn School of Medicine at Mount SinaiNew York, NY, USA
| | - Juan C. Sáez
- Departamento de Fisiología, Pontificia Universidad Católica de ChileSantiago, Chile
- Instituto Milenio, Centro Interdisciplinario de Neurociencias de Valparaíso, Universidad de ValparaísoValparaíso, Chile
| |
Collapse
|
36
|
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
|
37
|
Piccirillo R, Demontis F, Perrimon N, Goldberg AL. Mechanisms of muscle growth and atrophy in mammals and Drosophila. Dev Dyn 2014; 243:201-15. [PMID: 24038488 PMCID: PMC3980484 DOI: 10.1002/dvdy.24036] [Citation(s) in RCA: 98] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2013] [Revised: 08/01/2013] [Accepted: 08/01/2013] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND The loss of skeletal muscle mass (atrophy) that accompanies disuse and systemic diseases is highly debilitating. Although the pathogenesis of this condition has been primarily studied in mammals, Drosophila is emerging as an attractive system to investigate some of the mechanisms involved in muscle growth and atrophy. RESULTS In this review, we highlight the outstanding unsolved questions that may benefit from a combination of studies in both flies and mammals. In particular, we discuss how different environmental stimuli and signaling pathways influence muscle mass and strength and how a variety of disease states can cause muscle wasting. CONCLUSIONS Studies in Drosophila and mammals should help identify molecular targets for the treatment of muscle wasting in humans.
Collapse
Affiliation(s)
- Rosanna Piccirillo
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115
- Department of Oncology, IRCCS - Mario Negri Institute for Pharmacological Research, Milano, Italy
| | - Fabio Demontis
- Department of Genetics, Harvard Medical School, Boston, MA 02115
- Department of Developmental Neurobiology, Division of Developmental Biology, St. Jude Children’s Research Hospital, Memphis, TN 38105
| | - Norbert Perrimon
- Department of Genetics, Harvard Medical School, Boston, MA 02115
- Howard Hughes Medical Institute, Harvard Medical School, Boston, MA 02115
| | | |
Collapse
|
38
|
Fontes-Oliveira CC, Busquets S, Fuster G, Ametller E, Figueras M, Olivan M, Toledo M, López-Soriano FJ, Qu X, Demuth J, Stevens P, Varbanov A, Wang F, Isfort RJ, Argilés JM. A differential pattern of gene expression in skeletal muscle of tumor-bearing rats reveals dysregulation of excitation-contraction coupling together with additional muscle alterations. Muscle Nerve 2013; 49:233-48. [DOI: 10.1002/mus.23893] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2012] [Revised: 04/19/2013] [Accepted: 04/24/2013] [Indexed: 12/31/2022]
Affiliation(s)
- Cibely Cristine Fontes-Oliveira
- Cancer Research Group, Departament de Bioquímica i Biologia Molecular, Facultat de Biologia; Universitat de Barcelona; Diagonal 643 Barcelona 08028 Spain
| | - Sílvia Busquets
- Cancer Research Group, Departament de Bioquímica i Biologia Molecular, Facultat de Biologia; Universitat de Barcelona; Diagonal 643 Barcelona 08028 Spain
- Institut de Biomedicina de la Universitat de Barcelona; Barcelona Spain
| | - Gemma Fuster
- Cancer Research Group, Departament de Bioquímica i Biologia Molecular, Facultat de Biologia; Universitat de Barcelona; Diagonal 643 Barcelona 08028 Spain
| | - Elisabet Ametller
- Cancer Research Group, Departament de Bioquímica i Biologia Molecular, Facultat de Biologia; Universitat de Barcelona; Diagonal 643 Barcelona 08028 Spain
| | - Maite Figueras
- Cancer Research Group, Departament de Bioquímica i Biologia Molecular, Facultat de Biologia; Universitat de Barcelona; Diagonal 643 Barcelona 08028 Spain
| | - Mireia Olivan
- Cancer Research Group, Departament de Bioquímica i Biologia Molecular, Facultat de Biologia; Universitat de Barcelona; Diagonal 643 Barcelona 08028 Spain
| | - Míriam Toledo
- Cancer Research Group, Departament de Bioquímica i Biologia Molecular, Facultat de Biologia; Universitat de Barcelona; Diagonal 643 Barcelona 08028 Spain
| | - Francisco J. López-Soriano
- Cancer Research Group, Departament de Bioquímica i Biologia Molecular, Facultat de Biologia; Universitat de Barcelona; Diagonal 643 Barcelona 08028 Spain
- Institut de Biomedicina de la Universitat de Barcelona; Barcelona Spain
| | - Xiaoyan Qu
- Procter & Gamble; Mason Business Center; 8700 Mason-Montgomery Road Mason Ohio 45040 USA
| | - Jeffrey Demuth
- Procter & Gamble; Mason Business Center; 8700 Mason-Montgomery Road Mason Ohio 45040 USA
| | - Paula Stevens
- Procter & Gamble; Mason Business Center; 8700 Mason-Montgomery Road Mason Ohio 45040 USA
| | - Alex Varbanov
- Procter & Gamble; Mason Business Center; 8700 Mason-Montgomery Road Mason Ohio 45040 USA
| | - Feng Wang
- Procter & Gamble; Mason Business Center; 8700 Mason-Montgomery Road Mason Ohio 45040 USA
| | - Robert J. Isfort
- Procter & Gamble; Mason Business Center; 8700 Mason-Montgomery Road Mason Ohio 45040 USA
| | - Josep M. Argilés
- Cancer Research Group, Departament de Bioquímica i Biologia Molecular, Facultat de Biologia; Universitat de Barcelona; Diagonal 643 Barcelona 08028 Spain
- Institut de Biomedicina de la Universitat de Barcelona; Barcelona Spain
| |
Collapse
|
39
|
Wu Y, Collier L, Qin W, Creasey G, Bauman WA, Jarvis J, Cardozo C. Electrical stimulation modulates Wnt signaling and regulates genes for the motor endplate and calcium binding in muscle of rats with spinal cord transection. BMC Neurosci 2013; 14:81. [PMID: 23914941 PMCID: PMC3735397 DOI: 10.1186/1471-2202-14-81] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2013] [Accepted: 07/19/2013] [Indexed: 01/25/2023] Open
Abstract
BACKGROUND Spinal cord injury (SCI) results in muscle atrophy and a shift of slow oxidative to fast glycolytic fibers. Electrical stimulation (ES) at least partially restores muscle mass and fiber type distribution. The objective of this study was to was to characterize the early molecular adaptations that occur in rat soleus muscle after initiating isometric resistance exercise by ES for one hour per day for 1, 3 or 7 days when ES was begun 16 weeks after SCI. Additionally, changes in mRNA levels after ES were compared with those induced in soleus at the same time points after gastrocnemius tenotomy (GA). RESULTS ES increased expression of Hey1 and Pitx2 suggesting increased Notch and Wnt signaling, respectively, but did not normalize RCAN1.4, a measure of calcineurin/NFAT signaling, or PGC-1ß mRNA levels. ES increased PGC-1α expression but not that of slow myofibrillar genes. Microarray analysis showed that after ES, genes coding for calcium binding proteins and nicotinic acetylcholine receptors were increased, and the expression of genes involved in blood vessel formation and morphogenesis was altered. Of the 165 genes altered by ES only 16 were also differentially expressed after GA, of which 12 were altered in the same direction by ES and GA. In contrast to ES, GA induced expression of genes related to oxidative phosphorylation. CONCLUSIONS Notch and Wnt signaling may be involved in ES-induced increases in the mass of paralyzed muscle. Molecular adaptations of paralyzed soleus to resistance exercise are delayed or defective compared to normally innervated muscle.
Collapse
Affiliation(s)
- Yong Wu
- Center of Excellence for the Medical Consequences of SCI, James J. Peters VA Medical Center, 130 West Kingsbridge Road, Bronx, NY 10468, USA
| | - Lauren Collier
- Center of Excellence for the Medical Consequences of SCI, James J. Peters VA Medical Center, 130 West Kingsbridge Road, Bronx, NY 10468, USA
| | - Weiping Qin
- Center of Excellence for the Medical Consequences of SCI, James J. Peters VA Medical Center, 130 West Kingsbridge Road, Bronx, NY 10468, USA
- Department of Medicine, Mount Sinai School of Medicine, New York, NY, USA
| | - Graham Creasey
- VA Palo Alto Health Care System, Stanford University, Palo Alto, CA, USA
| | - William A Bauman
- Center of Excellence for the Medical Consequences of SCI, James J. Peters VA Medical Center, 130 West Kingsbridge Road, Bronx, NY 10468, USA
- Department of Medicine, Mount Sinai School of Medicine, New York, NY, USA
| | - Jonathan Jarvis
- School of Biomedical Sciences, University of Liverpool, Liverpool, UK
| | - Christopher Cardozo
- Center of Excellence for the Medical Consequences of SCI, James J. Peters VA Medical Center, 130 West Kingsbridge Road, Bronx, NY 10468, USA
- Department of Medicine, Mount Sinai School of Medicine, New York, NY, USA
- Department of Rehabilitation Medicine, Mount Sinai School of Medicine, New York, NY, USA
| |
Collapse
|
40
|
Sultan FA, Sweatt JD. The Role of the Gadd45 Family in the Nervous System: A Focus on Neurodevelopment, Neuronal Injury, and Cognitive Neuroepigenetics. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2013; 793:81-119. [DOI: 10.1007/978-1-4614-8289-5_6] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
41
|
von Walden F, Jakobsson F, Edström L, Nader GA. Altered autophagy gene expression and persistent atrophy suggest impaired remodeling in chronic hemiplegic human skeletal muscle. Muscle Nerve 2012; 46:785-92. [PMID: 22996233 DOI: 10.1002/mus.23387] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/13/2012] [Indexed: 11/10/2022]
Abstract
INTRODUCTION Upper motor neuron lesions after stroke are a major cause of disability. We aimed to determine whether skeletal muscles from these patients display typical molecular signatures of inflammation, growth arrest, and atrophy. METHODS Muscle biopsies were analyzed for morphological, histochemical, ultrastructural, and molecular features indicative of changes in gene expression involved in muscle atrophy. RESULTS Chronic hemiplegia resulted in ~9.5% atrophy, fiber type shifts, and histochemical and ultrastructural signs of impaired remodeling. TNF and TWEAK expressions were unaltered, but MSTN mRNA was lower (-73%, P < 0.05) in paretic tibialis anterior vs. age-matched controls. The expression of autophagy-related genes (BCN-1, LC3, and GABARAPL1) was lower in paretic tibialis anterior (-81%, -48%, and -60%, respectively, P < 0.01) and soleus (-85%, -54%, and -60% respectively, P < 0.01) compared with old controls. CONCLUSIONS Persistent atrophy in chronic spastic hemiplegia may be associated with impaired remodeling partly due to altered autophagy gene expression.
Collapse
Affiliation(s)
- Ferdinand von Walden
- Department of Medicine, Center for Molecular Medicine L8:04, Karolinska Institute, 171 76 Stockholm, Sweden
| | | | | | | |
Collapse
|
42
|
Fanzani A, Conraads VM, Penna F, Martinet W. Molecular and cellular mechanisms of skeletal muscle atrophy: an update. J Cachexia Sarcopenia Muscle 2012; 3:163-79. [PMID: 22673968 PMCID: PMC3424188 DOI: 10.1007/s13539-012-0074-6] [Citation(s) in RCA: 234] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2012] [Accepted: 05/13/2012] [Indexed: 02/06/2023] Open
Abstract
Skeletal muscle atrophy is defined as a decrease in muscle mass and it occurs when protein degradation exceeds protein synthesis. Potential triggers of muscle wasting are long-term immobilization, malnutrition, severe burns, aging as well as various serious and often chronic diseases, such as chronic heart failure, obstructive lung disease, renal failure, AIDS, sepsis, immune disorders, cancer, and dystrophies. Interestingly, a cooperation between several pathophysiological factors, including inappropriately adapted anabolic (e.g., growth hormone, insulin-like growth factor 1) and catabolic proteins (e.g., tumor necrosis factor alpha, myostatin), may tip the balance towards muscle-specific protein degradation through activation of the proteasomal and autophagic systems or the apoptotic pathway. Based on the current literature, we present an overview of the molecular and cellular mechanisms that contribute to muscle wasting. We also focus on the multifacetted therapeutic approach that is currently employed to prevent the development of muscle wasting and to counteract its progression. This approach includes adequate nutritional support, implementation of exercise training, and possible pharmacological compounds.
Collapse
Affiliation(s)
- Alessandro Fanzani
- Department of Biomedical Sciences and Biotechnologies and Interuniversitary Institute of Myology (IIM), University of Brescia, viale Europa 11, 25123, Brescia, Italy,
| | | | | | | |
Collapse
|
43
|
Ebert SM, Dyle MC, Kunkel SD, Bullard SA, Bongers KS, Fox DK, Dierdorff JM, Foster ED, Adams CM. Stress-induced skeletal muscle Gadd45a expression reprograms myonuclei and causes muscle atrophy. J Biol Chem 2012; 287:27290-301. [PMID: 22692209 DOI: 10.1074/jbc.m112.374777] [Citation(s) in RCA: 161] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Diverse stresses including starvation and muscle disuse cause skeletal muscle atrophy. However, the molecular mechanisms of muscle atrophy are complex and not well understood. Here, we demonstrate that growth arrest and DNA damage-inducible 45a protein (Gadd45a) is a critical mediator of muscle atrophy. We identified Gadd45a through an unbiased search for potential downstream mediators of the stress-inducible, pro-atrophy transcription factor ATF4. We show that Gadd45a is required for skeletal muscle atrophy induced by three distinct skeletal muscle stresses: fasting, muscle immobilization, and muscle denervation. Conversely, forced expression of Gadd45a in muscle or cultured myotubes induces atrophy in the absence of upstream stress. We show that muscle-specific ATF4 knock-out mice have a reduced capacity to induce Gadd45a mRNA in response to stress, and as a result, they undergo less atrophy in response to fasting or muscle immobilization. Interestingly, Gadd45a is a myonuclear protein that induces myonuclear remodeling and a comprehensive program for muscle atrophy. Gadd45a represses genes involved in anabolic signaling and energy production, and it induces pro-atrophy genes. As a result, Gadd45a reduces multiple barriers to muscle atrophy (including PGC-1α, Akt activity, and protein synthesis) and stimulates pro-atrophy mechanisms (including autophagy and caspase-mediated proteolysis). These results elucidate a critical stress-induced pathway that reprograms muscle gene expression to cause atrophy.
Collapse
Affiliation(s)
- Scott M Ebert
- Department of and Molecular Physiology and Biophysics and Fraternal Order of Eagles Diabetes Research Center, Roy J. and Lucille A. Carver College of Medicine, The University of Iowa, Iowa City, Iowa 52242, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Jayaraman A, Liu M, Ye F, Walter GA, Vandenborne K. Regenerative responses in slow- and fast-twitch muscles following moderate contusion spinal cord injury and locomotor training. Eur J Appl Physiol 2012; 113:191-200. [DOI: 10.1007/s00421-012-2429-2] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2011] [Accepted: 05/15/2012] [Indexed: 11/30/2022]
|
45
|
Pereira BP, Tan BL, Han HC, Zou Y, Aung KZ, Leong DT. Intramuscular nerve damage in lacerated skeletal muscles may direct the inflammatory cytokine response during recovery. J Cell Biochem 2012; 113:2330-45. [DOI: 10.1002/jcb.24105] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
46
|
Wu Y, Zhao J, Zhao W, Pan J, Bauman WA, Cardozo CP. Nandrolone normalizes determinants of muscle mass and fiber type after spinal cord injury. J Neurotrauma 2012; 29:1663-75. [PMID: 22208735 DOI: 10.1089/neu.2011.2203] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Spinal cord injury (SCI) results in atrophy of skeletal muscle and changes from slow oxidative to fast glycolytic fibers, which may reflect reduced levels of peroxisome proliferator-activated receptor gamma coactivator-1α (PGC-1α), increased myostatin signaling, or both. In animals, testosterone reduces loss of muscle fiber cross-sectional area and activity of enzymes of energy metabolism. To identify the molecular mechanisms behind the benefits of androgens on paralyzed muscle, male rats were spinal cord transected and treated for 8 weeks with vehicle, testosterone at a physiological replacement dose, or testosterone plus nandrolone, an anabolic steroid. Treatments were initiated immediately after SCI and continued until the day animals were euthanized. In the SCI animals, gastrocnemius muscle mass was significantly increased by testosterone plus nandrolone, but not by testosterone alone. Both treatments significantly reduced nuclear content of Smad2/3 and mRNA levels of activin receptor IIB and follistatin-like 3. Testosterone alone or with nandrolone reversed SCI-induced declines in cellular and nuclear levels of PGC-1α protein and PGC-1α mRNA levels. For PGC-1α target genes, testosterone plus nandrolone partially reversed SCI-induced decreases in levels of proteins without corresponding increases in their mRNA levels. Thus, the findings demonstrate that following SCI, signaling through activin receptors and Smad2/3 is increased, and that androgens suppress activation of this signaling pathway. The findings also indicate that androgens upregulate PGC-1α in paralyzed muscle and promote its nuclear localization, but that these effects are insufficient to fully activate transcription of PGC-1α target genes. Furthermore, the transcription of these genes is not tightly coupled with their translation.
Collapse
Affiliation(s)
- Yong Wu
- Center of Excellence for the Medical Consequences of SCI, James J. Peters VA Medical Center, Bronx, NY, USA
| | | | | | | | | | | |
Collapse
|
47
|
Lee S, Shin J, Hong Y, Lee M, Kim K, Lee SR, Chang KT, Hong Y. Beneficial effects of melatonin on stroke-induced muscle atrophy in focal cerebral ischemic rats. Lab Anim Res 2012; 28:47-54. [PMID: 22474474 PMCID: PMC3315192 DOI: 10.5625/lar.2012.28.1.47] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2012] [Revised: 02/27/2012] [Accepted: 03/02/2012] [Indexed: 12/26/2022] Open
Abstract
MUSCLE ATROPHY IS THE RESULT OF TWO OPPOSING CONDITIONS THAT CAN BE FOUND IN PATHOLOGICAL OR DISEASED MUSCLES: an imbalance in protein synthesis and degradation mechanisms. Thus, we investigated whether exogenous melatonin could regulate muscle components in stroke-induced muscle atrophy in rats. Comparing muscle phenotypes, we found that long-term melatonin administration could influence muscle mass. Muscle atrophy-related genes, including muscle atrophy F-box (MAFbx) and muscle ring finger 1 (MuRF1) were significantly down-regulated in melatonin-administered rats in the gastrocnemius. However, only MAFbx at the mRNA level was attenuated in the soleus of melatonin-administered rats. Insulin-like growth factor-1 receptor (IGF-1R) was significantly over-expressed in melatonin-administered rats in both the gastrocnemius and soleus muscles. Comparing myosin heavy chain (MHC) components, in the gastrocnemius, expression of both slow- and fast-type isoforms were significantly enhanced in melatonin-administered rats. These results suggest that long-term exogenous melatonin-administration may have a prophylactic effect on muscle atrophy through the MuRF1/MAFbx signaling pathway, as well as a potential therapeutic effect on muscle atrophy through the IGF-1-mediated hypertrophic signaling pathway in a stroke animal model.
Collapse
Affiliation(s)
- Seunghoon Lee
- Department of Rehabilitation Science in Interdisciplinary PhD Program, Graduate School of Inje University, Gimhae, Korea
| | - Jinhee Shin
- Department of Physical Therapy, College of Biomedical Science & Engineering, Inje University, Gimhae, Korea
| | - Yunkyung Hong
- Department of Rehabilitation Science in Interdisciplinary PhD Program, Graduate School of Inje University, Gimhae, Korea
| | - Minkyung Lee
- Department of Rehabilitation Science in Interdisciplinary PhD Program, Graduate School of Inje University, Gimhae, Korea
| | - Koo Kim
- Department of Physical Therapy, College of Biomedical Science & Engineering, Inje University, Gimhae, Korea
| | - Sang-Rae Lee
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology, Ochang, Korea
| | - Kyu-Tae Chang
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology, Ochang, Korea
| | - Yonggeun Hong
- Department of Rehabilitation Science in Interdisciplinary PhD Program, Graduate School of Inje University, Gimhae, Korea
- Department of Physical Therapy, College of Biomedical Science & Engineering, Inje University, Gimhae, Korea
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology, Ochang, Korea
- Cardiovascular & Metabolic Disease Center, College of Biomedical Science & Engineering, Inje University, Gimhae, Korea
| |
Collapse
|
48
|
Esteves AM, Squarcini CFR, Lancellotti CLP, Tufik S, de Mello MT. Characteristics of muscle fibers in rats with limb movements during sleep after spinal cord injury. Eur Neurol 2012; 67:107-15. [PMID: 22236775 DOI: 10.1159/000334102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2011] [Accepted: 10/03/2011] [Indexed: 11/19/2022]
Abstract
BACKGROUND/AIMS Previous studies have demonstrated that spinal cord injury (SCI) results in changes in sleep patterns through increased arousals and limb movements during sleep. Dramatic changes in muscle myosin heavy-chain isoforms have also been reported. The aim of this study was to investigate the characteristics of muscle fibers after SCI in rats with limb movements during sleep. METHODS Forty male Wistar rats were divided into four groups: SHAM, SCI 3, 7 and 15 days. Animals were subjected to electrode insertion surgery, 24-hour baseline sleep recording, SCI, and subsequent sleep recording for 3, 7, or 15 consecutive days. In addition, the gastrocnemius muscle and spinal cord were collected for histopathological/histochemical analyses. RESULTS Our results indicate a rapid and progressive decrease in the cross-sectional area of type I fibers in the gastrocnemius muscle (35.76-24.74 μm(2)) after SCI. Additionally, we found SCI-induced changes in sleep patterns. Following SCI, we also observed limb movements in sleeping rats, as well as significant negative moderate correlations between type I fibers and limb movement. CONCLUSION Our study strengthened the hypothesis by correlation between changes in types of muscle fiber (decline in type I fibers) and an increase in limb movements during sleep after SCI.
Collapse
Affiliation(s)
- Andrea M Esteves
- Departamento de Psicobiologia, Universidade Federal de São Paulo, São Paulo, Brazil
| | | | | | | | | |
Collapse
|
49
|
Park S, Lee SK, Park K, Lee Y, Hong Y, Lee S, Jeon JC, Kim JH, Lee SR, Chang KT, Hong Y. Beneficial effects of endogenous and exogenous melatonin on neural reconstruction and functional recovery in an animal model of spinal cord injury. J Pineal Res 2012; 52:107-19. [PMID: 21854445 DOI: 10.1111/j.1600-079x.2011.00925.x] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The purpose of this study was to investigate the beneficial effects of endogenous and exogenous melatonin on functional recovery in an animal model of spinal cord injury (SCI). Eight-week-old male Sprague-Dawley (SD, 250-260 g) rats were used for contusion SCI surgery. All experimental groups were maintained under one of the following conditions: 12/12-hr light/dark (L/D) or 24:0-hr constant light (LL). Melatonin (10 mg/kg) was injected subcutaneously for 4 wk, twice daily (07:00, 19:00). Locomotor recovery, inducible nitric oxide synthase (iNOS), glial fibrillary acidic protein gene expression, and muscle atrophy-related genes, including muscle atrophy F-box (MAFbx) and muscle-specific ring-finger protein 1 (MuRF1) gene expression were evaluated. Furthermore, autophagic signaling such as Beclin-1 and LC3 protein expression was examined in the spinal cord and in skeletal muscle. The melatonin treatment resulted in increased hind-limb motor function and decreased iNOS mRNA expression in the L/D condition compared with the LL condition (P < 0.05), indicating that endogenous melatonin had neuroprotective effects. Furthermore, the MAFbx, MuRF1 mRNA level, and converted LC3 II protein expression were decreased in the melatonin-treated SCI groups under the LL (P < 0.05), possibly in response to the exogenous melatonin treatment. Therefore, it seems that both endogenous and exogenous melatonin contribute to neural recovery and to the prevention of skeletal muscle atrophy, promoting functional recovery after SCI. Finally, this study supports the benefit of endogenous melatonin and use of exogenous melatonin as a therapeutic intervention for SCI.
Collapse
Affiliation(s)
- Sookyoung Park
- Cardiovascular & Metabolic Disease Center, College of Biomedical Science & Engineering, Inje University, Gimhae, Korea
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Wu Y, Hou J, Collier L, Pan J, Hou L, Qin W, Bauman WA, Cardozo CP. The administration of high-dose methylprednisolone for 24 h reduced muscle size and increased atrophy-related gene expression in spinal cord-injured rats. Spinal Cord 2011; 49:867-73. [DOI: 10.1038/sc.2011.28] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
|