1
|
Wu J, Tang J, Huang D, Wang Y, Zhou E, Ru Q, Xu G, Chen L, Wu Y. Study on the comorbid mechanisms of sarcopenia and late-life depression. Behav Brain Res 2025; 485:115538. [PMID: 40122287 DOI: 10.1016/j.bbr.2025.115538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Revised: 03/06/2025] [Accepted: 03/09/2025] [Indexed: 03/25/2025]
Abstract
The increasing global aging population has brought greater focus to age-related diseases, particularly muscle-brain comorbidities such as sarcopenia and late-life depression. Sarcopenia, defined by the gradual loss of muscle mass and function, is notably prevalent among older individuals, while late-life depression profoundly affects their mental health and overall well-being. Epidemiological evidence suggests a high co-occurrence of these two conditions, although the precise biological mechanisms linking them remain inadequately understood. This review synthesizes the existing body of literature on sarcopenia and late-life depression, examining their definitions, prevalence, clinical presentations, and available treatments. The goal is to clarify the potential connections between these comorbidities and offer a theoretical framework for the development of future preventive and therapeutic strategies.
Collapse
Affiliation(s)
- Jiale Wu
- Institute of Intelligent Sport and Proactive Health, Department of Health and Physical Education, Jianghan University, Wuhan 430056, China
| | - Jun Tang
- Institute of Intelligent Sport and Proactive Health, Department of Health and Physical Education, Jianghan University, Wuhan 430056, China
| | - Di Huang
- Institute of Intelligent Sport and Proactive Health, Department of Health and Physical Education, Jianghan University, Wuhan 430056, China
| | - Yu Wang
- Institute of Intelligent Sport and Proactive Health, Department of Health and Physical Education, Jianghan University, Wuhan 430056, China
| | - Enyuan Zhou
- Institute of Intelligent Sport and Proactive Health, Department of Health and Physical Education, Jianghan University, Wuhan 430056, China
| | - Qin Ru
- Institute of Intelligent Sport and Proactive Health, Department of Health and Physical Education, Jianghan University, Wuhan 430056, China
| | - Guodong Xu
- Institute of Intelligent Sport and Proactive Health, Department of Health and Physical Education, Jianghan University, Wuhan 430056, China
| | - Lin Chen
- Institute of Intelligent Sport and Proactive Health, Department of Health and Physical Education, Jianghan University, Wuhan 430056, China.
| | - Yuxiang Wu
- Institute of Intelligent Sport and Proactive Health, Department of Health and Physical Education, Jianghan University, Wuhan 430056, China.
| |
Collapse
|
2
|
Gulley Cox LI, Dias N, Zhang C, Zhang Y, Gorniak SL. Effects of Type II Diabetes on upper extremity muscle characteristics in older adults. Neurosci Lett 2025; 844:138039. [PMID: 39549828 DOI: 10.1016/j.neulet.2024.138039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 10/28/2024] [Accepted: 11/11/2024] [Indexed: 11/18/2024]
Abstract
With one in every four older adults living with T2D and one in every two older adults meeting the criteria for prediabetes, neuromuscular changes due to T2D are likely to impact functional activities in this population. Limited work in evaluating motor unit number and size across muscles in the upper extremity in persons with Type II Diabetes (T2D) exists, mostly due to the traditional belief bias that the upper extremity is relatively spared in T2D as compared to the lower extremities. The purpose of the current study was to evaluate motor unit number and size (using electrophysiological motor unit number index (MUNIX) and motor unit size index (MUSIX)) across the upper extremity in older adults with T2D (n = 13) as compared to healthy age- and sex-matched controls (n = 12). Persons with T2D presented with more motor units and larger motor unit sizes (p < 0.05) as compared to age- and sex-matched control participants. These changes were not dependent upon muscle location within a limb, indicating systemic neuromuscular changes associated with T2D. These group effects were clarified when health state covariates (e.g., blood pressure) were accounted for. Findings are consistent with emerging data that show altered neuromuscular characteristics with health state considerations in persons with T2D.
Collapse
Affiliation(s)
- Lauren I Gulley Cox
- Department of Health and Human Performance, University of Houston, Houston, TX 77204, USA
| | - Nicholas Dias
- Department of Biomedical Engineering, University of Houston, Houston, TX 77204, USA
| | - Chuan Zhang
- Department of Biomedical Engineering, University of Houston, Houston, TX 77204, USA
| | - Yingchun Zhang
- Department of Biomedical Engineering, University of Houston, Houston, TX 77204, USA
| | - Stacey L Gorniak
- Department of Health and Human Performance, University of Houston, Houston, TX 77204, USA.
| |
Collapse
|
3
|
Iyer CC, Chugh D, Bobbili PJ, Iii AJB, Crum AE, Yi AF, Kaspar BK, Meyer KC, Burghes AHM, Arnold WD. Follistatin-induced muscle hypertrophy in aged mice improves neuromuscular junction innervation and function. Neurobiol Aging 2021; 104:32-41. [PMID: 33964607 PMCID: PMC8225567 DOI: 10.1016/j.neurobiolaging.2021.03.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Revised: 02/28/2021] [Accepted: 03/07/2021] [Indexed: 12/23/2022]
Abstract
Sarcopenia, or age-related loss of muscle mass and strength, is an important contributor to loss of physical function in older adults. The pathogenesis of sarcopenia is likely multifactorial, but recently the role of neurological degeneration, such as motor unit loss, has received increased attention. Here, we investigated the longitudinal effects of muscle hypertrophy (via overexpression of human follistatin, a myostatin antagonist) on neuromuscular integrity in C57BL/6J mice between the ages of 24 and 27 months. Following follistatin overexpression (delivered via self-complementary adeno-associated virus subtype 9 injection), muscle weight and torque production were significantly improved. Follistatin treatment resulted in improvements of neuromuscular junction innervation and transmission but had no impact on age-related losses of motor units. These studies demonstrate that follistatin overexpression-induced muscle hypertrophy not only increased muscle weight and torque production but also countered age-related degeneration at the neuromuscular junction in mice.
Collapse
Affiliation(s)
- Chitra C Iyer
- Department of Neurology, Neuromuscular Division, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Deepti Chugh
- Department of Neurology, Neuromuscular Division, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Prameela J Bobbili
- Department of Neurology, Neuromuscular Division, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Anton J Blatnik Iii
- Department of Biological Chemistry and Pharmacology, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Alexander E Crum
- Department of Neurology, Neuromuscular Division, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Allen F Yi
- The Ohio State University College of Medicine, Columbus, OH, USA
| | - Brian K Kaspar
- The Research Institute, Nationwide Children's Hospital, Columbus, OH, USA
| | - Kathrin C Meyer
- The Research Institute, Nationwide Children's Hospital, Columbus, OH, USA; Department of Pediatrics, The Ohio State University, Columbus, OH, USA
| | - Arthur H M Burghes
- Department of Biological Chemistry and Pharmacology, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - W David Arnold
- Department of Neurology, Neuromuscular Division, The Ohio State University Wexner Medical Center, Columbus, OH, USA; Department of Physical Medicine and Rehabilitation, The Ohio State University Wexner Medical Center, Columbus, OH, USA; Department of Neuroscience, The Ohio State University Wexner Medical Center, Columbus, OH, USA; Department of Physiology and Cell Biology, The Ohio State University Wexner Medical Center, Columbus, OH, USA.
| |
Collapse
|
4
|
Padilla CJ, Harrigan ME, Harris H, Schwab JM, Rutkove SB, Rich MM, Clark BC, Arnold WD. Profiling age-related muscle weakness and wasting: neuromuscular junction transmission as a driver of age-related physical decline. GeroScience 2021; 43:1265-1281. [PMID: 33895959 PMCID: PMC8190265 DOI: 10.1007/s11357-021-00369-3] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Accepted: 04/19/2021] [Indexed: 12/22/2022] Open
Abstract
Pathological age-related loss of skeletal muscle strength and mass contribute to impaired physical function in older adults. Factors that promote the development of these conditions remain incompletely understood, impeding development of effective and specific diagnostic and therapeutic approaches. Inconclusive evidence across species suggests disruption of action potential signal transmission at the neuromuscular junction (NMJ), the crucial connection between the nervous and muscular systems, as a possible contributor to age-related muscle dysfunction. Here we investigated age-related loss of NMJ function using clinically relevant, electrophysiological measures (single-fiber electromyography (SFEMG) and repetitive nerve stimulation (RNS)) in aged (26 months) versus young (6 months) F344 rats. Measures of muscle function (e.g., grip strength, peak plantarflexion contractility torque) and mass were assessed for correlations with physiological measures (e.g., indices of NMJ transmission). Other outcomes also included plantarflexion muscle contractility tetanic torque fade during 1-s trains of stimulation as well as gastrocnemius motor unit size and number. Profiling NMJ function in aged rats identified significant declines in NMJ transmission stability and reliability. Further, NMJ deficits were tightly correlated with hindlimb grip strength, gastrocnemius muscle weight, loss of peak contractility torque, degree of tetanic fade, and motor unit loss. Thus, these findings provide direct evidence for NMJ dysfunction as a potential mechanism of age-related muscle dysfunction pathogenesis and severity. These findings also suggest that NMJ transmission modulation may serve as a target for therapeutic development for age-related loss of physical function.
Collapse
Affiliation(s)
- Carlos J Padilla
- Division of Neuromuscular Diseases, Department of Neurology, The Ohio State University Wexner Medical Center, 1060 Carmack Road, Room 207, Columbus, OH, 43210, USA
| | - Markus E Harrigan
- Division of Neuromuscular Diseases, Department of Neurology, The Ohio State University Wexner Medical Center, 1060 Carmack Road, Room 207, Columbus, OH, 43210, USA
| | - Hallie Harris
- Division of Neuromuscular Diseases, Department of Neurology, The Ohio State University Wexner Medical Center, 1060 Carmack Road, Room 207, Columbus, OH, 43210, USA
| | - Jan M Schwab
- Division of Neuromuscular Diseases, Department of Neurology, The Ohio State University Wexner Medical Center, 1060 Carmack Road, Room 207, Columbus, OH, 43210, USA
- Belford Center for Spinal Cord Injury, The Ohio State University Wexner Medical Center, Columbus, OH, USA
- Department of Physical Medicine and Rehabilitation, The Ohio State University Wexner Medical Center, Columbus, OH, USA
- Center for Brain and Spinal Cord Repair, The Ohio State University Wexner Medical Center, Columbus, OH, USA
- The Neurological Institute, The Ohio State University Wexner Medical Center, Columbus, OH, USA
- Department of Neuroscience, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Seward B Rutkove
- Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Mark M Rich
- Department of Neuroscience, Cell Biology and Physiology, Wright State University, Dayton, OH, 45435, USA
| | - Brian C Clark
- Department of Biomedical Sciences, Ohio Musculoskeletal and Neurological Institute, Athens, OH, 45701, USA
| | - W David Arnold
- Division of Neuromuscular Diseases, Department of Neurology, The Ohio State University Wexner Medical Center, 1060 Carmack Road, Room 207, Columbus, OH, 43210, USA.
- Department of Physical Medicine and Rehabilitation, The Ohio State University Wexner Medical Center, Columbus, OH, USA.
- The Neurological Institute, The Ohio State University Wexner Medical Center, Columbus, OH, USA.
- Department of Neuroscience, The Ohio State University Wexner Medical Center, Columbus, OH, USA.
- Department of Physiology and Cell Biology, The Ohio State University Wexner Medical Center, Columbus, OH, USA.
| |
Collapse
|
5
|
Chugh D, Iyer CC, Bobbili P, Blatnik AJ, Kaspar BK, Meyer K, Burghes AH, Clark BC, Arnold WD. Voluntary wheel running with and without follistatin overexpression improves NMJ transmission but not motor unit loss in late life of C57BL/6J mice. Neurobiol Aging 2021; 101:285-296. [PMID: 33678425 PMCID: PMC8122043 DOI: 10.1016/j.neurobiolaging.2021.01.012] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Revised: 12/23/2020] [Accepted: 01/16/2021] [Indexed: 01/17/2023]
Abstract
Sarcopenia, or pathological loss of muscle mass and strength during aging, is an important contributor to loss of physical function in older adults. Sarcopenia is a multifactorial syndrome associated with intrinsic muscle and upstream neurological dysfunction. Exercise is well-established as an effective intervention for sarcopenia, but less is known about the long-term neurobiological impact of exercise. The goals of this study were to investigate the effects of exercise, alone or in combination with follistatin (FST) overexpression (antagonist of myostatin), on neuromuscular junction transmission and motor unit numbers in mice between the age of 22 and 27 months, ages at which prior studies have demonstrated that some motor unit loss is already evident. C57BL/6J mice underwent baseline assessment and were randomized to housing with or without voluntary running wheels and injection with adeno-associated virus to overexpress FST or vehicle. Groups for comparison included sedentary and running with and without FST. Longitudinal assessments showed significantly increased muscle mass and contractility in the 'running plus FST' group, but running, with and without FST, showed no effect on motor unit degeneration. In contrast, running, with and without FST, demonstrated marked improvement of neuromuscular junction transmission stability.
Collapse
Affiliation(s)
- Deepti Chugh
- Department of Neurology, Neuromuscular Division, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Chitra C Iyer
- Department of Neurology, Neuromuscular Division, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Prameela Bobbili
- Department of Neurology, Neuromuscular Division, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Anton J Blatnik
- Department of Biological Chemistry and Pharmacology, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Brian K Kaspar
- The Research Institute, Nationwide Children's Hospital, Columbus, OH, USA
| | - Kathrin Meyer
- The Research Institute, Nationwide Children's Hospital, Columbus, OH, USA; Department of Pediatrics, The Ohio State University, Columbus, OH, USA
| | - Arthur Hm Burghes
- Department of Biological Chemistry and Pharmacology, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Brian C Clark
- Ohio Musculoskeletal and Neurological Institute & the Department of Biomedical Sciences, Athens, OH, USA
| | - W David Arnold
- Department of Neurology, Neuromuscular Division, The Ohio State University Wexner Medical Center, Columbus, OH, USA; Department of Physical Medicine and Rehabilitation, The Ohio State University Wexner Medical Center, Columbus, OH, USA; Department of Neuroscience, The Ohio State University Wexner Medical Center, Columbus, OH, USA; Department of Physiology and Cell Biology, The Ohio State University Wexner Medical Center, Columbus, OH, USA.
| |
Collapse
|
6
|
Blasco A, Gras S, Mòdol-Caballero G, Tarabal O, Casanovas A, Piedrafita L, Barranco A, Das T, Pereira SL, Navarro X, Rueda R, Esquerda JE, Calderó J. Motoneuron deafferentation and gliosis occur in association with neuromuscular regressive changes during ageing in mice. J Cachexia Sarcopenia Muscle 2020; 11:1628-1660. [PMID: 32691534 PMCID: PMC7749545 DOI: 10.1002/jcsm.12599] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 06/05/2020] [Accepted: 06/15/2020] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND The cellular mechanisms underlying the age-associated loss of muscle mass and function (sarcopenia) are poorly understood, hampering the development of effective treatment strategies. Here, we performed a detailed characterization of age-related pathophysiological changes in the mouse neuromuscular system. METHODS Young, adult, middle-aged, and old (1, 4, 14, and 24-30 months old, respectively) C57BL/6J mice were used. Motor behavioural and electrophysiological tests and histological and immunocytochemical procedures were carried out to simultaneously analyse structural, molecular, and functional age-related changes in distinct cellular components of the neuromuscular system. RESULTS Ageing was not accompanied by a significant loss of spinal motoneurons (MNs), although a proportion (~15%) of them in old mice exhibited an abnormally dark appearance. Dark MNs were also observed in adult (~9%) and young (~4%) animals, suggesting that during ageing, some MNs undergo early deleterious changes, which may not lead to MN death. Old MNs were depleted of cholinergic and glutamatergic inputs (~40% and ~45%, respectively, P < 0.01), suggestive of age-associated alterations in MN excitability. Prominent microgliosis and astrogliosis [~93% (P < 0.001) and ~100% (P < 0.0001) increase vs. adults, respectively] were found in old spinal cords, with increased density of pro-inflammatory M1 microglia and A1 astroglia (25-fold and 4-fold increase, respectively, P < 0.0001). Ageing resulted in significant reductions in the nerve conduction velocity and the compound muscle action potential amplitude (~30%, P < 0.05, vs. adults) in old distal plantar muscles. Compared with adult muscles, old muscles exhibited significantly higher numbers of both denervated and polyinnervated neuromuscular junctions, changes in fibre type composition, higher proportion of fibres showing central nuclei and lipofuscin aggregates, depletion of satellite cells, and augmented expression of different molecules related to development, plasticity, and maintenance of neuromuscular junctions, including calcitonin gene-related peptide, growth associated protein 43, agrin, fibroblast growth factor binding protein 1, and transforming growth factor-β1. Overall, these alterations occurred at varying degrees in all the muscles analysed, with no correlation between the age-related changes observed and myofiber type composition or muscle topography. CONCLUSIONS Our data provide a global view of age-associated neuromuscular changes in a mouse model of ageing and help to advance understanding of contributing pathways leading to development of sarcopenia.
Collapse
Affiliation(s)
- Alba Blasco
- Unitat de Neurobiologia Cel·lular, Departament de Medicina Experimental, Facultat de Medicina, Universitat de Lleida, Institut de Recerca Biomèdica de Lleida (IRBLleida), Lleida, Spain
| | - Sílvia Gras
- Unitat de Neurobiologia Cel·lular, Departament de Medicina Experimental, Facultat de Medicina, Universitat de Lleida, Institut de Recerca Biomèdica de Lleida (IRBLleida), Lleida, Spain
| | - Guillem Mòdol-Caballero
- Grup de Neuroplasticitat i Regeneració, Institut de Neurociències, Departament de Biologia Cel·lular, Fisiologia i Immunologia, Universitat Autònoma de Barcelona, CIBERNED, Bellaterra, Spain
| | - Olga Tarabal
- Unitat de Neurobiologia Cel·lular, Departament de Medicina Experimental, Facultat de Medicina, Universitat de Lleida, Institut de Recerca Biomèdica de Lleida (IRBLleida), Lleida, Spain
| | - Anna Casanovas
- Unitat de Neurobiologia Cel·lular, Departament de Medicina Experimental, Facultat de Medicina, Universitat de Lleida, Institut de Recerca Biomèdica de Lleida (IRBLleida), Lleida, Spain
| | - Lídia Piedrafita
- Unitat de Neurobiologia Cel·lular, Departament de Medicina Experimental, Facultat de Medicina, Universitat de Lleida, Institut de Recerca Biomèdica de Lleida (IRBLleida), Lleida, Spain
| | | | - Tapas Das
- Abbott Nutrition Research and Development, Columbus, OH, USA
| | | | - Xavier Navarro
- Grup de Neuroplasticitat i Regeneració, Institut de Neurociències, Departament de Biologia Cel·lular, Fisiologia i Immunologia, Universitat Autònoma de Barcelona, CIBERNED, Bellaterra, Spain
| | - Ricardo Rueda
- Abbott Nutrition Research and Development, Granada, Spain
| | - Josep E Esquerda
- Unitat de Neurobiologia Cel·lular, Departament de Medicina Experimental, Facultat de Medicina, Universitat de Lleida, Institut de Recerca Biomèdica de Lleida (IRBLleida), Lleida, Spain
| | - Jordi Calderó
- Unitat de Neurobiologia Cel·lular, Departament de Medicina Experimental, Facultat de Medicina, Universitat de Lleida, Institut de Recerca Biomèdica de Lleida (IRBLleida), Lleida, Spain
| |
Collapse
|
7
|
Castets P, Ham DJ, Rüegg MA. The TOR Pathway at the Neuromuscular Junction: More Than a Metabolic Player? Front Mol Neurosci 2020; 13:162. [PMID: 32982690 PMCID: PMC7485269 DOI: 10.3389/fnmol.2020.00162] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Accepted: 08/05/2020] [Indexed: 12/18/2022] Open
Abstract
The neuromuscular junction (NMJ) is the chemical synapse connecting motor neurons and skeletal muscle fibers. NMJs allow all voluntary movements, and ensure vital functions like breathing. Changes in the structure and function of NMJs are hallmarks of numerous pathological conditions that affect muscle function including sarcopenia, the age-related loss of muscle mass and function. However, the molecular mechanisms leading to the morphological and functional perturbations in the pre- and post-synaptic compartments of the NMJ remain poorly understood. Here, we discuss the role of the metabolic pathway associated to the kinase TOR (Target of Rapamycin) in the development, maintenance and alterations of the NMJ. This is of particular interest as the TOR pathway has been implicated in aging, but its role at the NMJ is still ill-defined. We highlight the respective functions of the two TOR-associated complexes, TORC1 and TORC2, and discuss the role of localized protein synthesis and autophagy regulation in motor neuron terminals and sub-synaptic regions of muscle fibers and their possible effects on NMJ maintenance.
Collapse
Affiliation(s)
- Perrine Castets
- Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | | | | |
Collapse
|
8
|
Khosa S, Trikamji B, Khosa GS, Khanli HM, Mishra SK. An Overview of Neuromuscular Junction Aging Findings in Human and Animal Studies. Curr Aging Sci 2020; 12:28-34. [PMID: 31161982 PMCID: PMC6971950 DOI: 10.2174/1874609812666190603165746] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Revised: 05/14/2019] [Accepted: 05/14/2019] [Indexed: 12/15/2022]
Abstract
Background: Aging is a complex irreversible process that is not only related to an individual’s genetic make-up but also to lifestyle choices and environmental exposures. Like every other structure in human body, the Neuromuscular Junction (NMJ) is not averse to aging. Objectives: The prime objective is to analyse the microscopic and macroscopic changes at the NMJs with aging. Methods: For the purpose of review we evaluated data from resources like Pubmed, Ovid, UCLA libraries and USC libraries. Results: We review various morphological, physiological, immunological, and biochemical changes in NMJs with aging and their management. Conclusion: The alterations in NMJs secondary to aging are inevitable. It is vital that neurologists clearly understand the pathophysiology of NMJ aging and differentiate between physiological and pathological effects of aging. With the current knowledge of science, the changes in NMJ aging can be better prevented rather than cured.
Collapse
Affiliation(s)
- Shaweta Khosa
- Department of Neurology, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, CA, United States
| | - Bhavesh Trikamji
- Department of Neurology, Harbor UCLA Medical Center, Torrance, CA 90502, United States
| | - Gurveer S Khosa
- Department of Medicine, Indira Gandhi Medical College, Shimla, Himachal Pradesh, India
| | - Hadi M Khanli
- Department of Neurology, George Washington University, Washington, DC 20052, United States
| | - Shri K Mishra
- Department of Neurology, University of Southern California, Keck School of Medicine, Los Angeles, CA 90033, United States
| |
Collapse
|
9
|
Jamali A, Shahrbanian S, Morteza Tayebi S. The Effects of Exercise Training on the Brain-Derived Neurotrophic Factor (BDNF) in the Patients with Type 2 Diabetes: A Systematic Review of the Randomized Controlled Trials. J Diabetes Metab Disord 2020; 19:633-643. [PMID: 32550216 DOI: 10.1007/s40200-020-00529-w] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Accepted: 04/13/2020] [Indexed: 02/06/2023]
Abstract
Purpose Glucose dysregulation is one of the distinctive features of type 2 diabetes that is associated with an increased risk of cognitive impairment and dementia. The low concentrations of brain-derived neurotrophic factor (BDNF) are reported in people with insulin resistance, metabolic syndrome, and type 2 diabetes. BDNF can be increased by an adjustment in lifestyle including caloric restriction and exercise training. Studies have reported controversial findings about physical activity and its association with BDNF, but there is no comprehensive conclusions on this issue. The aim of this study was to systematically review the effects of exercise training on BDNF levels in patients with type 2 diabetes. Methods The electronic databases of Embase, Pedro, PubMed, Medline, Cochrane Library, as well as the Google Scholar search engine were used to obtain the related data about the role of exercise training on BDNF levels in patients with type 2 diabetes. The search period was set from inception to August 2019. Keywords of "exercise", "training", "physical activity", "brain-derived neurotrophic factor", "type 2 diabetes", and "randomized clinical trials", were used in persian and English. The PEDro scale was used to evaluate the quality of the included articles. Results. Finally, 11 articles (four human and seven animal articles) with medium to high quality were included in the study which 5 articles reported elevation (one human and four animal articles), 4 articles reported a reduction (one human and three animal articles), and 2 articles reported no changes (both of them in human articles) in BDNF level following the exercise training. Conclusion Decreased energy intake and increased energy expenditure through exercise training may modulate BDNF levels in patients with type 2 diabetes.
Collapse
Affiliation(s)
- Afsaneh Jamali
- Department of Sport Science, Faculty of Humanities, Tarbiat Modares University, Tehran, Iran
| | - Shahnaz Shahrbanian
- Department of Sport Science, Faculty of Humanities, Tarbiat Modares University, Tehran, Iran
| | - Seyed Morteza Tayebi
- Department of Exercise Physiology, Faculty of Sport Science, Allameh Tabataba'i University, Tehran, Iran
| |
Collapse
|
10
|
Borzuola R, Giombini A, Torre G, Campi S, Albo E, Bravi M, Borrione P, Fossati C, Macaluso A. Central and Peripheral Neuromuscular Adaptations to Ageing. J Clin Med 2020; 9:jcm9030741. [PMID: 32182904 PMCID: PMC7141192 DOI: 10.3390/jcm9030741] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 02/27/2020] [Accepted: 03/04/2020] [Indexed: 12/31/2022] Open
Abstract
Ageing is accompanied by a severe muscle function decline presumably caused by structural and functional adaptations at the central and peripheral level. Although researchers have reported an extensive analysis of the alterations involving muscle intrinsic properties, only a limited number of studies have recognised the importance of the central nervous system, and its reorganisation, on neuromuscular decline. Neural changes, such as degeneration of the human cortex and function of spinal circuitry, as well as the remodelling of the neuromuscular junction and motor units, appear to play a fundamental role in muscle quality decay and culminate with considerable impairments in voluntary activation and motor performance. Modern diagnostic techniques have provided indisputable evidence of a structural and morphological rearrangement of the central nervous system during ageing. Nevertheless, there is no clear insight on how such structural reorganisation contributes to the age-related functional decline and whether it is a result of a neural malfunction or serves as a compensatory mechanism to preserve motor control and performance in the elderly population. Combining leading-edge techniques such as high-density surface electromyography (EMG) and improved diagnostic procedures such as functional magnetic resonance imaging (fMRI) or high-resolution electroencephalography (EEG) could be essential to address the unresolved controversies and achieve an extensive understanding of the relationship between neural adaptations and muscle decline.
Collapse
Affiliation(s)
- Riccardo Borzuola
- Department of Movement, Human and Health Sciences, University of Rome “Foro Italico”, 00135 Rome, Italy; (R.B.); (A.G.); (P.B.); (C.F.); (A.M.)
| | - Arrigo Giombini
- Department of Movement, Human and Health Sciences, University of Rome “Foro Italico”, 00135 Rome, Italy; (R.B.); (A.G.); (P.B.); (C.F.); (A.M.)
| | - Guglielmo Torre
- Department of Orthopaedic And Trauma Surgery, Campus Bio-Medico University of Rome, 00128 Rome, Italy; (S.C.); (E.A.)
- Correspondence: ; Tel.: +6-225-418-825
| | - Stefano Campi
- Department of Orthopaedic And Trauma Surgery, Campus Bio-Medico University of Rome, 00128 Rome, Italy; (S.C.); (E.A.)
| | - Erika Albo
- Department of Orthopaedic And Trauma Surgery, Campus Bio-Medico University of Rome, 00128 Rome, Italy; (S.C.); (E.A.)
| | - Marco Bravi
- Department of Physical Medicine and Rehabilitation, Campus Bio-Medico University of Rome, 00128 Rome, Italy;
| | - Paolo Borrione
- Department of Movement, Human and Health Sciences, University of Rome “Foro Italico”, 00135 Rome, Italy; (R.B.); (A.G.); (P.B.); (C.F.); (A.M.)
| | - Chiara Fossati
- Department of Movement, Human and Health Sciences, University of Rome “Foro Italico”, 00135 Rome, Italy; (R.B.); (A.G.); (P.B.); (C.F.); (A.M.)
| | - Andrea Macaluso
- Department of Movement, Human and Health Sciences, University of Rome “Foro Italico”, 00135 Rome, Italy; (R.B.); (A.G.); (P.B.); (C.F.); (A.M.)
| |
Collapse
|
11
|
Spartano NL, Davis-Plourde KL, Himali JJ, Murabito JM, Vasan RS, Beiser AS, Seshadri S. Self-Reported Physical Activity and Relations to Growth and Neurotrophic Factors in Diabetes Mellitus: The Framingham Offspring Study. J Diabetes Res 2019; 2019:2718465. [PMID: 30729134 PMCID: PMC6343169 DOI: 10.1155/2019/2718465] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2018] [Revised: 11/28/2018] [Accepted: 12/03/2018] [Indexed: 01/02/2023] Open
Abstract
AIMS Circulating insulin-like growth factor- (IGF-) 1, vascular endothelial growth factor (VEGF), and brain-derived neurotrophic factor (BDNF) levels are often lower in individuals with diabetes mellitus (DM) and are important for repairing vascular and neuronal dysfunction. The purpose of this investigation was to determine the cross-sectional relations of physical activity to circulating concentrations of IGF-1, VEGF, and BDNF in individuals with and without DM. METHODS In 1730 participants from the Framingham Offspring Study examination cycle 7, including those with DM (n = 179, mean age 64 years, 39% women) and without DM (n = 1551, mean age 60 years, 46% women), we related self-reported physical activity variables to circulating concentrations of IGF-1, VEGF, and BDNF using linear multivariable regression models. We also tested for interactions by age. Participants with prevalent cardiovascular disease, stroke, and dementia or taking hormone replacement therapy were excluded. RESULTS In participants with DM, more ambulatory physical activity was associated with higher IGF-1 levels (β ± standard error (SE) = 0.22 ± 0.08, p = 0.009), and more total physical activity was related to higher BDNF levels (β ± SE = 0.18 ± 0.08, p = 0.035), but physical activity was not significantly related to circulating VEGF. In participants without DM, no associations were observed. Moreover, in the examination of interactions by age, the association of ambulatory physical activity with IGF-1 levels was only observed in older adults with DM (age ≥ 60 years, β ± SE = 0.23 ± 0.11, p = 0.042) but not in middle-aged adults with DM (age < 60 years, β ± SE = 0.06 ± 0.13, p = 0.645). CONCLUSION Our results suggest that more physical activity is associated with higher circulating IGF-1 and BDNF in participants with DM. These results, dissecting interactions by both age and DM status, may also help to explain some of the inconsistent results in studies relating physical activity to growth and neurotrophic factors.
Collapse
Affiliation(s)
- Nicole L. Spartano
- Department of Endocrinology, Diabetes, Nutrition & Weight Management, Boston University School of Medicine (BUSM), Boston, MA, USA
- Framingham Heart Study, Framingham, MA, USA
| | - Kendra L. Davis-Plourde
- Framingham Heart Study, Framingham, MA, USA
- Department of Biostatistics, Boston University School of Public Health (BUSPH), Boston, MA, USA
| | - Jayandra J. Himali
- Framingham Heart Study, Framingham, MA, USA
- Department of Biostatistics, Boston University School of Public Health (BUSPH), Boston, MA, USA
- Department of Neurology, BUSM, Boston, MA, USA
| | - Joanne M. Murabito
- Framingham Heart Study, Framingham, MA, USA
- Departments of Medicine and Epidemiology, BUSM and BUSPH, Boston, MA, USA
| | - Ramachandran S. Vasan
- Framingham Heart Study, Framingham, MA, USA
- Departments of Medicine and Epidemiology, BUSM and BUSPH, Boston, MA, USA
| | - Alexa S. Beiser
- Framingham Heart Study, Framingham, MA, USA
- Department of Biostatistics, Boston University School of Public Health (BUSPH), Boston, MA, USA
- Department of Neurology, BUSM, Boston, MA, USA
| | - Sudha Seshadri
- Framingham Heart Study, Framingham, MA, USA
- Department of Neurology, BUSM, Boston, MA, USA
- Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases, University of Texas Health Sciences Center, San Antonio, TX, USA
| |
Collapse
|
12
|
Shanmukha S, Narayanappa G, Nalini A, Alladi PA, Raju TR. Sporadic amyotrophic lateral sclerosis (SALS) - skeletal muscle response to cerebrospinal fluid from SALS patients in a rat model. Dis Model Mech 2018; 11:11/4/dmm031997. [PMID: 29666144 PMCID: PMC5963857 DOI: 10.1242/dmm.031997] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Accepted: 03/05/2018] [Indexed: 01/17/2023] Open
Abstract
Skeletal muscle atrophy is the most prominent feature of amyotrophic lateral sclerosis (ALS), an adult-onset neurodegenerative disease of motor neurons. However, the contribution of skeletal muscle to disease progression remains elusive. Our previous studies have shown that intrathecal injection of cerebrospinal fluid from sporadic ALS patients (ALS-CSF) induces several degenerative changes in motor neurons and glia of neonatal rats. Here, we describe various pathologic events in the rat extensor digitorum longus muscle following intrathecal injection of ALS-CSF. Adenosine triphosphatase staining and electron microscopic (EM) analysis revealed significant atrophy and grouping of type 2 fibres in ALS-CSF-injected rats. Profound neuromuscular junction (NMJ) damage, such as fragmentation accompanied by denervation, were revealed by α-bungarotoxin immunostaining. Altered expression of key NMJ proteins, rapsyn and calpain, was also observed by immunoblotting. In addition, EM analysis showed sarcolemmal folding, Z-line streaming, structural alterations of mitochondria and dilated sarcoplasmic reticulum. The expression of trophic factors was affected, with significant downregulation of vascular endothelial growth factor (VEGF), marginal reduction in insulin-like growth factor-1 (IGF-1), and upregulation of brain-derived neurotrophic factor (BDNF) and glial-derived neurotrophic factor (GDNF). However, motor neurons might be unable to harness the enhanced levels of BDNF and GDNF, owing to impaired NMJs. We propose that ALS-CSF triggers motor neuronal degeneration, resulting in pathological changes in the skeletal muscle. Muscle damage further aggravates the motor neuronal pathology, because of the interdependency between them. This sets in a vicious cycle, leading to rapid and progressive loss of motor neurons, which could explain the relentless course of ALS.This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
- Shruthi Shanmukha
- Department of Neurophysiology, National Institute of Mental Health and Neurosciences, Hosur Road, Bangalore 560 029, India
| | - Gayathri Narayanappa
- Department of Neuropathology, National Institute of Mental Health and Neurosciences, Hosur Road, Bangalore 560 029, India
| | - Atchayaram Nalini
- Department of Neurology, National Institute of Mental Health and Neurosciences, Hosur Road, Bangalore 560 029, India
| | - Phalguni Anand Alladi
- Department of Neurophysiology, National Institute of Mental Health and Neurosciences, Hosur Road, Bangalore 560 029, India
| | - Trichur R Raju
- Department of Neurophysiology, National Institute of Mental Health and Neurosciences, Hosur Road, Bangalore 560 029, India
| |
Collapse
|
13
|
Bendella H, Rink S, Grosheva M, Sarikcioglu L, Gordon T, Angelov DN. Putative roles of soluble trophic factors in facial nerve regeneration, target reinnervation, and recovery of vibrissal whisking. Exp Neurol 2017; 300:100-110. [PMID: 29104116 DOI: 10.1016/j.expneurol.2017.10.029] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2017] [Revised: 10/25/2017] [Accepted: 10/30/2017] [Indexed: 12/15/2022]
Abstract
It is well-known that, after nerve transection and surgical repair, misdirected regrowth of regenerating motor axons may occur in three ways. The first way is that the axons enter into endoneurial tubes that they did not previously occupy, regenerate through incorrect fascicles and reinnervate muscles that they did not formerly supply. Consequently the activation of these muscles results in inappropriate movements. The second way is that, in contrast with the precise target-directed pathfinding by elongating motor nerves during embryonic development, several axons rather than a single axon grow out from each transected nerve fiber. The third way of misdirection occurs by the intramuscular terminal branching (sprouting) of each regenerating axon to culminate in some polyinnervation of neuromuscular junctions, i.e. reinnervation of junctions by more than a single axon. Presently, "fascicular" or "topographic specificity" cannot be achieved and hence target-directed nerve regeneration is, as yet, unattainable. Nonetheless, motor and sensory reinnervation of appropriate endoneurial tubes does occur and can be promoted by brief nerve electrical stimulation. This review considers the expression of neurotrophic factors in the neuromuscular system and how this expression can promote functional recovery, with emphasis on the whisking of vibrissae on the rat face in relationship to the expression of the factors. Evidence is reviewed for a role of neurotrophic factors as short-range diffusible sprouting stimuli in promoting complete functional recovery of vibrissal whisking in blind Sprague Dawley (SD)/RCS rats but not in SD rats with normal vision, after facial nerve transection and surgical repair. Briefly, a complicated time course of growth factor expression in the nerves and denervated muscles include (1) an early increase in FGF2 and IGF2, (2) reduced NGF between 2 and 14days after nerve transection and surgical repair, (3) a late rise in BDNF and (4) reduced IGF1 protein in the denervated muscles at 28days. These findings suggest that recovery of motor function after peripheral nerve injury is due, at least in part, to a complex regulation of nerve injury-associated neurotrophic factors and cytokines at the neuromuscular junctions of denervated muscles. In particular, the increase of FGF2 and concomittant decrease of NGF during the first week after facial nerve-nerve anastomosis in SD/RCS blind rats may prevent intramuscular axon sprouting and, in turn, reduce poly-innervation of the neuromuscular junction.
Collapse
Affiliation(s)
- Habib Bendella
- Department of Neurosurgery, University of Witten/Herdecke, Cologne Merheim Medical Center (CMMC), Cologne, Germany
| | - Svenja Rink
- Department of Prosthetic Dentistry, School of Dental and Oral Medicine, University of Cologne, Germany
| | - Maria Grosheva
- Department of Oto-Rhino-Laryngology, University of Cologne, Germany
| | | | - Tessa Gordon
- Department of Surgery, The Hospital for Sick Children, Toronto, Ontario M5G 1X8, Canada
| | | |
Collapse
|
14
|
Grosheva M, Nohroudi K, Schwarz A, Rink S, Bendella H, Sarikcioglu L, Klimaschewski L, Gordon T, Angelov DN. Comparison of trophic factors' expression between paralyzed and recovering muscles after facial nerve injury. A quantitative analysis in time course. Exp Neurol 2016; 279:137-148. [PMID: 26940083 DOI: 10.1016/j.expneurol.2016.02.020] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2015] [Revised: 02/07/2016] [Accepted: 02/26/2016] [Indexed: 01/08/2023]
Abstract
After peripheral nerve injury, recovery of motor performance negatively correlates with the poly-innervation of neuromuscular junctions (NMJ) due to excessive sprouting of the terminal Schwann cells. Denervated muscles produce short-range diffusible sprouting stimuli, of which some are neurotrophic factors. Based on recent data that vibrissal whisking is restored perfectly during facial nerve regeneration in blind rats from the Sprague Dawley (SD)/RCS strain, we compared the expression of brain derived neurotrophic factor (BDNF), fibroblast growth factor-2 (FGF2), insulin growth factors 1 and 2 (IGF1, IGF2) and nerve growth factor (NGF) between SD/RCS and SD-rats with normal vision but poor recovery of whisking function after facial nerve injury. To establish which trophic factors might be responsible for proper NMJ-reinnervation, the transected facial nerve was surgically repaired (facial-facial anastomosis, FFA) for subsequent analysis of mRNA and proteins expressed in the levator labii superioris muscle. A complicated time course of expression included (1) a late rise in BDNF protein that followed earlier elevated gene expression, (2) an early increase in FGF2 and IGF2 protein after 2 days with sustained gene expression, (3) reduced IGF1 protein at 28 days coincident with decline of raised mRNA levels to baseline, and (4) reduced NGF protein between 2 and 14 days with maintained gene expression found in blind rats but not the rats with normal vision. These findings suggest that recovery of motor function after peripheral nerve injury is due, at least in part, to a complex regulation of lesion-associated neurotrophic factors and cytokines in denervated muscles. The increase of FGF-2 protein and concomittant decrease of NGF (with no significant changes in BDNF or IGF levels) during the first week following FFA in SD/RCS blind rats possibly prevents the distal branching of regenerating axons resulting in reduced poly-innervation of motor endplates.
Collapse
Affiliation(s)
- Maria Grosheva
- Department of Oto-Rhino-Laryngology, University of Cologne, Germany
| | | | - Alisa Schwarz
- Department of Anatomy I, University of Cologne, Germany
| | - Svenja Rink
- Department of Anatomy I, University of Cologne, Germany
| | - Habib Bendella
- Department of Neurosurgery, Hospital Merheim, University of Witten-Herdecke, Cologne, Germany
| | | | - Lars Klimaschewski
- Division of Neuroanatomy Innsbruck Medical University, 6020 Innsbruck, Austria
| | - Tessa Gordon
- Department of Surgery,The Hospital for Sick Children, Toronto, Ontario M5G 1X8, Canada
| | | |
Collapse
|
15
|
Messi ML, Li T, Wang ZM, Marsh AP, Nicklas B, Delbono O. Resistance Training Enhances Skeletal Muscle Innervation Without Modifying the Number of Satellite Cells or their Myofiber Association in Obese Older Adults. J Gerontol A Biol Sci Med Sci 2015; 71:1273-80. [PMID: 26447161 DOI: 10.1093/gerona/glv176] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2015] [Accepted: 09/17/2015] [Indexed: 11/14/2022] Open
Abstract
Studies in humans and animal models provide compelling evidence for age-related skeletal muscle denervation, which may contribute to muscle fiber atrophy and loss. Skeletal muscle denervation seems relentless; however, long-term, high-intensity physical activity appears to promote muscle reinnervation. Whether 5-month resistance training (RT) enhances skeletal muscle innervation in obese older adults is unknown. This study found that neural cell-adhesion molecule, NCAM+ muscle area decreased with RT and was inversely correlated with muscle strength. NCAM1 and RUNX1 gene transcripts significantly decreased with the intervention. Type I and type II fiber grouping in the vastus lateralis did not change significantly but increases in leg press and knee extensor strength inversely correlated with type I, but not with type II, fiber grouping. RT did not modify the total number of satellite cells, their number per area, or the number associated with specific fiber subtypes or innervated/denervated fibers. Our results suggest that RT has a beneficial impact on skeletal innervation, even when started late in life by sedentary obese older adults.
Collapse
Affiliation(s)
- María Laura Messi
- Department of Internal Medicine, Section on Gerontology and Geriatric Medicine and J Paul Sticht Center on Aging, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Tao Li
- Department of Internal Medicine, Section on Gerontology and Geriatric Medicine and J Paul Sticht Center on Aging, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Zhong-Min Wang
- Department of Internal Medicine, Section on Gerontology and Geriatric Medicine and J Paul Sticht Center on Aging, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Anthony P Marsh
- Department of Health and Exercise Science, Wake Forest University, Winston-Salem, North Carolina
| | - Barbara Nicklas
- Department of Internal Medicine, Section on Gerontology and Geriatric Medicine and J Paul Sticht Center on Aging, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Osvaldo Delbono
- Department of Internal Medicine, Section on Gerontology and Geriatric Medicine and J Paul Sticht Center on Aging, Wake Forest School of Medicine, Winston-Salem, North Carolina.
| |
Collapse
|
16
|
Park KHJ. Mechanisms of Muscle Denervation in Aging: Insights from a Mouse Model of Amyotrophic Lateral Sclerosis. Aging Dis 2015; 6:380-9. [PMID: 26425392 DOI: 10.14336/ad.2015.0506] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2015] [Accepted: 05/06/2015] [Indexed: 12/31/2022] Open
Abstract
Muscle denervation at the neuromuscular junction (NMJ) is thought to be a contributing factor in age-related muscle weakness. Therefore, understanding the mechanisms that modulate NMJ innervation is a key to developing therapies to combat age-related muscle weakness affecting the elderly. Two mouse models, one lacking the Cu/Zn superoxide dismutase (SOD1) gene and another harboring the transgenic mutant human SOD1 gene, display progressive changes at the NMJ, including muscle endplate fragmentation, nerve terminal sprouting, and denervation. These changes at the NMJ share many of the common features observed in the NMJs of aged mice. In this review, research findings demonstrating the effects of PGC-1α, IGF-1, GDNF, MyoD, myogenin, and miR-206 on NMJ innervation patterns in the G93A SOD1 mice will be highlighted in the context of age-related muscle denervation.
Collapse
Affiliation(s)
- Kevin H J Park
- Department of Psychology and Neuroscience Program, Central Michigan University, Mount Pleasant, MI 48859, USA
| |
Collapse
|
17
|
Moyer AL, Wagner KR. Mammalian Mss51 is a skeletal muscle-specific gene modulating cellular metabolism. J Neuromuscul Dis 2015; 2:371-385. [PMID: 26634192 PMCID: PMC4664537 DOI: 10.3233/jnd-150119] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
BACKGROUND The transforming growth factor β (TGF-β) signaling pathways modulate skeletal muscle growth, regeneration, and cellular metabolism. Several recent gene expression studies have shown that inhibition of myostatin and TGF-β1 signaling consistently leads to a significant reduction in expression of Mss51, also named Zmynd17. The function of mammalian Mss51 is unknown although a putative homolog in yeast is a mitochondrial translational activator. OBJECTIVE The objective of this work was to characterize mammalian Mss51. METHODS Quantitative RT-PCR and immunoblot of subcellular fractionation were used to determine expression patterns and localization of Mss51. The CRISPR/Cas9 system was used to reduce expression of Mss51 in C2C12 myoblasts and the function of Mss51 was evaluated in assays of proliferation, differentiation and cellular metabolism. RESULTS Mss51 was predominantly expressed in skeletal muscle and in those muscles dominated by fast-twitch fibers. In vitro, its expression was upregulated upon differentiation of C2C12 myoblasts into myotubes. Expression of Mss51 was modulated in response to altered TGF-β family signaling. In human muscle, Mss51 localized to the mitochondria. Its genetic disruption resulted in increased levels of cellular ATP, β-oxidation, glycolysis, and oxidative phosphorylation. CONCLUSIONS Mss51 is a novel, skeletal muscle-specific gene and a key target of myostatin and TGF-β1 signaling. Unlike myostatin, TGF-β1 and IGF-1, Mss51 does not regulate myoblast proliferation or differentiation. Rather, Mss51 appears to be one of the effectors of these growth factors on metabolic processes including fatty acid oxidation, glycolysis and oxidative phosphorylation.
Collapse
Affiliation(s)
- Adam L. Moyer
- The Hugo W. Moser Research Institute, Kennedy Krieger Institute, Baltimore, MD, USA
- Graduate Program in Cellular and Molecular Medicine, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Kathryn R. Wagner
- The Hugo W. Moser Research Institute, Kennedy Krieger Institute, Baltimore, MD, USA
- Graduate Program in Cellular and Molecular Medicine, Johns Hopkins School of Medicine, Baltimore, MD, USA
- Departments of Neurology and Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD, USA
| |
Collapse
|
18
|
Gonzalez-Freire M, de Cabo R, Studenski SA, Ferrucci L. The Neuromuscular Junction: Aging at the Crossroad between Nerves and Muscle. Front Aging Neurosci 2014; 6:208. [PMID: 25157231 PMCID: PMC4127816 DOI: 10.3389/fnagi.2014.00208] [Citation(s) in RCA: 215] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2014] [Accepted: 07/25/2014] [Indexed: 01/19/2023] Open
Abstract
Aging is associated with a progressive loss of muscle mass and strength and a decline in neurophysiological functions. Age-related neuromuscular junction (NMJ) plays a key role in musculoskeletal impairment that occurs with aging. However, whether changes in the NMJ precede or follow the decline of muscle mass and strength remains unresolved. Many factors such as mitochondrial dysfunction, oxidative stress, inflammation, changes in the innervation of muscle fibers, and mechanical properties of the motor units probably perform an important role in NMJ degeneration and muscle mass and strength decline in late life. This review addresses the primary events that might lead to NMJ dysfunction with aging, including studies on biomarkers, signaling pathways, and animal models. Interventions such as caloric restriction and exercise may positively affect the NMJ through this mechanism and attenuate the age-related progressive impairment in motor function.
Collapse
Affiliation(s)
- Marta Gonzalez-Freire
- Translational Gerontology Branch, National Institute on Aging, Intramural Research Program, National Institutes of Health , Baltimore, MD , USA ; Longitudinal Studies Section, Baltimore Longitudinal Study of Aging, National Institute on Aging, National Institutes of Health , Baltimore, MD , USA
| | - Rafael de Cabo
- Translational Gerontology Branch, National Institute on Aging, Intramural Research Program, National Institutes of Health , Baltimore, MD , USA
| | - Stephanie A Studenski
- Translational Gerontology Branch, National Institute on Aging, Intramural Research Program, National Institutes of Health , Baltimore, MD , USA ; Longitudinal Studies Section, Baltimore Longitudinal Study of Aging, National Institute on Aging, National Institutes of Health , Baltimore, MD , USA
| | - Luigi Ferrucci
- Translational Gerontology Branch, National Institute on Aging, Intramural Research Program, National Institutes of Health , Baltimore, MD , USA ; Longitudinal Studies Section, Baltimore Longitudinal Study of Aging, National Institute on Aging, National Institutes of Health , Baltimore, MD , USA
| |
Collapse
|
19
|
Cheng A, Morsch M, Murata Y, Ghazanfari N, Reddel SW, Phillips WD. Sequence of age-associated changes to the mouse neuromuscular junction and the protective effects of voluntary exercise. PLoS One 2013; 8:e67970. [PMID: 23844140 PMCID: PMC3701007 DOI: 10.1371/journal.pone.0067970] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2013] [Accepted: 05/23/2013] [Indexed: 11/18/2022] Open
Abstract
Loss of connections between motor neurons and skeletal muscle fibers contribute to motor impairment in old age, but the sequence of age-associated changes that precede loss of the neuromuscular synapse remains uncertain. Here we determine changes in the size of neuromuscular synapses within the tibialis anterior muscle across the life span of C57BL/6J mice. Immunofluorescence, confocal microscopy and morphometry were used to measure the area occupied by nerve terminal synaptophysin staining and postsynaptic acetylcholine receptors at motor endplates of 2, 14, 19, 22, 25 and 28month old mice. The key findings were: 1) At middle age (14-months) endplate acetylcholine receptors occupied 238±11 µm2 and nerve terminal synaptophysin 168±14 µm2 (mean ± SEM). 2) Between 14-months and 19-months (onset of old age) the area occupied by postsynaptic acetylcholine receptors declined 30%. At many endplates the large acetylcholine receptor plaque became fragmented into multiple smaller acetylcholine receptor clusters. 3) Between 19- and 25-months, the fraction of endplate acetylcholine receptors covered by synaptophysin fell 21%. By 28-months, half of the endplates imaged retained ≤50 µm2 area of synaptophysin staining. 4) Within aged muscles, the degree to which an endplate remained covered by synaptophysin did not depend upon the total area of acetylcholine receptors, nor upon the number of discrete receptor clusters. 5) Voluntary wheel-running exercise, beginning late in middle-age, prevented much of the age-associated loss of nerve terminal synaptophysin. In summary, a decline in the area of endplate acetylcholine receptor clusters at the onset of old age was followed by loss of nerve terminal synaptophysin from the endplate. Voluntary running exercise, begun late in middle age, substantially inhibited the loss of nerve terminal from aging motor endplates.
Collapse
Affiliation(s)
- Anson Cheng
- Physiology and Bosch Institute, University of Sydney, Sydney, New South Wales, Australia
| | - Marco Morsch
- Physiology and Bosch Institute, University of Sydney, Sydney, New South Wales, Australia
| | - Yui Murata
- Physiology and Bosch Institute, University of Sydney, Sydney, New South Wales, Australia
| | - Nazanin Ghazanfari
- Physiology and Bosch Institute, University of Sydney, Sydney, New South Wales, Australia
| | - Stephen W. Reddel
- Department of Molecular Medicine, Concord Hospital, Concord, New South Wales, Australia
| | - William D. Phillips
- Physiology and Bosch Institute, University of Sydney, Sydney, New South Wales, Australia
- * E-mail:
| |
Collapse
|
20
|
Kim JC, Kalantar-Zadeh K, Kopple JD. Frailty and protein-energy wasting in elderly patients with end stage kidney disease. J Am Soc Nephrol 2012; 24:337-51. [PMID: 23264684 DOI: 10.1681/asn.2012010047] [Citation(s) in RCA: 210] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Older people constitute an increasingly greater proportion of patients with advanced CKD, including those patients undergoing maintenance dialysis treatment. Frailty is a biologic syndrome of decreased reserve and resistance to stressors that results from cumulative declines across multiple physiologic systems and causes vulnerability to adverse outcomes. Frailty is common in elderly CKD patients, and it may be associated with protein-energy wasting (PEW), sarcopenia, dynapenia, and other complications of CKD. Causes of frailty with or without PEW in the elderly with CKD can be classified into three categories: causes primarily caused by aging per se, advanced CKD per se, or a combination of both conditions. Frailty and PEW in elderly CKD patients are associated with impaired physical performance, disability, poorer quality of life, and reduced survival. Prevention and treatment of these conditions in the elderly CKD patients often require a multifaceted approach. Here, we examine the causes and consequences of these conditions and examine the interplay between frailty and PEW in elderly CKD patients.
Collapse
Affiliation(s)
- Jun Chul Kim
- Division of Nephrology and Hypertension, Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, California 90502, USA
| | | | | |
Collapse
|
21
|
Weber H, Rauch A, Adamski S, Chakravarthy K, Kulkarni A, Dogdas B, Bendtsen C, Kath G, Alves SE, Wilkinson HA, Chiu CS. Automated rodent in situ muscle contraction assay and myofiber organization analysis in sarcopenia animal models. J Appl Physiol (1985) 2012; 112:2087-98. [PMID: 22461442 DOI: 10.1152/japplphysiol.00871.2011] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Age-related sarcopenia results in frailty and decreased mobility, which are associated with increased falls and long-term disability in the elderly. Given the global increase in lifespan, sarcopenia is a growing, unmet medical need. This report aims to systematically characterize muscle aging in preclinical models, which may facilitate the development of sarcopenia therapies. Naïve rats and mice were subjected to noninvasive micro X-ray computed tomography (micro-CT) imaging, terminal in situ muscle function characterizations, and ATPase-based myofiber analysis. We developed a Definiens (Parsippany, NJ)-based algorithm to automate micro-CT image analysis, which facilitates longitudinal in vivo muscle mass analysis. We report development and characterization of translational in situ skeletal muscle performance assay systems in rat and mouse. The systems incorporate a custom-designed animal assay stage, resulting in enhanced force measurement precision, and LabVIEW (National Instruments, Austin, TX)-based algorithms to support automated data acquisition and data analysis. We used ATPase-staining techniques for myofibers to characterize fiber subtypes and distribution. Major parameters contributing to muscle performance were identified using data mining and integration, enabled by Labmatrix (BioFortis, Columbia, MD). These technologies enabled the systemic and accurate monitoring of muscle aging from a large number of animals. The data indicated that longitudinal muscle cross-sectional area measurement effectively monitors change of muscle mass and function during aging. Furthermore, the data showed that muscle performance during aging is also modulated by myofiber remodeling factors, such as changes in myofiber distribution patterns and changes in fiber shape, which affect myofiber interaction. This in vivo muscle assay platform has been applied to support identification and validation of novel targets for the treatment of sarcopenia.
Collapse
Affiliation(s)
- H. Weber
- Musculo-Skeletal Biology Program Team, Merck Research Laboratories, West Point, Pennsylvania
| | - A. Rauch
- Bioelectronics, Merck Research Laboratories, West Point, Pennsylvania
| | - S. Adamski
- Musculo-Skeletal Biology Program Team, Merck Research Laboratories, West Point, Pennsylvania
| | - K. Chakravarthy
- Musculo-Skeletal Biology Program Team, Merck Research Laboratories, West Point, Pennsylvania
| | - A. Kulkarni
- Musculo-Skeletal Biology Program Team, Merck Research Laboratories, West Point, Pennsylvania
| | - B. Dogdas
- Informatics IT, Merck Research Laboratories, West Point, Pennsylvania
| | - C. Bendtsen
- Merck Research Laboratories, IRBM, Rome, Italy; and
| | - G. Kath
- Bioelectronics, Merck Research Laboratories, Rahway, New Jersey
| | - S. E. Alves
- Musculo-Skeletal Biology Program Team, Merck Research Laboratories, West Point, Pennsylvania
| | - H. A. Wilkinson
- Musculo-Skeletal Biology Program Team, Merck Research Laboratories, West Point, Pennsylvania
| | - C-S. Chiu
- Musculo-Skeletal Biology Program Team, Merck Research Laboratories, West Point, Pennsylvania
| |
Collapse
|
22
|
Abstract
Insulin-like growth factor 1 (IGF-1) is a pleiotropic polypeptide. Its expression is tightly regulated and it plays significant roles during early development, maturation, and adulthood. This article discusses the roles of IGF-1 in determination of body size, skeletal acquisition, muscle growth, carbohydrate metabolism, and longevity, as learned from mouse models.
Collapse
Affiliation(s)
- Shoshana Yakar
- Department of Basic Science and Craniofacial Biology, David B. Kriser Dental Center, New York University College of Dentistry, New York, NY 10010-4086, USA.
| | | |
Collapse
|
23
|
Delbono O. Expression and regulation of excitation-contraction coupling proteins in aging skeletal muscle. Curr Aging Sci 2011; 4:248-59. [PMID: 21529320 PMCID: PMC9634721 DOI: 10.2174/1874609811104030248] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2010] [Revised: 05/10/2010] [Accepted: 05/10/2010] [Indexed: 11/22/2022]
Abstract
Functional and structural decline of the neuromuscular system is a recognized cause of decreased strength, impaired performance of daily living activities, and loss of independence in the elderly. However, in mammals, including humans, age-related loss of strength is greater than loss of muscle mass, so the underlying mechanisms remain only partially understood. This review focuses on the mechanisms underlying impaired skeletal muscle function with aging, including external calcium-dependent skeletal muscle contraction; increased voltage-sensitive calcium channel Cav1.1 β1asubunit and junctional face protein JP-45 and decreased Cav1.1 (α1) expression, and the potential role of these and other recently discovered molecules of the muscle T-tubule/sarcoplasmic reticulum junction in excitation-contraction uncoupling. We also examined neural influences and trophic factors, particularly insulin-like growth factor-I (IGF-1). Better insight into the triad proteins' involvement in muscle ECC and nerve/muscle interactions and regulation will lead to more rational interventions to delay or prevent muscle weakness with aging. The focus of this review is on the proteins mediating excitation-contraction coupling (ECC) and their expression and regulation in humans and rodent models of skeletal muscle functional decline with aging. Age-dependent changes in proteins other than those related to ECC, muscle composition, clinical assessment and interventions, have been extensively reviewed recently [1-3].
Collapse
Affiliation(s)
- Osvaldo Delbono
- Department of Internal Medicine, Section on Gerontology and Geriatric Medicine, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA.
| |
Collapse
|
24
|
Larkin LM, Davis CS, Sims-Robinson C, Kostrominova TY, Van Remmen H, Richardson A, Feldman EL, Brooks SV. Skeletal muscle weakness due to deficiency of CuZn-superoxide dismutase is associated with loss of functional innervation. Am J Physiol Regul Integr Comp Physiol 2011; 301:R1400-7. [PMID: 21900648 DOI: 10.1152/ajpregu.00093.2011] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
An association between oxidative stress and muscle atrophy and weakness in vivo is supported by elevated oxidative damage and accelerated loss of muscle mass and force with aging in CuZn-superoxide dismutase-deficient (Sod1(-/-)) mice. The purpose was to determine the basis for low specific force (N/cm(2)) of gastrocnemius muscles in Sod1(-/-) mice and establish the extent to which structural and functional changes in muscles of Sod1(-/-) mice resemble those associated with normal aging. We tested the hypothesis that muscle weakness in Sod1(-/-) mice is due to functionally denervated fibers by comparing forces during nerve and direct muscle stimulation. No differences were observed for wild-type mice at any age in the forces generated in response to nerve and muscle stimulation. Nerve- and muscle-stimulated forces were also not different for 4-wk-old Sod1(-/-) mice, whereas, for 8- and 20-mo-old mice, forces during muscle stimulation were 16 and 30% greater, respectively, than those obtained using nerve stimulation. In addition to functional evidence of denervation with aging, fiber number was not different for Sod1(-/-) and wild-type mice at 4 wk, but 50% lower for Sod1(-/-) mice by 20 mo, and denervated motor end plates were prevalent in Sod1(-/-) mice at both 8 and 20 mo and in WT mice by 28 mo. The data suggest ongoing denervation in muscles of Sod1(-/-) mice that results in fiber loss and muscle atrophy. Moreover, the findings support using Sod1(-/-) mice to explore mechanistic links between oxidative stress and the progression of deficits in muscle structure and function.
Collapse
Affiliation(s)
- Lisa M Larkin
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan 48109-2200, USA
| | | | | | | | | | | | | | | |
Collapse
|
25
|
Sood S, Hanson ED, Delmonico MJ, Kostek MC, Hand BD, Roth SM, Hurley BF. Does insulin-like growth factor 1 genotype influence muscle power response to strength training in older men and women? Eur J Appl Physiol 2011; 112:743-53. [DOI: 10.1007/s00421-011-2028-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2011] [Accepted: 05/25/2011] [Indexed: 12/25/2022]
|
26
|
Motor End Plate Innervation Loss in Diabetes and the Role of Insulin. J Neuropathol Exp Neurol 2011; 70:323-39. [DOI: 10.1097/nen.0b013e318215669a] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
|
27
|
Bosch-Marcé M, Wee CD, Martinez TL, Lipkes CE, Choe DW, Kong L, Van Meerbeke JP, Musarò A, Sumner CJ. Increased IGF-1 in muscle modulates the phenotype of severe SMA mice. Hum Mol Genet 2011; 20:1844-53. [PMID: 21325354 DOI: 10.1093/hmg/ddr067] [Citation(s) in RCA: 80] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Spinal muscular atrophy (SMA) is an inherited motor neuron disease caused by the mutation of the survival motor neuron 1 (SMN1) gene and deficiency of the SMN protein. Severe SMA mice have abnormal motor function and small, immature myofibers early in development suggesting that SMN protein deficiency results in retarded muscle growth. Insulin-like growth factor 1 (IGF-1) stimulates myoblast proliferation, induces myogenic differentiation and generates myocyte hypertrophy in vitro and in vivo. We hypothesized that increased expression of IGF-1 specifically in skeletal muscle would attenuate disease features of SMAΔ7 mice. SMAΔ7 mice overexpressing a local isoform of IGF-1 (mIGF-1) in muscle showed enlarged myofibers and a 40% increase in median survival compared with mIGF-1-negative SMA littermates (median survival = 14 versus 10 days, respectively, log-rank P = 0.025). Surprisingly, this was not associated with a significant improvement in motor behavior. Treatment of both mIGF-1(NEG) and mIGF-1(POS) SMA mice with the histone deacetylase inhibitor, trichostatin A (TSA), resulted in a further extension of survival and improved motor behavior, but the combination of mIGF-1 and TSA treatment was not synergistic. These results show that increased mIGF-1 expression restricted to muscle can modulate the phenotype of SMA mice indicating that therapeutics targeted to muscle alone should not be discounted as potential disease-modifying therapies in SMA. IGF-1 may warrant further investigation in mild SMA animal models and perhaps SMA patients.
Collapse
Affiliation(s)
- Marta Bosch-Marcé
- Department of Neurology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Velloso CP, Harridge SDR. Insulin-like growth factor-I E peptides: implications for aging skeletal muscle. Scand J Med Sci Sports 2010; 20:20-7. [PMID: 19883387 DOI: 10.1111/j.1600-0838.2009.00997.x] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
In skeletal muscle there is good evidence to suggest that locally produced insulin-like growth factor-1 (IGF-I), rather than circulating IGF-I, is important in regard to muscle mass maintenance, repair and hypertrophy. This "mature" IGF-I comprises exons 3 and 4 of the IGF-I gene, but during processing the full length gene (which contains six exons) is subject to a process of alternative splicing. As a result smaller peptides (E peptides) are believed to be cleaved from the mature IGF-I peptide during processing of the prohormone and the likelihood is that they have different biological roles. In human skeletal muscle three transcripts encoding for these splice variants (IGF-IEa, IGF-IEb and IGF-IEc, also known as MGF) can be identified. When studied at the mRNA level these three transcripts are known to be upregulated in the muscles of elderly people following high resistance exercise, albeit with different time courses. However, compared with mature IGF-I relatively little is known about the mechanism of action of the different E peptides.
Collapse
Affiliation(s)
- Cristina P Velloso
- Division of Applied Biomedical Research, King's College London, London, UK
| | | |
Collapse
|
29
|
Lang T, Streeper T, Cawthon P, Baldwin K, Taaffe DR, Harris TB. Sarcopenia: etiology, clinical consequences, intervention, and assessment. Osteoporos Int 2010; 21:543-59. [PMID: 19779761 PMCID: PMC2832869 DOI: 10.1007/s00198-009-1059-y] [Citation(s) in RCA: 501] [Impact Index Per Article: 33.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2009] [Accepted: 08/18/2009] [Indexed: 02/07/2023]
Abstract
The aging process is associated with loss of muscle mass and strength and decline in physical functioning. The term sarcopenia is primarily defined as low level of muscle mass resulting from age-related muscle loss, but its definition is often broadened to include the underlying cellular processes involved in skeletal muscle loss as well as their clinical manifestations. The underlying cellular changes involve weakening of factors promoting muscle anabolism and increased expression of inflammatory factors and other agents which contribute to skeletal muscle catabolism. At the cellular level, these molecular processes are manifested in a loss of muscle fiber cross-sectional area, loss of innervation, and adaptive changes in the proportions of slow and fast motor units in muscle tissue. Ultimately, these alterations translate to bulk changes in muscle mass, strength, and function which lead to reduced physical performance, disability, increased risk of fall-related injury, and, often, frailty. In this review, we summarize current understanding of the mechanisms underlying sarcopenia and age-related changes in muscle tissue morphology and function. We also discuss the resulting long-term outcomes in terms of loss of function, which causes increased risk of musculoskeletal injuries and other morbidities, leading to frailty and loss of independence.
Collapse
Affiliation(s)
- T Lang
- Department of Radiology and Biomedical Imaging, University of California, UCSF, San Francisco, CA 94143-0946, USA.
| | | | | | | | | | | |
Collapse
|
30
|
Kiryakova S, Söhnchen J, Grosheva M, Schuetz U, Marinova T, Dzhupanova R, Sinis N, Hübbers CU, Skouras E, Ankerne J, Fries JWU, Irintchev A, Dunlop SA, Angelov DN. Recovery of whisking function promoted by manual stimulation of the vibrissal muscles after facial nerve injury requires insulin-like growth factor 1 (IGF-1). Exp Neurol 2010; 222:226-34. [PMID: 20067789 DOI: 10.1016/j.expneurol.2009.12.031] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2009] [Accepted: 12/30/2009] [Indexed: 01/04/2023]
Abstract
Recently, we showed that manual stimulation (MS) of denervated vibrissal muscles enhanced functional recovery following facial nerve cut and suture (FFA) by reducing poly-innervation at the neuro-muscular junctions (NMJ). Although the cellular correlates of poly-innervation are established, with terminal Schwann cells (TSC) processes attracting axon sprouts to "bridge" adjacent NMJ, molecular correlates are poorly understood. Since quantitative RT-PCR revealed a rapid increase of IGF-1 mRNA in denervated muscles, we examined the effect of daily MS for 2 months after FFA in IGF-1(+/-) heterozygous mice; controls were wild-type (WT) littermates including intact animals. We quantified vibrissal motor performance and the percentage of NMJ bridged by S100-positive TSC. There were no differences between intact WT and IGF-1(+/-) mice for vibrissal whisking amplitude (48 degrees and 49 degrees ) or the percentage of bridged NMJ (0%). After FFA and handling alone (i.e. no MS) in WT animals, vibrissal whisking amplitude was reduced (60% lower than intact) and the percentage of bridged NMJ increased (42% more than intact). MS improved both the amplitude of vibrissal whisking (not significantly different from intact) and the percentage of bridged NMJ (12% more than intact). After FFA and handling in IGF-1(+/-) mice, the pattern was similar (whisking amplitude 57% lower than intact; proportion of bridged NMJ 42% more than intact). However, MS did not improve outcome (whisking amplitude 47% lower than intact; proportion of bridged NMJ 40% more than intact). We conclude that IGF-I is required to mediate the effects of MS on target muscle reinnervation and recovery of whisking function.
Collapse
Affiliation(s)
- S Kiryakova
- Department of Anatomy I, University of Cologne, D-50924 Cologne, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Saini A, Faulkner S, Al-Shanti N, Stewart C. Powerful signals for weak muscles. Ageing Res Rev 2009; 8:251-67. [PMID: 19716529 DOI: 10.1016/j.arr.2009.02.001] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2009] [Accepted: 02/10/2009] [Indexed: 12/19/2022]
Abstract
The aim of the present review is to summarise, evaluate and critique the different mechanisms involved in anabolic growth of skeletal muscle and the catabolic processes involved in cancer cachexia and sarcopenia of ageing. This is highly relevant, since they represent targets for future promising clinical investigations. Sarcopenia is an inevitable process associated with a gradual reduction in muscle mass and strength, associated with a reduction in motor unit number and atrophy of muscle fibres, especially the fast type IIa fibres. The loss of muscle mass with ageing is clinically important because it leads to diminished functional ability and associated complications. Cachexia is widely recognised as severe and rapid wasting accompanying disease states such as cancer or immunodeficiency disease. One of the main characteristics of cancer cachexia is asthenia or lack of strength, which is directly related to the muscle loss. Indeed, apart from the speed of loss, muscle wasting during cancer and ageing share many common metabolic pathways and mediators. In healthy young individuals, muscles maintain their mass and function because of a balance between protein synthesis and protein degradation associated with rates of anabolic and catabolic processes, respectively. Muscles grow (hypertrophy) when protein synthesis exceeds protein degradation. Conversely, muscles shrink (atrophy) when protein degradation dominates. These processes are not occurring independently of each other, but are finely coordinated by a web of intricate signalling networks. Such signalling networks are in charge of executing environmental and cellular cues that ultimately determine whether muscle proteins are synthesised or degraded. Increasing our understanding for the pathways involved in hypertrophy and atrophy and particularly the interaction of these pathways is essential in designing therapeutic strategies for both prevention and treatment of muscle wasting conditions with age and with disease.
Collapse
Affiliation(s)
- Amarjit Saini
- Institute for Biomedical Research into Human Movement and Health, Manchester Metropolitan University, Manchester, United Kingdom.
| | | | | | | |
Collapse
|
32
|
Augustin H, Partridge L. Invertebrate models of age-related muscle degeneration. Biochim Biophys Acta Gen Subj 2009; 1790:1084-94. [PMID: 19563864 DOI: 10.1016/j.bbagen.2009.06.011] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2009] [Revised: 06/12/2009] [Accepted: 06/20/2009] [Indexed: 12/26/2022]
Abstract
Functional and structural deterioration of muscles is an inevitable consequence of ageing in a wide variety of animal species. What underlies these changes is a complex network of interactions between the muscle-intrinsic and muscle-extrinsic factors, making it very difficult to distinguish between the cause and the consequence. Many of the genes, structures, and processes implicated in mammalian skeletal muscle ageing are preserved in invertebrate species Drosophila melanogaster and Caenorhabditis elegans. The absence in these organisms of mechanisms that promote muscle regeneration, and substantially different hormonal environment, warrant caution when extrapolating experimental data from studies conducted in invertebrates to mammalian species. The simplicity and accessibility of these models, however, offer ample opportunities for studying age-related myopathologies as well as investigating drugs and therapies to alleviate them.
Collapse
Affiliation(s)
- Hrvoje Augustin
- Institute of Healthy Ageing and GEE, University College London, London WC1E 6BT, UK
| | | |
Collapse
|
33
|
Schuenke MD, Kopchick JJ, Hikida RS, Kraemer WJ, Staron RS. Effects of growth hormone overexpression vs. growth hormone receptor gene disruption on mouse hindlimb muscle fiber type composition. Growth Horm IGF Res 2008; 18:479-486. [PMID: 18499495 DOI: 10.1016/j.ghir.2008.04.003] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2008] [Revised: 04/07/2008] [Accepted: 04/09/2008] [Indexed: 02/06/2023]
Abstract
OBJECTIVE The present study characterizes the fiber type composition of selected hindlimb muscles from two transgenic mouse lines specifically engineered to alter the amounts of circulating growth hormone (GH) and insulin-like growth factor-1 (IFG-1). DESIGN The triceps surae muscle group (soleus m., gastrocnemius m., and plantaris m.) was harvested en masse from mice that were: (1) giant due to the expression of a bovine GH transgene (bGH), (2) dwarf due to the disruption of the GH receptor/binding protein gene (GHR-/-), and (3) normal-sized controls [non-transgenic (NT)]. Histochemical and immunohistochemical methods were utilized on serial cross sections to delineate eight fiber types (I, IC, IIC, IIA, IIAD, IID, IIDB, and IIB). Cross-sectional areas were subsequently determined on approximately 50 fibers/type. RESULTS Compared to NT littermates, muscles from bGH mice demonstrated a significant (p<0.05) fast-to-slow shift in fiber phenotype, as well as significantly larger fibers for most types. In contrast, significantly smaller fibers were found for all fiber types in the GHR-/- mice, with no significant differences in fiber type percentages compared to NT. Regardless of mouse genotype, the hierarchy of fiber size was maintained in each muscle with type I the largest in the soleus m. and type IIB the largest in the predominantly fast muscles (plantaris, superficial and deep gastrocnemius muscles). CONCLUSION In conclusion, the genetic manipulation of GH expression (bGH) and its receptor binding (GHR-/-) had profound and divergent effects on muscle phenotype. It is hoped that continued research in this area will help elucidate the direct (independent of IGF-1) vs. indirect (via IGF-1 mediating mechanisms) effects of GH.
Collapse
Affiliation(s)
- Mark D Schuenke
- Department of Anatomy, College of Osteopathic Medicine, University of New England, Biddeford, ME 04005, USA.
| | | | | | | | | |
Collapse
|
34
|
Abstract
IGF-I and -II are potent neuronal mitogens and survival factors. The actions of IGF-I and -II are mediated via the type I IGF receptor (IGF-IR) and IGF binding proteins regulate the bioavailability of the IGFs. Cell viability correlates with IGF-IR expression and intact IGF-I/IGF-IR signaling pathways, including activation of MAPK/phosphatidylinositol-3 kinase. The expression of IGF-I and -II, IGF-IR, and IGF binding proteins are developmentally regulated in the central and peripheral nervous system. IGF-I therapy demonstrates mixed therapeutic results in the treatment of peripheral nerve injury, neuropathy, and motor neuron diseases such as amyotrophic lateral sclerosis. In this review we discuss the role of IGFs during peripheral nervous system development and the IGF signaling system as the potential therapeutic target for the treatment of nerve injury and motor neuron diseases.
Collapse
Affiliation(s)
- Kelli A Sullivan
- Department of Neurology, University of Michigan, Ann Arbor, Michigan 48109-2200, USA
| | | | | |
Collapse
|
35
|
Zacchigna S, Lambrechts D, Carmeliet P. Neurovascular signalling defects in neurodegeneration. Nat Rev Neurosci 2008; 9:169-81. [DOI: 10.1038/nrn2336] [Citation(s) in RCA: 275] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
36
|
Cellular and molecular mechanisms underlying age-related skeletal muscle wasting and weakness. Biogerontology 2008; 9:213-28. [PMID: 18299960 DOI: 10.1007/s10522-008-9131-0] [Citation(s) in RCA: 283] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2007] [Accepted: 02/06/2008] [Indexed: 01/02/2023]
Abstract
Some of the most serious consequences of ageing are its effects on skeletal muscle. The term 'sarcopenia' describes the slow but progressive loss of muscle mass with advancing age and is characterised by a deterioration of muscle quantity and quality leading to a gradual slowing of movement and a decline in strength. The loss of muscle mass and strength is thought to be attributed to the progressive atrophy and loss of individual muscle fibres associated with the loss of motor units, and a concomitant reduction in muscle 'quality' due to the infiltration of fat and other non-contractile material. These age-related changes in skeletal muscle can be largely attributed to the complex interaction of factors affecting neuromuscular transmission, muscle architecture, fibre composition, excitation-contraction coupling, and metabolism. Given the magnitude of the growing public health problems associated with sarcopenia, there is considerable interest in the development and evaluation of therapeutic strategies to attenuate, prevent, or ultimately reverse age-related muscle wasting and weakness. The aim is to review our current understanding of some of the cellular and molecular mechanisms responsible for age-related changes in skeletal muscle.
Collapse
|
37
|
Matsuoka Y, Inoue A. Controlled differentiation of myoblast cells into fast and slow muscle fibers. Cell Tissue Res 2008; 332:123-32. [DOI: 10.1007/s00441-008-0582-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2007] [Accepted: 01/14/2008] [Indexed: 10/22/2022]
|
38
|
Ai J, Gozal D, Li L, Wead WB, Chapleau MW, Wurster R, Yang B, Li H, Liu R, Cheng Z. Degeneration of vagal efferent axons and terminals in cardiac ganglia of aged rats. J Comp Neurol 2007; 504:74-88. [PMID: 17614301 DOI: 10.1002/cne.21431] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Baroreflex control of the heart rate is significantly reduced during aging. However, neural mechanisms that underlie such a functional reduction are not fully understood. We injected the tracer DiI into the left nucleus ambiguus (NA), then used confocal microscopy and a Neurolucida Digitization System to examine qualitatively and quantitatively vagal efferent projections to cardiac ganglia of young adult (5-6 months) and aged (24-25 months) rats (Sprague Dawley). Fluoro-Gold was injected intraperitoneally to counterstain cardiac ganglionic principal neurons (PNs). In aged, as in young rats, NA axons projected to all cardiac ganglia and formed numerous basket endings around PNs in the hearts. However, significant structural changes were found in aged rats compared with young rats. Vagal efferent axons contained abnormally swollen axonal segments and exhibited reduced or even absent synaptic-like terminals around PNs, such that the numbers of vagal fibers and basket endings around PNs were substantially reduced (P < 0.01). Furthermore, synaptic-like varicose contacts of vagal cardiac axons with PNs were significantly reduced by approximately 50% (P < 0.01). These findings suggest that vagal efferents continue to maintain homeostatic control over the heart during aging. However, the marked morphological reorganization of vagal efferent axons and terminals in cardiac ganglia may represent the structural substrate for reduced vagal control of the heart rate and attenuated baroreflex function during aging.
Collapse
Affiliation(s)
- Jing Ai
- Biomolecular Science Center, Burnett College of Biomedical Sciences, University of Central Florida, Orlando, Florida 32816, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Lynch GS, Schertzer JD, Ryall JG. Therapeutic approaches for muscle wasting disorders. Pharmacol Ther 2007; 113:461-87. [PMID: 17258813 DOI: 10.1016/j.pharmthera.2006.11.004] [Citation(s) in RCA: 100] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2006] [Revised: 11/10/2006] [Accepted: 11/10/2006] [Indexed: 12/12/2022]
Abstract
Muscle wasting and weakness are common in many disease states and conditions including aging, cancer cachexia, sepsis, denervation, disuse, inactivity, burns, HIV-acquired immunodeficiency syndrome (AIDS), chronic kidney or heart failure, unloading/microgravity, and muscular dystrophies. Although the maintenance of muscle mass is generally regarded as a simple balance between protein synthesis and protein degradation, these mechanisms are not strictly independent, but in fact they are coordinated by a number of different and sometimes complementary signaling pathways. Clearer details are now emerging about these different molecular pathways and the extent to which these pathways contribute to the etiology of various muscle wasting disorders. Therapeutic strategies for attenuating muscle wasting and improving muscle function vary in efficacy. Exercise and nutritional interventions have merit for slowing the rate of muscle atrophy in some muscle wasting conditions, but in most cases they cannot halt or reverse the wasting process. Hormonal and/or other drug strategies that can target key steps in the molecular pathways that regulate protein synthesis and protein degradation are needed. This review describes the signaling pathways that maintain muscle mass and provides an overview of some of the major conditions where muscle wasting and weakness are indicated. The review provides details on some therapeutic strategies that could potentially attenuate muscle atrophy, promote muscle growth, and ultimately improve muscle function. The emphasis is on therapies that can increase muscle mass and improve functional outcomes that will ultimately lead to improvement in the quality of life for affected patients.
Collapse
Affiliation(s)
- Gordon S Lynch
- Basic and Clinical Myology Laboratory, Department of Physiology, The University of Melbourne, Victoria 3010, Australia.
| | | | | |
Collapse
|
40
|
Moreno RJ, Messi ML, Zheng Z, Wang ZM, Ye P, D'Ercole JA, Delbono O. Role of sustained overexpression of central nervous system IGF-I in the age-dependent decline of mouse excitation-contraction coupling. J Membr Biol 2007; 212:147-61. [PMID: 17334835 DOI: 10.1007/s00232-006-0044-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2006] [Revised: 09/18/2006] [Indexed: 12/20/2022]
Abstract
We investigated the effects of exclusive and sustained transgenic overexpression of insulin-like growth factor (IGF)-I in the central nervous system (CNS) on the age-dependent decline in muscle strength, excitation-contraction coupling, muscle innervation and neuromuscular junction postterminal architecture. We found that (1) transgenic IGF-I overexpression in the CNS does not modify the decline in extensor digitorum longus (EDL) and soleus muscle weight with aging and (2) strength significantly decreases in transgenic (Tg) compared to wild-type mice. The latter finding is consistent with (3) the decreased absolute and specific force measured in the EDL muscle in vitro and (4) the decreased charge movement and peak intracellular Ca(2+) mobilization in individual muscle fibers from old IGF-I Tg mice compared to young wild-type mice, which also is associated with (5) decreased dihydropyridine receptor alpha(1)-subunit expression in old compared to young IGF-I Tg mice. (6) Tg IGF-I prevents a change in muscle fiber type that is associated with (7) improved muscle innervation and postterminal neuromuscular structure. (8) IGF-I is expressed extensively across the spinal cord gray matter and the lateral motor column. Our results raise questions about the timing and cell location of CNS IGF-I overexpression necessary to prevent or to ameliorate age-dependent alterations in the structure and function of skeletal muscle.
Collapse
Affiliation(s)
- Ramón Jiménez Moreno
- Department of Physiology and Pharmacology, Wake Forest University School of Medicine, Winston-Salem, NC, 27157, USA
| | | | | | | | | | | | | |
Collapse
|
41
|
Ramji N, Toth C, Kennedy J, Zochodne DW. Does diabetes mellitus target motor neurons? Neurobiol Dis 2006; 26:301-11. [PMID: 17337195 DOI: 10.1016/j.nbd.2006.11.016] [Citation(s) in RCA: 75] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2006] [Revised: 11/02/2006] [Accepted: 11/04/2006] [Indexed: 11/18/2022] Open
Abstract
A pattern of peripheral neurodegeneration occurs in chronic diabetes mellitus in which an early, but selective retraction of distal axons may occur prior to any irretrievable neuronal loss. Clinical observations suggest that sensory systems undergo damage before those of motor neurons. In this work, we examined the fate of the spinal motor neuron in a long-term chronic model of experimental (streptozotocin-induced) diabetes already known to be associated with substantial loss of sensory neurons. The integrity, physiological function, and critical forms of protein expression of the full motor neuron tree was examined in mice exposed to 8 months of diabetes. Motor neurons developed progressive features of distal loss of axonal terminals but without perikaryal dropout, indicating distal axon retraction. While numbers and caliber of motor neuron perikarya and their nerve trunk axons were preserved, axons developed conduction velocity slowing, loss of motor units and neuromuscular junctions, and compensatory single motor unit action potential enlargement. Four critical proteins directly linked to diabetic complications were altered in motor neurons of diabetic mice: an elevated perikaryal expression of RAGE and PARP, molecules associated with cellular stress, along with concurrent rises in HSP-27 and pAKT, molecules alternatively identified with neuroprotective survival. Moreover, Akt mRNA was increased in diabetic lumbar spinal cords. Overall these findings indicate that although motor neurons are resistant to irretrievable dropout, they are targeted nonetheless by diabetes and gradually withdraw their terminals from distal innervation.
Collapse
Affiliation(s)
- Noor Ramji
- University of Calgary, Department of Clinical Neurosciences, Room 168, 3330 Hospital Drive, N.W., Calgary, Alberta, Canada T2N 4N1
| | | | | | | |
Collapse
|
42
|
Payne AM, Messi ML, Zheng Z, Delbono O. Motor neuron targeting of IGF-1 attenuates age-related external Ca2+-dependent skeletal muscle contraction in senescent mice. Exp Gerontol 2006; 42:309-19. [PMID: 17174053 PMCID: PMC2063746 DOI: 10.1016/j.exger.2006.11.003] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2006] [Revised: 10/25/2006] [Accepted: 11/03/2006] [Indexed: 11/25/2022]
Abstract
A population of fast muscle fibers from aging mice is dependent on external Ca(2+) to maintain tetanic force during repeated contractions. We hypothesized that age-related denervation in muscle fibers plays a role in initiating this contractile deficit, and that prevention of denervation by IGF-1 overexpression would prevent external Ca(2+)-dependent contraction in aging mice. IGF-1 overexpression in skeletal muscle prevents age-related denervation, and prevented external Ca(2+)-dependent contraction in this work. To determine if the effects of IGF-1 overexpression are on muscle or nerve, aging mice were injected with a tetanus toxin fragment-C (TTC) fusion protein that targets IGF-1 to spinal cord motor neurons. This treatment prevented external Ca(2+)-dependent contraction. We also show evidence that injections of the IGF-1-TTC fusion protein prevent age-related alterations to the nerve terminals at the neuromuscular junctions. We conclude that the slow age-related denervation of fast muscle fibers underlies dependence on external Ca(2+) to maintain tetanic force in a population of muscle fibers from senescent mice.
Collapse
Affiliation(s)
- Anthony M. Payne
- Department of Physiology and Pharmacology, Wake Forest University School of Medicine, Winston-Salem, NC 27157
| | - María Laura Messi
- Department of Physiology and Pharmacology, Wake Forest University School of Medicine, Winston-Salem, NC 27157
| | - Zhenlin Zheng
- Department of Physiology and Pharmacology, Wake Forest University School of Medicine, Winston-Salem, NC 27157
| | - Osvaldo Delbono
- Department of Physiology and Pharmacology, Wake Forest University School of Medicine, Winston-Salem, NC 27157
- Department of Internal Medicine, Section on Gerontology, Wake Forest University School of Medicine, Winston-Salem, NC 27157
- Neuroscience Program, Wake Forest University School of Medicine, Winston-Salem, NC 27157
- * Corresponding Author: Osvaldo Delbono, Department of Physiology and Pharmacology, Wake Forest University School of Medicine, 1 Medical Center Boulevard, Winston-Salem, NC 27157, USA, Phone: (336) 716-9802, Fax: (336) 716-2273,
| |
Collapse
|
43
|
Payne AM, Zheng Z, Messi ML, Milligan CE, González E, Delbono O. Motor neurone targeting of IGF-1 prevents specific force decline in ageing mouse muscle. J Physiol 2005; 570:283-94. [PMID: 16293644 PMCID: PMC1464304 DOI: 10.1113/jphysiol.2005.100032] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
IGF-1 is a potent growth factor for both motor neurones and skeletal muscle. Muscle IGF-1 is known to provide target-derived trophic effects on motor neurones. Therefore, IGF-1 overexpression in muscle is effective in delaying or preventing deleterious effects of ageing in both tissues. Since age-related decline in muscle function stems partly from motor neurone loss, a tetanus toxin fragment-C (TTC) fusion protein was created to target IGF-1 to motor neurones. IGF-1-TTC retains IGF-1 activity as indicated by [(3)H]thymidine incorporation into L6 myoblasts. Spinal cord motor neurones effectively bound and internalized the IGF-1-TTC in vitro. Similarly, IGF-1-TTC injected into skeletal muscles was taken up and retrogradely transported to the spinal cord in vivo, a process prevented by denervation of injected muscles. Three monthly IGF-1-TTC injections into muscles of ageing mice did not increase muscle weight or muscle fibre size, but significantly increased single fibre specific force over aged controls injected with saline, IGF-1, or TTC. None of the injections changed muscle fibre type composition, but neuromuscular junction post-terminals were larger and more complex in muscle fibres injected with IGF-1-TTC, compared to the other groups, suggesting preservation of muscle fibre innervation. This work demonstrates that induced overexpression of IGF-1 in spinal cord motor neurones of ageing mice prevents muscle fibre specific force decline, a hallmark of ageing skeletal muscle.
Collapse
Affiliation(s)
- Anthony M Payne
- Department of Physiology and Pharmacology, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA
| | | | | | | | | | | |
Collapse
|
44
|
Kostek MC, Delmonico MJ, Reichel JB, Roth SM, Douglass L, Ferrell RE, Hurley BF. Muscle strength response to strength training is influenced by insulin-like growth factor 1 genotype in older adults. J Appl Physiol (1985) 2005; 98:2147-54. [PMID: 15894537 DOI: 10.1152/japplphysiol.00817.2004] [Citation(s) in RCA: 72] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Strength training (ST) is considered an intervention of choice for the prevention and treatment of sarcopenia. Reports in the literature have suggested that the insulin-like growth factor I protein (IGF-I) plays a major role in ST-induced skeletal muscle hypertrophy and strength improvements. A microsatellite repeat in the promoter region of the IGF1 gene has been associated with IGF-I blood levels and phenotypes related to IGF-I in adult men and women. To examine the influence of this polymorphism on muscle hypertrophic and strength responses to ST, we studied 67 Caucasian men and women before and after a 10-wk single-leg knee-extension ST program. One repetition maximum strength, muscle volume via computed tomography, and muscle quality were assessed at baseline and after 10 wk of training. The IGF1 repeat promoter polymorphism and three single-nucleotide polymorphisms were genotyped. For the promoter polymorphism, subjects were grouped as homozygous for the 192 allele, heterozygous, or noncarriers of the 192 allele. After 10 wk of training, 1-repetition maximum, muscle volume, and muscle quality increased significantly for all groups combined ( P < 0.001). However, carriers of the 192 allele gained significantly more strength with ST than noncarriers of the 192 allele ( P = 0.02). There was also a nonsignificant trend for a greater increase in muscle volume in 192 carriers than noncarriers ( P = 0.08). No significant associations were observed for the other polymorphisms studied. Thus these data suggest that the IGF1 promoter polymorphism may influence the strength response to ST. Larger sample sizes should be used in future studies to verify these results.
Collapse
Affiliation(s)
- Matthew C Kostek
- Department of Kinesiology, University of Maryland, College Park, MD 20742, USA
| | | | | | | | | | | | | |
Collapse
|
45
|
Dion P, Shanmugam V, Gaspar C, Messaed C, Meijer I, Toulouse A, Laganiere J, Roussel J, Rochefort D, Laganiere S, Allen C, Karpati G, Bouchard JP, Brais B, Rouleau GA. Transgenic expression of an expanded (GCG)13 repeat PABPN1 leads to weakness and coordination defects in mice. Neurobiol Dis 2005; 18:528-36. [PMID: 15755680 DOI: 10.1016/j.nbd.2004.09.021] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2004] [Revised: 07/08/2004] [Accepted: 09/29/2004] [Indexed: 11/12/2022] Open
Abstract
Oculopharyngeal muscular dystrophy (OPMD) is a late-onset disorder caused by a (GCG)n trinucleotide repeat expansion in the poly(A) binding protein nuclear-1 (PABPN1) gene, which in turn leads to an expanded polyalanine tract in the protein. We generated transgenic mice expressing either the wild type or the expanded form of human PABPN1, and transgenic animals with the expanded form showed clear signs of abnormal limb clasping, muscle weakness, coordination deficits, and peripheral nerves alterations. Analysis of mitotic and postmitotic tissues in those transgenic animals revealed ubiquitinated PABPN1-positive intranuclear inclusions (INIs) in neuronal cells. This latter observation led us to test and confirm the presence of similar INIs in postmortem brain sections from an OPMD patient. Our results indicate that expanded PABPN1, presumably via the toxic effects of its polyalanine tract, can lead to inclusion formation and neurodegeneration in both the mouse and the human.
Collapse
Affiliation(s)
- Patrick Dion
- McGill University Health Center Research Institute and the Centre for Research in Neurosciences, McGill University, 1650 Cedar Avenue (L7-224), Montréal, Québec, Canada, H3G 1A4
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Lynch GS. Update on therapies for sarcopenia: novel approaches for age-related muscle wasting and weakness. Expert Opin Ther Pat 2005. [DOI: 10.1517/13543776.14.9.1329] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
47
|
Abstract
Sarcopenia is the term widely used to describe the progressive loss of muscle mass with advancing age. Even before significant muscle wasting becomes apparent, ageing is associated with a slowing of movement and a gradual decline in muscle strength, factors that increase the risk of injury from sudden falls and the reliance of the frail elderly on assistance in accomplishing even basic tasks of independent living. Sarcopenia is recognised as one of the major public health problems now facing industrialised nations, and its effects are expected to place increasing demands on public healthcare systems worldwide. Although the effects of ageing on skeletal muscle are unlikely to be halted or reversed, the underlying mechanisms responsible for these deleterious changes present numerous targets for drug discovery with potential opportunities to attenuate muscle wasting, improve muscle function, and preserve functional independence. Very few drugs have been developed with sarcopenia specifically in mind. However, because many of the effects of ageing on skeletal muscle resemble those indicated in many neuromuscular disorders, drugs that target neurodegenerative diseases may also have important relevance for treating age-related muscle wasting and weakness. This review describes a selection of the emerging drugs that have been developed during the period 1997 - 2004, relevant to sarcopenia.
Collapse
Affiliation(s)
- Gordon S Lynch
- The University of Melbourne, Department of Physiology and Centre for Neuroscience, Victoria, 3010, Australia.
| |
Collapse
|
48
|
Monti B, Contestabile A. Selective alteration of DNA fragmentation and caspase activity in the spinal cord of aged rats and effect of dietary restriction. Brain Res 2003; 992:137-41. [PMID: 14604782 DOI: 10.1016/s0006-8993(03)03456-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Age-related alterations in CNS may preferentially affect spinal cord. We report here that in spinal cord of aged (30-month-old) rats, a significant increase of DNA fragmentation accompanied by an increased activity of caspase-1, but not of caspase-3, can be measured. The increase in caspase-1 activation, but not the increased level of DNA fragmentation, was substantially reverted by long-lasting dietary restriction.
Collapse
Affiliation(s)
- Barbara Monti
- Department of Biology, University of Bologna, Via Selmi 3, 40126, Bologna, Italy
| | | |
Collapse
|