1
|
Majchrzak K, Hentschel E, Hönzke K, Geithe C, von Maltzahn J. We need to talk-how muscle stem cells communicate. Front Cell Dev Biol 2024; 12:1378548. [PMID: 39050890 PMCID: PMC11266305 DOI: 10.3389/fcell.2024.1378548] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Accepted: 06/18/2024] [Indexed: 07/27/2024] Open
Abstract
Skeletal muscle is one of the tissues with the highest ability to regenerate, a finely controlled process which is critically depending on muscle stem cells. Muscle stem cell functionality depends on intrinsic signaling pathways and interaction with their immediate niche. Upon injury quiescent muscle stem cells get activated, proliferate and fuse to form new myofibers, a process involving the interaction of multiple cell types in regenerating skeletal muscle. Receptors in muscle stem cells receive the respective signals through direct cell-cell interaction, signaling via secreted factors or cell-matrix interactions thereby regulating responses of muscle stem cells to external stimuli. Here, we discuss how muscle stem cells interact with their immediate niche focusing on how this controls their quiescence, activation and self-renewal and how these processes are altered in age and disease.
Collapse
Affiliation(s)
- Karolina Majchrzak
- Faculty of Health Sciences Brandenburg, Brandenburg University of Technology Cottbus–Senftenberg, Senftenberg, Germany
| | - Erik Hentschel
- Faculty of Health Sciences Brandenburg, Brandenburg University of Technology Cottbus–Senftenberg, Senftenberg, Germany
| | - Katja Hönzke
- Faculty of Health Sciences Brandenburg, Brandenburg University of Technology Cottbus–Senftenberg, Senftenberg, Germany
- Department of Infectious Diseases and Respiratory Medicine, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt Universität zu Berlin, Berlin, Germany
| | - Christiane Geithe
- Faculty of Health Sciences Brandenburg, Brandenburg University of Technology Cottbus–Senftenberg, Senftenberg, Germany
| | - Julia von Maltzahn
- Faculty of Health Sciences Brandenburg, Brandenburg University of Technology Cottbus–Senftenberg, Senftenberg, Germany
- Leibniz Institute on Aging, Fritz Lipmann Institute, Jena, Germany
- Faculty for Environment and Natural Sciences, Brandenburg University of Technology Cottbus—Senftenberg, Senftenberg, Germany
| |
Collapse
|
2
|
Henze H, Hüttner SS, Koch P, Schüler SC, Groth M, von Eyss B, von Maltzahn J. Denervation alters the secretome of myofibers and thereby affects muscle stem cell lineage progression and functionality. NPJ Regen Med 2024; 9:10. [PMID: 38424446 PMCID: PMC10904387 DOI: 10.1038/s41536-024-00353-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Accepted: 02/14/2024] [Indexed: 03/02/2024] Open
Abstract
Skeletal muscle function crucially depends on innervation while repair of skeletal muscle relies on resident muscle stem cells (MuSCs). However, it is poorly understood how innervation affects MuSC properties and thereby regeneration of skeletal muscle. Here, we report that loss of innervation causes precocious activation of MuSCs concomitant with the expression of markers of myogenic differentiation. This aberrant activation of MuSCs after loss of innervation is accompanied by profound alterations on the mRNA and protein level. Combination of muscle injury with loss of innervation results in impaired regeneration of skeletal muscle including shifts in myogenic populations concomitant with delayed maturation of regenerating myofibers. We further demonstrate that loss of innervation leads to alterations in myofibers and their secretome, which then affect MuSC behavior. In particular, we identify an increased secretion of Osteopontin and transforming growth factor beta 1 (Tgfb1) by myofibers isolated from mice which had undergone sciatic nerve transection. The altered secretome results in the upregulation of early activating transcription factors, such as Junb, and their target genes in MuSCs. However, the combination of different secreted factors from myofibers after loss of innervation is required to cause the alterations observed in MuSCs after loss of innervation. These data demonstrate that loss of innervation first affects myofibers causing alterations in their secretome which then affect MuSCs underscoring the importance of proper innervation for MuSC functionality and regeneration of skeletal muscle.
Collapse
Affiliation(s)
- Henriette Henze
- Leibniz Institute on Aging - Fritz Lipmann Institute, Beutenbergstrasse 11, 07745, Jena, Germany
| | - Sören S Hüttner
- Leibniz Institute on Aging - Fritz Lipmann Institute, Beutenbergstrasse 11, 07745, Jena, Germany
| | - Philipp Koch
- Leibniz Institute on Aging - Fritz Lipmann Institute, Beutenbergstrasse 11, 07745, Jena, Germany
| | - Svenja C Schüler
- Leibniz Institute on Aging - Fritz Lipmann Institute, Beutenbergstrasse 11, 07745, Jena, Germany
| | - Marco Groth
- Leibniz Institute on Aging - Fritz Lipmann Institute, Beutenbergstrasse 11, 07745, Jena, Germany
| | - Björn von Eyss
- Leibniz Institute on Aging - Fritz Lipmann Institute, Beutenbergstrasse 11, 07745, Jena, Germany
| | - Julia von Maltzahn
- Leibniz Institute on Aging - Fritz Lipmann Institute, Beutenbergstrasse 11, 07745, Jena, Germany.
- Faculty of Health Sciences Brandenburg, Brandenburg University of Technology Cottbus - Senftenberg, Universitätsplatz 1, 01968, Senftenberg, Germany.
| |
Collapse
|
3
|
Careccia G, Mangiavini L, Cirillo F. Regulation of Satellite Cells Functions during Skeletal Muscle Regeneration: A Critical Step in Physiological and Pathological Conditions. Int J Mol Sci 2023; 25:512. [PMID: 38203683 PMCID: PMC10778731 DOI: 10.3390/ijms25010512] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Revised: 12/20/2023] [Accepted: 12/28/2023] [Indexed: 01/12/2024] Open
Abstract
Skeletal muscle regeneration is a complex process involving the generation of new myofibers after trauma, competitive physical activity, or disease. In this context, adult skeletal muscle stem cells, also known as satellite cells (SCs), play a crucial role in regulating muscle tissue homeostasis and activating regeneration. Alterations in their number or function have been associated with various pathological conditions. The main factors involved in the dysregulation of SCs' activity are inflammation, oxidative stress, and fibrosis. This review critically summarizes the current knowledge on the role of SCs in skeletal muscle regeneration. It examines the changes in the activity of SCs in three of the most common and severe muscle disorders: sarcopenia, muscular dystrophy, and cancer cachexia. Understanding the molecular mechanisms involved in their dysregulations is essential for improving current treatments, such as exercise, and developing personalized approaches to reactivate SCs.
Collapse
Affiliation(s)
- Giorgia Careccia
- Department of Biosciences, University of Milan, 20133 Milan, Italy;
| | - Laura Mangiavini
- IRCCS Istituto Ortopedico Galeazzi, 20161 Milan, Italy;
- Department of Biomedical Sciences for Health, University of Milan, 20133 Milan, Italy
| | - Federica Cirillo
- IRCCS Policlinico San Donato, 20097 San Donato Milanese, Italy
- Institute for Molecular and Translational Cardiology (IMTC), 20097 San Donato Milanese, Italy
| |
Collapse
|
4
|
AMPK Phosphorylation Impacts Apoptosis in Differentiating Myoblasts Isolated from Atrophied Rat Soleus Muscle. Cells 2023; 12:cells12060920. [PMID: 36980261 PMCID: PMC10047078 DOI: 10.3390/cells12060920] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 02/25/2023] [Accepted: 03/10/2023] [Indexed: 03/19/2023] Open
Abstract
Regrowth of atrophied myofibers depends on muscle satellite cells (SCs) that exist outside the plasma membrane. Muscle atrophy appears to result in reduced number of SCs due to apoptosis. Given reduced AMP-activated protein kinase (AMPK) activity during differentiation of primary myoblasts derived from atrophic muscle, we hypothesized that there may be a potential link between AMPK and susceptibility of differentiating myoblasts to apoptosis. The aim of this study was to estimate the effect of AMPK activation (via AICAR treatment) on apoptosis in differentiating myoblasts derived from atrophied rat soleus muscle. Thirty rats were randomly assigned to the following two groups: control (C, n = 10) and 7-day hindlimb suspension (HS, n = 20). Myoblasts derived from the soleus muscles of HS rats were divided into two parts: AICAR-treated cells and non-treated cells. Apoptotic processes were evaluated by using TUNEL assay, RT-PCR and WB. In differentiating myoblasts derived from the atrophied soleus, there was a significant decrease (p < 0.05) in AMPK and ACC phosphorylation in parallel with increased number of apoptotic nuclei and a significant upregulation of pro-apoptotic markers (caspase-3, -9, BAX, p53) compared to the cells derived from control muscles. AICAR treatment of atrophic muscle-derived myoblasts during differentiation prevented reductions in AMPK and ACC phosphorylation as well as maintained the number of apoptotic nuclei and the expression of pro-apoptotic markers at the control levels. Thus, the maintenance of AMPK activity can suppress enhanced apoptosis in differentiating myoblasts derived from atrophied rat soleus muscle.
Collapse
|
5
|
Peña-Toledo MA, Luque E, LaTorre M, Jimena I, Leiva-Cepas F, Ruz-Caracuel I, Agüera E, Peña-Amaro J, Tunez I. The ultrastructure of muscle fibers and satellite cells in experimental autoimmune encephalomyelitis after treatment with transcranial magnetic stimulation. Ultrastruct Pathol 2022; 46:401-412. [PMID: 35994513 DOI: 10.1080/01913123.2022.2112330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/15/2022]
Abstract
In this study, we investigated the effect of transcranial magnetic stimulation (TMS) on the ultrastructure of muscle fibers and satellite cells in rats with experimental autoimmune encephalomyelitis (EAE). EAE-induced animals were treated with TMS (60 Hz at 0.7 mT) for 2 hours in the morning, once a day, 5 days a week, for 3 weeks, starting on day 15 post-immunization. The rats were sacrificed on day 36 post-immunization, and the soleus muscles were evaluated by light microscopy and transmission electron microscopy. Findings were compared with a non-treated EAE group. Electron microscopy analysis showed the presence of degenerated mitochondria, autophagic vacuoles, and altered myofibrils in non-treated EAE group. This correlates with the presence of acid phosphatase activity in muscle fibers and core-targetoid lesions with desmin immunohistochemistry. Most myonuclei in the EAE group showed apoptotic features. In contrast, EAE induced-TMS treated animals had less ultrastructural changes in the mitochondria and the myofibrils, together with less frequent apoptotic nuclear features. Peripheral desmin+ protrusions, as a marker of active satellite cells, were significantly increased in TMS-treated group. This correlates ultrastructurally with the presence of active features in satellite cells in the TMS group. In conclusion, the attenuation of ultrastructural alterations in muscle fibers and activation response of satellite cells caused by EAE indicated that skeletal muscle had a regenerative response to TMS.
Collapse
Affiliation(s)
- María Angeles Peña-Toledo
- Dementia and Multiple Sclerosis Unit, Neurology Service, Reina Sofia University Hospital, Cordoba, Spain.,Maimonides Institute for Biomedical Research IMIBIC, Cordoba, Spain
| | - Evelio Luque
- Maimonides Institute for Biomedical Research IMIBIC, Cordoba, Spain.,Department of Morphological Sciences, Histology Section, Faculty of Medicine and Nursing, University of Cordoba, Cordoba, Spain
| | - Manuel LaTorre
- Maimonides Institute for Biomedical Research IMIBIC, Cordoba, Spain.,Department of Biochemistry and Molecular Biology, Faculty of Medicine and Nursing, University of Cordoba, Cordoba, Spain
| | - Ignacio Jimena
- Maimonides Institute for Biomedical Research IMIBIC, Cordoba, Spain.,Department of Morphological Sciences, Histology Section, Faculty of Medicine and Nursing, University of Cordoba, Cordoba, Spain
| | - Fernando Leiva-Cepas
- Maimonides Institute for Biomedical Research IMIBIC, Cordoba, Spain.,Department of Morphological Sciences, Histology Section, Faculty of Medicine and Nursing, University of Cordoba, Cordoba, Spain.,Department of Pathology, Reina Sofía University Hospital, Córdoba, Spain
| | - Ignacio Ruz-Caracuel
- Department of Morphological Sciences, Histology Section, Faculty of Medicine and Nursing, University of Cordoba, Cordoba, Spain.,Department of Pathology, Ramon y Cajal University Hospital, IRYCIS, Madrid, Spain
| | - Eduardo Agüera
- Dementia and Multiple Sclerosis Unit, Neurology Service, Reina Sofia University Hospital, Cordoba, Spain.,Maimonides Institute for Biomedical Research IMIBIC, Cordoba, Spain
| | - J Peña-Amaro
- Maimonides Institute for Biomedical Research IMIBIC, Cordoba, Spain.,Department of Morphological Sciences, Histology Section, Faculty of Medicine and Nursing, University of Cordoba, Cordoba, Spain
| | - Isaac Tunez
- Maimonides Institute for Biomedical Research IMIBIC, Cordoba, Spain.,Department of Biochemistry and Molecular Biology, Faculty of Medicine and Nursing, University of Cordoba, Cordoba, Spain.,Cooperative Research Thematic Excellent Network on Brain Stimulation (REDESTIM), Ministery for Economy, Industry and Competitiveness, Madrid, Spain
| |
Collapse
|
6
|
A Long Journey before Cycling: Regulation of Quiescence Exit in Adult Muscle Satellite Cells. Int J Mol Sci 2022; 23:ijms23031748. [PMID: 35163665 PMCID: PMC8836154 DOI: 10.3390/ijms23031748] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 01/28/2022] [Accepted: 01/30/2022] [Indexed: 02/04/2023] Open
Abstract
Skeletal muscle harbors a pool of stem cells called muscle satellite cells (MuSCs) that are mainly responsible for its robust regenerative capacities. Adult satellite cells are mitotically quiescent in uninjured muscles under homeostasis, but they exit quiescence upon injury to re-enter the cell cycle to proliferate. While most of the expanded satellites cells differentiate and fuse to form new myofibers, some undergo self-renewal to replenish the stem cell pool. Specifically, quiescence exit describes the initial transition of MuSCs from quiescence to the first cell cycle, which takes much longer than the time required for subsequent cell cycles and involves drastic changes in cell size, epigenetic and transcriptomic profiles, and metabolic status. It is, therefore, an essential period indispensable for the success of muscle regeneration. Diverse mechanisms exist in MuSCs to regulate quiescence exit. In this review, we summarize key events that occur during quiescence exit in MuSCs and discuss the molecular regulation of this process with an emphasis on multiple levels of intrinsic regulatory mechanisms. A comprehensive understanding of how quiescence exit is regulated will facilitate satellite cell-based muscle regenerative therapies and advance their applications in various disease and aging conditions.
Collapse
|
7
|
Wong A, Garcia SM, Tamaki S, Striedinger K, Barruet E, Hansen SL, Young DM, Pomerantz JH. Satellite cell activation and retention of muscle regenerative potential after long-term denervation. Stem Cells 2021; 39:331-344. [PMID: 33326654 DOI: 10.1002/stem.3316] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Accepted: 12/01/2020] [Indexed: 12/17/2022]
Abstract
Irreversible denervation atrophy remains an unsolved clinical problem, and the role of skeletal muscle stem cell (MuSC, satellite cell) depletion in this process is unclear. We investigated the ability of MuSCs to regenerate muscle in the context of denervation. Three to 12 months following sciatic denervation in mice, MuSC number, size, EdU uptake, rate of division, and mitochondrial activity were increased. Following acute myotoxin injury, denervated muscles formed new muscle fibers in situ. MuSCs isolated via flow cytometry from denervated mouse muscle, or from atrophic denervated gluteus maximus muscles of humans with complete spinal cord injuries two decades prior, formed new muscle fibers and reoccupied the anatomic niche after transplantation into uninjured muscle. Our results show unequivocally that, even after prolonged denervation, MuSCs retain intrinsic regenerative potential similar to that of uninjured MuSCs. Treatment of denervation atrophy will require elucidating the non-MuSC environmental changes in muscle that prevent functional regeneration.
Collapse
Affiliation(s)
- Alvin Wong
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Program in Craniofacial Biology, Eli and Edythe Broad Center of Regeneration Medicine, University of California, San Francisco, California, USA
| | - Steven M Garcia
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Program in Craniofacial Biology, Eli and Edythe Broad Center of Regeneration Medicine, University of California, San Francisco, California, USA
| | - Stanley Tamaki
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Program in Craniofacial Biology, Eli and Edythe Broad Center of Regeneration Medicine, University of California, San Francisco, California, USA
| | - Katharine Striedinger
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Program in Craniofacial Biology, Eli and Edythe Broad Center of Regeneration Medicine, University of California, San Francisco, California, USA
| | - Emilie Barruet
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Program in Craniofacial Biology, Eli and Edythe Broad Center of Regeneration Medicine, University of California, San Francisco, California, USA
| | - Scott L Hansen
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Program in Craniofacial Biology, Eli and Edythe Broad Center of Regeneration Medicine, University of California, San Francisco, California, USA
| | - David M Young
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Program in Craniofacial Biology, Eli and Edythe Broad Center of Regeneration Medicine, University of California, San Francisco, California, USA
| | - Jason H Pomerantz
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Program in Craniofacial Biology, Eli and Edythe Broad Center of Regeneration Medicine, University of California, San Francisco, California, USA
- Department of Orofacial Sciences, University of California, San Francisco, California, USA
| |
Collapse
|
8
|
Rhode SC, Beier JP, Ruhl T. Adipose tissue stem cells in peripheral nerve regeneration-In vitro and in vivo. J Neurosci Res 2020; 99:545-560. [PMID: 33070351 DOI: 10.1002/jnr.24738] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 09/16/2020] [Accepted: 09/30/2020] [Indexed: 12/12/2022]
Abstract
After peripheral nerve injury, Schwann cells (SCs) are crucially involved in several steps of the subsequent regenerative processes, such as the Wallerian degeneration. They promote lysis and phagocytosis of myelin, secrete numbers of neurotrophic factors and cytokines, and recruit macrophages for a biological debridement. However, nerve injuries with a defect size of >1 cm do not show proper tissue regeneration and require a surgical nerve gap reconstruction. To find a sufficient alternative to the current gold standard-the autologous nerve transplant-several cell-based therapies have been developed and were experimentally investigated. One approach aims on the use of adipose tissue stem cells (ASCs). These are multipotent mesenchymal stromal cells that can differentiate into multiple phenotypes along the mesodermal lineage, such as osteoblasts, chondrocytes, and myocytes. Furthermore, ASCs also possess neurotrophic features, that is, they secrete neurotrophic factors like the nerve growth factor, brain-derived neurotrophic factor, neurotrophin-3, ciliary neurotrophic factor, glial cell-derived neurotrophic factor, and artemin. They can also differentiate into the so-called Schwann cell-like cells (SCLCs). These cells share features with naturally occurring SCs, as they also promote nerve regeneration in the periphery. This review gives a comprehensive overview of the use of ASCs in peripheral nerve regeneration and peripheral nerve tissue engineering both in vitro and in vivo. While the sustainability of differentiation of ASCs to SCLCs in vivo is still questionable, ASCs used with different nerve conduits, such as hydrogels or silk fibers, have been shown to promote nerve regeneration.
Collapse
Affiliation(s)
- Sophie Charlotte Rhode
- Department of Plastic Surgery, Hand Surgery and Burn Center, University Hospital RWTH Aachen, Aachen, Germany
| | - Justus Patrick Beier
- Department of Plastic Surgery, Hand Surgery and Burn Center, University Hospital RWTH Aachen, Aachen, Germany
| | - Tim Ruhl
- Department of Plastic Surgery, Hand Surgery and Burn Center, University Hospital RWTH Aachen, Aachen, Germany
| |
Collapse
|
9
|
Bennett BT, Mohamed JS, Alway SE. The Effects of Calcium- β-Hydroxy- β-Methylbutyrate on Aging-Associated Apoptotic Signaling and Muscle Mass and Function in Unloaded but Nonatrophied Extensor Digitorum Longus Muscles of Aged Rats. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:3938672. [PMID: 32774671 PMCID: PMC7396042 DOI: 10.1155/2020/3938672] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 06/12/2020] [Accepted: 06/19/2020] [Indexed: 11/18/2022]
Abstract
Beta-hydroxy-beta-methylbutyrate (HMB), a naturally occurring leucine metabolite, has been shown to attenuate plantar flexor muscle loss and increase myogenic stem cell activation during reloading after a period of significant muscle wasting by disuse in old rodents. However, it was less clear if HMB would alter dorsiflexor muscle response to unloading or reloading when there was no significant atrophy that was induced by unloading. In this study, we tested if calcium HMB (Ca-HMB) would improve muscle function and alter apoptotic signaling in the extensor digitorum longus (EDL) of aged animals that were unloaded but did not undergo atrophy. The EDL muscle was unloaded for 14 days by hindlimb suspension (HS) in aged (34-36 mo.) male Fisher 344 × Brown Norway rats. The rats were removed from HS and allowed normal cage ambulation for 14 days of reloading (R). Throughout the study, the rats were gavaged daily with 170 mg of Ca-HMB or water 7 days prior to HS, then throughout 14 days of HS and 14 days of recovery after removing HS. The animals' body weights were significantly reduced by ~18% after 14 days of HS and continued to decline by ~22% during R as compared to control conditions; however, despite unloading, EDL did not atrophy by HS, nor did it increase in mass after R. No changes were observed in EDL twitch contraction time, force production, fatigue resistance, fiber cross-sectional area, or markers of nuclear apoptosis (myonuclei + satellite cells) after HS or R. While HS and R increased the proapoptotic Bax protein abundance, BCL-2 abundance was also increased as was the frequency of TUNEL-positive myonuclei and satellite cells, yet muscle mass and fiber cross-sectional area did not change and Ca-HMB treatment had no effect reducing apoptotic signaling. These data indicate that (i) increased apoptotic signaling preceded muscle atrophy or occurred without significant EDL atrophy and (ii) that Ca-HMB treatment did not improve EDL signaling, muscle mass, or muscle function in aged rats, when HS and R did not impact mass or function.
Collapse
Affiliation(s)
- Brian T. Bennett
- Laboratory of Muscle Biology and Sarcopenia, Department of Exercise Physiology, West Virginia University School of Medicine, Morgantown, WV, USA 26506
| | - Junaith S. Mohamed
- Center for Muscle, Metabolism and Neuropathology, Division of Rehabilitation Sciences, College of Health Professions, University of Tennessee Health Science Center, Memphis, TN, USA 38163
- Laboratory of Nerve and Muscle, Department of Diagnostic and Health Sciences, College of Health Professions, University of Tennessee Health Science Center, Memphis, TN, USA 38163
- Laboratory of Muscle Biology and Sarcopenia, Department of Physical Therapy, College of Health Professions, University of Tennessee Health Science Center, Memphis, TN, USA 38163
| | - Stephen E. Alway
- Laboratory of Muscle Biology and Sarcopenia, Department of Exercise Physiology, West Virginia University School of Medicine, Morgantown, WV, USA 26506
- Center for Muscle, Metabolism and Neuropathology, Division of Rehabilitation Sciences, College of Health Professions, University of Tennessee Health Science Center, Memphis, TN, USA 38163
- Laboratory of Muscle Biology and Sarcopenia, Department of Physical Therapy, College of Health Professions, University of Tennessee Health Science Center, Memphis, TN, USA 38163
- Department of Physiology, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, USA 38163
| |
Collapse
|
10
|
Farhang-Sardroodi S, Wilkie KP. Mathematical Model of Muscle Wasting in Cancer Cachexia. J Clin Med 2020; 9:jcm9072029. [PMID: 32605273 PMCID: PMC7409297 DOI: 10.3390/jcm9072029] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 06/17/2020] [Accepted: 06/19/2020] [Indexed: 12/12/2022] Open
Abstract
Cancer cachexia is a debilitating condition characterized by an extreme loss of skeletal muscle mass, which negatively impacts patients' quality of life, reduces their ability to sustain anti-cancer therapies, and increases the risk of mortality. Recent discoveries have identified the myostatin/activin A/ActRIIB pathway as critical to muscle wasting by inducing satellite cell quiescence and increasing muscle-specific ubiquitin ligases responsible for atrophy. Remarkably, pharmacological blockade of the ActRIIB pathway has been shown to reverse muscle wasting and prolong the survival time of tumor-bearing animals. To explore the implications of this signaling pathway and potential therapeutic targets in cachexia, we construct a novel mathematical model of muscle tissue subjected to tumor-derived cachectic factors. The model formulation tracks the intercellular interactions between cancer cell, satellite cell, and muscle cell populations. The model is parameterized by fitting to colon-26 mouse model data, and the analysis provides insight into tissue growth in healthy, cancerous, and post-cachexia treatment conditions. Model predictions suggest that cachexia fundamentally alters muscle tissue health, as measured by the stem cell ratio, and this is only partially recovered by anti-cachexia treatment. Our mathematical findings suggest that after blocking the myostatin/activin A pathway, partial recovery of cancer-induced muscle loss requires the activation and proliferation of the satellite cell compartment with a functional differentiation program.
Collapse
|
11
|
Lu X, Liang B, Li S, Chen Z, Chang W. Modulation of HOXA9 after skeletal muscle denervation and reinnervation. Am J Physiol Cell Physiol 2020; 318:C1154-C1165. [PMID: 32233950 DOI: 10.1152/ajpcell.00055.2020] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Homeobox A9 (HOXA9), the expression of which is promoted by mixed lineage leukemia 1 (MLL1) and WD-40 repeat protein 5 (WDR5), is a homeodomain-containing transcription factor that plays an essential role in regulating stem cell activity. HOXA9 has been found to inhibit skeletal muscle regeneration and delay recovery after muscle wounding in aged mice, but little is known about its role in denervated/reinnervated muscles. We performed detailed time-dependent expression analyses of HOXA9 and its promoters, MLL1 and WDR5, in rat gastrocnemius muscles after the following three types of sciatic nerve surgeries: nerve transection (denervation), end-to-end repair (repair), and sham operation (sham). Then, the specific mechanisms of HOXA9 were detected in vitro by transfecting primary satellite cells with empty pIRES2-DsRed2, pIRES2-DsRed2-HOXA9, empty pPLK/GFP-Puro, and pPLK/GFP-Puro-HOXA9 small hairpin RNA (shRNA) plasmids. We found, for the first time, that HOXA9 protein expression simultaneously increased with increasing denervated muscle atrophy severity and that upregulated MLL1 and WDR5 expression was partly associated with denervation. Indeed, in vitro experiments revealed that HOXA9 inhibited myogenic differentiation, affected the best known atrophic signaling pathways, and promoted apoptosis but did not eliminate the differentiation potential of primary satellite cells. HOXA9 may promote denervated muscle atrophy by regulating the activity of satellite cells.
Collapse
Affiliation(s)
- Xiaomei Lu
- Department of Orthopedics, The Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Bingsheng Liang
- Department of Orthopedics, The Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Shuaijie Li
- Department of Orthopedics, Tongji Hospital, Tongji University School of Medicine, Shanghai, China
| | - Zhi Chen
- Department of Orthopedics, The Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Wenkai Chang
- Department of Orthopedics, The Second Hospital of Shanxi Medical University, Taiyuan, China
| |
Collapse
|
12
|
The Role of Muscle Stem Cells in Regeneration and Recovery after Denervation: A Review. Plast Reconstr Surg 2019; 143:779-788. [PMID: 30817650 DOI: 10.1097/prs.0000000000005370] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Skeletal muscle denervation is a complex clinical problem that still lacks a comprehensive solution. Previous studies have suggested that prolonged periods of denervation lead to a decline in the muscle stem cell population, negatively affecting the ability of muscle to regenerate following reinnervation. Recent advances in the understanding of muscle stem cell biology, along with new techniques that increase the ability to identify and manipulate these cells, provide an opportunity to definitively address the impact of muscle stem cells in recovery from denervation and their potential role in treatment. METHODS A comprehensive review of the literature on the biology of muscle denervation, and the effect of denervation injury on muscle stem cell behavior, was performed. RESULTS In this review, the authors discuss the current understanding of muscle stem cell biology in the setting of denervation atrophy, review barriers to successful reinnervation, and review options available to patients following denervation injury. The authors also discuss potential use of muscle stem cells in future therapies. CONCLUSIONS Although the clinical treatment of prolonged denervation injury has improved in recent years, regeneration of native muscle remains elusive. Muscle stem cells have been demonstrated to be of central importance in muscle regeneration following injury, and may be a powerful tool that provides effective new options for future treatments. Additional work clarifying the effect of denervation injury on satellite cells is needed to determine whether they are a limiting factor in recovery and to demonstrate whether their clinical use as a cell-based therapy in denervation injury can be efficacious.
Collapse
|
13
|
Gordon T, Wood P, Sulaiman OAR. Long-Term Denervated Rat Schwann Cells Retain Their Capacity to Proliferate and to Myelinate Axons in vitro. Front Cell Neurosci 2019; 12:511. [PMID: 30666188 PMCID: PMC6330764 DOI: 10.3389/fncel.2018.00511] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Accepted: 12/10/2018] [Indexed: 12/19/2022] Open
Abstract
Functional recovery is poor after peripheral nerve injury and delayed surgical repair or when nerves must regenerate over long distances to reinnervate distant targets. A reduced capacity of Schwann cells (SCs) in chronically denervated distal nerve stumps to support and interact with regenerating axons may account for the poor outcome. In an in vitro system, we examined the capacity of adult, long-term denervated rat SCs to proliferate and to myelinate neurites in co-cultures with fetal dorsal root ganglion (DRG) neurons. Non-neuronal cells were counted immediately after their isolation from the distal sciatic nerve stumps that were subjected to acute denervation of 7 days or chronic denervation of either 7 weeks or 17 months. Thereafter, equal numbers of the non-neural cells were co-cultured with purified dissociated DRG neurons for 5 days. The co-cultures were then treated with 3H-Thymidine for 24 h to quantitate SC proliferation with S100 immunostaining and autoradiography. After a 24-day period of co-culture, Sudan Black staining was used to visualize and count myelin segments that were elaborated around DRG neurites by the SCs. Isolated non-neural cells from 7-week chronically denervated nerve stumps increased 2.5-fold in number compared to ~2 million in 7 day acutely denervated stumps. There were only <0.2 million cells in the 17-week chronically denervated stumps. Nonetheless, these chronically denervated SCs maintained their proliferative capacity although the capacity was reduced to 30% in the 17-month chronically denervated distal nerve stumps. Moreover, the chronically denervated SCs retained their capacity to myelinate DRG neurites: there was extensive myelination of the neurites by the acutely and chronically denervated SCs after 24 days co-culture. There were no significant differences in the extent of myelination. We conclude that the low numbers of surviving SCs in chronically denervated distal nerve stumps retain their ability to respond to axonal signals to divide and to elaborate myelin. However, their low numbers consequent to their poor survival and their reduced capacity to proliferate account, at least in part, for the poor functional recovery after delayed surgical repair of injured nerve and/or the repair of injured nerves far from their target organs.
Collapse
Affiliation(s)
- Tessa Gordon
- Division of Neuroscience, Faculty of Medicine, University of Alberta, Edmonton, AB, Canada
| | - Patrick Wood
- The Miami Project to Cure Paralysis/Department of Neurological Surgery, University of Miami School of Medicine, Miami, FL, United States
| | - Olawale A R Sulaiman
- Department of Neurosurgery, Ochsner Medical Center, New Orleans, LA, United States
| |
Collapse
|
14
|
Forcina L, Miano C, Musarò A. The physiopathologic interplay between stem cells and tissue niche in muscle regeneration and the role of IL-6 on muscle homeostasis and diseases. Cytokine Growth Factor Rev 2018; 41:1-9. [PMID: 29778303 DOI: 10.1016/j.cytogfr.2018.05.001] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Accepted: 05/03/2018] [Indexed: 12/11/2022]
Abstract
Skeletal muscle is a complex, dynamic tissue characterized by an elevated plasticity. Although the adult muscle is mainly composed of multinucleated fibers with post mitotic nuclei, it retains a remarkable ability to regenerate in response to traumatic events. The regenerative potential of the adult skeletal muscle relies in the activity of satellite cells, mononucleated cells residing within the muscle in intimate association with myofibers. Satellite cells normally remain quiescent in their sublaminar position, sporadically entering the cell cycle to guarantee an efficient cellular turnover, by fusing with pre-existing myofibers, and to maintain the stem cell pool. However, after muscle injury satellite cells undergo an extensive increase of their activity in response to environmental stimuli, thereby participating to the regeneration of a functional muscle tissue. Nevertheless, regeneration is affected in several pathologic conditions and by a wide range of environmental signals that are highly variable, not only through time, but also depending on the physiological or pathological conditions of the musculature. Among these factors, the interleukin-6 (IL-6) plays a critical physiopathologic role on muscle homeostasis and diseases. The basis of muscle regeneration and the impact of IL-6 on the physiopathology of skeletal muscle will be discussed.
Collapse
Affiliation(s)
- Laura Forcina
- DAHFMO-Unit of Histology and Medical Embryology, Sapienza University of Rome, Laboratory Affiliated to Istituto Pasteur Italia - Fondazione Cenci Bolognetti, Via A. Scarpa, 14, Rome 00161, Italy
| | - Carmen Miano
- DAHFMO-Unit of Histology and Medical Embryology, Sapienza University of Rome, Laboratory Affiliated to Istituto Pasteur Italia - Fondazione Cenci Bolognetti, Via A. Scarpa, 14, Rome 00161, Italy
| | - Antonio Musarò
- DAHFMO-Unit of Histology and Medical Embryology, Sapienza University of Rome, Laboratory Affiliated to Istituto Pasteur Italia - Fondazione Cenci Bolognetti, Via A. Scarpa, 14, Rome 00161, Italy; Center for Life Nano Science@Sapienza, Istituto Italiano di Tecnologia, Rome 00161, Italy.
| |
Collapse
|
15
|
Dinulovic I, Furrer R, Handschin C. Plasticity of the Muscle Stem Cell Microenvironment. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1041:141-169. [PMID: 29204832 DOI: 10.1007/978-3-319-69194-7_8] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Satellite cells (SCs) are adult muscle stem cells capable of repairing damaged and creating new muscle tissue throughout life. Their functionality is tightly controlled by a microenvironment composed of a wide variety of factors, such as numerous secreted molecules and different cell types, including blood vessels, oxygen, hormones, motor neurons, immune cells, cytokines, fibroblasts, growth factors, myofibers, myofiber metabolism, the extracellular matrix and tissue stiffness. This complex niche controls SC biology-quiescence, activation, proliferation, differentiation or renewal and return to quiescence. In this review, we attempt to give a brief overview of the most important players in the niche and their mutual interaction with SCs. We address the importance of the niche to SC behavior under physiological and pathological conditions, and finally survey the significance of an artificial niche both for basic and translational research purposes.
Collapse
|
16
|
Wang Y, Bai X, Wang Z, Cao J, Dong Y, Dong Y, Chen Y. Various LED Wavelengths Affected Myofiber Development and Satellite Cell Proliferation of Chick Embryos via the IGF-1 Signaling Pathway. Photochem Photobiol 2017; 93:1492-1501. [PMID: 28708285 DOI: 10.1111/php.12806] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2017] [Accepted: 06/16/2017] [Indexed: 01/08/2023]
Abstract
An effect of monochromatic light illumination on muscle mass has been discovered in chickens; however, its effect on the development of embryonic muscle remains unclear. Our previous studies demonstrated that monochromatic green light promoted satellite cell proliferation and muscle growth in posthatching broilers. In this study, we investigated the effects and mechanisms of monochromatic light exposure on muscle development in late embryogenesis. Seven hundred and fifty fertile broiler eggs were randomly assigned to blue (B-group), green (G-group), red (R-group), white (W-group) lights or darkness (D-group) throughout the incubation period. The muscle weight and fiber size were highest in the G-group compared to the other groups during embryonic days (E) 17 to E20. The proliferation of satellite cells isolated from the G-group was highest, and in vivo green light remarkably increased the number of proliferating cell nuclear antigen (PCNA)-positive cells in skeletal muscle. Meanwhile, plasma IGF-1 was higher (15.5-16.2%) in the G-group than that in D- and R-groups, and the satellite cells isolated from the G-group had a more sensitive response to IGF-1. These findings demonstrate green monochromatic photobiomodulation promoted the muscle growth and satellite cell proliferation was related to the IGF-1 signaling pathway in late embryogenesis.
Collapse
Affiliation(s)
- Yao Wang
- Laboratory of Anatomy of Domestic Animals, College of Animal Medicine, China Agricultural University, Beijing, China
| | - Xinjie Bai
- Laboratory of Anatomy of Domestic Animals, College of Animal Medicine, China Agricultural University, Beijing, China
| | - Zixu Wang
- Laboratory of Anatomy of Domestic Animals, College of Animal Medicine, China Agricultural University, Beijing, China
| | - Jing Cao
- Laboratory of Anatomy of Domestic Animals, College of Animal Medicine, China Agricultural University, Beijing, China
| | - Yulan Dong
- Laboratory of Anatomy of Domestic Animals, College of Animal Medicine, China Agricultural University, Beijing, China
| | - Yanjun Dong
- Laboratory of Anatomy of Domestic Animals, College of Animal Medicine, China Agricultural University, Beijing, China
| | - Yaoxing Chen
- Laboratory of Anatomy of Domestic Animals, College of Animal Medicine, China Agricultural University, Beijing, China
| |
Collapse
|
17
|
Handschin C, Mortezavi A, Plock J, Eberli D. External physical and biochemical stimulation to enhance skeletal muscle bioengineering. Adv Drug Deliv Rev 2015; 82-83:168-175. [PMID: 25453267 PMCID: PMC4444527 DOI: 10.1016/j.addr.2014.10.021] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2014] [Revised: 09/30/2014] [Accepted: 10/15/2014] [Indexed: 12/13/2022]
Abstract
PURPOSE OF REVIEW Cell based muscle tissue engineering carries the potential to revert the functional loss of muscle tissue caused by disease and trauma. Although muscle tissue can be bioengineered using various precursor cells, major limitations still remain. RECENT FINDINGS In the last decades several cellular pathways playing a crucial role in muscle tissue regeneration have been described. These pathways can be influenced by external stimuli and they not only orchestrate the regenerative process after physiologic wear and muscle trauma, but also play an important part in aging and maintaining the stem cell niche, which is required to maintain long-term muscle function. SUMMARY In this review article we will highlight possible new avenues using external physical and biochemical stimulation in order to optimize muscle bioengineering.
Collapse
Affiliation(s)
| | | | | | - Daniel Eberli
- corresponding author: Daniel Eberli MD PhD, Division of Urology, University Hospital Zürich, University of Zürich, Frauenklinikstrasse 10, 8091 Zürich, Switzerland, Phone: +41 44 255 11 11, Fax: +41 44 255 96 20,
| |
Collapse
|
18
|
Brooks NE, Myburgh KH. Skeletal muscle wasting with disuse atrophy is multi-dimensional: the response and interaction of myonuclei, satellite cells and signaling pathways. Front Physiol 2014; 5:99. [PMID: 24672488 PMCID: PMC3955994 DOI: 10.3389/fphys.2014.00099] [Citation(s) in RCA: 130] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2013] [Accepted: 02/27/2014] [Indexed: 12/25/2022] Open
Abstract
Maintenance of skeletal muscle is essential for health and survival. There are marked losses of skeletal muscle mass as well as strength and physiological function under conditions of low mechanical load, such as space flight, as well as ground based models such as bed rest, immobilization, disuse, and various animal models. Disuse atrophy is caused by mechanical unloading of muscle and this leads to reduced muscle mass without fiber attrition. Skeletal muscle stem cells (satellite cells) and myonuclei are integrally involved in skeletal muscle responses to environmental changes that induce atrophy. Myonuclear domain size is influenced differently in fast and slow twitch muscle, but also by different models of muscle wasting, a factor that is not yet understood. Although the myonuclear domain is 3-dimensional this is rarely considered. Apoptosis as a mechanism for myonuclear loss with atrophy is controversial, whereas cell death of satellite cells has not been considered. Molecular signals such as myostatin/SMAD pathway, MAFbx, and MuRF1 E3 ligases of the ubiquitin proteasome pathway and IGF1-AKT-mTOR pathway are 3 distinctly different contributors to skeletal muscle protein adaptation to disuse. Molecular signaling pathways activated in muscle fibers by disuse are rarely considered within satellite cells themselves despite similar exposure to unloading or low mechanical load. These molecular pathways interact with each other during atrophy and also when various interventions are applied that could alleviate atrophy. Re-applying mechanical load is an obvious method to restore muscle mass, however how nutrient supplementation (e.g., amino acids) may further enhance recovery (or reduce atrophy despite unloading or ageing) is currently of great interest. Satellite cells are particularly responsive to myostatin and to growth factors. Recently, the hibernating squirrel has been identified as an innovative model to study resistance to atrophy.
Collapse
Affiliation(s)
- Naomi E Brooks
- Health and Exercise Science Research Group, School of Sport, University of Stirling Stirling, UK
| | - Kathryn H Myburgh
- Muscle Research Group, Department of Physiological Sciences, Stellenbosch University Stellenbosch, South Africa
| |
Collapse
|
19
|
Montarras D, L'honoré A, Buckingham M. Lying low but ready for action: the quiescent muscle satellite cell. FEBS J 2013; 280:4036-50. [DOI: 10.1111/febs.12372] [Citation(s) in RCA: 151] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2013] [Revised: 05/24/2013] [Accepted: 05/28/2013] [Indexed: 12/21/2022]
Affiliation(s)
- Didier Montarras
- Department of Developmental and Stem Cell Biology; CNRS URA 2578; Institut Pasteur; Paris; France
| | - Aurore L'honoré
- Department of Developmental and Stem Cell Biology; CNRS URA 2578; Institut Pasteur; Paris; France
| | - Margaret Buckingham
- Department of Developmental and Stem Cell Biology; CNRS URA 2578; Institut Pasteur; Paris; France
| |
Collapse
|
20
|
Abstract
Adult skeletal muscle in mammals is a stable tissue under normal circumstances but has remarkable ability to repair after injury. Skeletal muscle regeneration is a highly orchestrated process involving the activation of various cellular and molecular responses. As skeletal muscle stem cells, satellite cells play an indispensible role in this process. The self-renewing proliferation of satellite cells not only maintains the stem cell population but also provides numerous myogenic cells, which proliferate, differentiate, fuse, and lead to new myofiber formation and reconstitution of a functional contractile apparatus. The complex behavior of satellite cells during skeletal muscle regeneration is tightly regulated through the dynamic interplay between intrinsic factors within satellite cells and extrinsic factors constituting the muscle stem cell niche/microenvironment. For the last half century, the advance of molecular biology, cell biology, and genetics has greatly improved our understanding of skeletal muscle biology. Here, we review some recent advances, with focuses on functions of satellite cells and their niche during the process of skeletal muscle regeneration.
Collapse
Affiliation(s)
- Hang Yin
- Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | | | | |
Collapse
|
21
|
Garland CB, Pomerantz JH. Regenerative strategies for craniofacial disorders. Front Physiol 2012; 3:453. [PMID: 23248598 PMCID: PMC3521957 DOI: 10.3389/fphys.2012.00453] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2012] [Accepted: 11/12/2012] [Indexed: 01/26/2023] Open
Abstract
Craniofacial disorders present markedly complicated problems in reconstruction because of the complex interactions of the multiple, simultaneously affected tissues. Regenerative medicine holds promise for new strategies to improve treatment of these disorders. This review addresses current areas of unmet need in craniofacial reconstruction and emphasizes how craniofacial tissues differ from their analogs elsewhere in the body. We present a problem-based approach to illustrate current treatment strategies for various craniofacial disorders, to highlight areas of need, and to suggest regenerative strategies for craniofacial bone, fat, muscle, nerve, and skin. For some tissues, current approaches offer excellent reconstructive solutions using autologous tissue or prosthetic materials. Thus, new “regenerative” approaches would need to offer major advantages in order to be adopted. In other tissues, the unmet need is great, and we suggest the greatest regenerative need is for muscle, skin, and nerve. The advent of composite facial tissue transplantation and the development of regenerative medicine are each likely to add important new paradigms to our treatment of craniofacial disorders.
Collapse
Affiliation(s)
- Catharine B Garland
- Department of Surgery, Division of Plastic and Reconstructive Surgery, University of California San Francisco San Francisco, CA, USA ; Craniofacial and Mesenchymal Biology Program, University of California San Francisco San Francisco, CA, USA
| | | |
Collapse
|
22
|
Nagpal P, Plant PJ, Correa J, Bain A, Takeda M, Kawabe H, Rotin D, Bain JR, Batt JAE. The ubiquitin ligase Nedd4-1 participates in denervation-induced skeletal muscle atrophy in mice. PLoS One 2012; 7:e46427. [PMID: 23110050 PMCID: PMC3482220 DOI: 10.1371/journal.pone.0046427] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2012] [Accepted: 08/29/2012] [Indexed: 11/23/2022] Open
Abstract
Skeletal muscle atrophy is a consequence of muscle inactivity resulting from denervation, unloading and immobility. It accompanies many chronic disease states and also occurs as a pathophysiologic consequence of normal aging. In all these conditions, ubiquitin-dependent proteolysis is a key regulator of the loss of muscle mass, and ubiquitin ligases confer specificity to this process by interacting with, and linking ubiquitin moieties to target substrates through protein∶protein interaction domains. Our previous work suggested that the ubiquitin-protein ligase Nedd4-1 is a potential mediator of skeletal muscle atrophy associated with inactivity (denervation, unloading and immobility). Here we generated a novel tool, the Nedd4-1 skeletal muscle-specific knockout mouse (myoCre;Nedd4-1flox/flox) and subjected it to a well validated model of denervation induced skeletal muscle atrophy. The absence of Nedd4-1 resulted in increased weights and cross-sectional area of type II fast twitch fibres of denervated gastrocnemius muscle compared with wild type littermates controls, at seven and fourteen days following tibial nerve transection. These effects are not mediated by the Nedd4-1 substrates MTMR4, FGFR1 and Notch-1. These results demonstrate that Nedd4-1 plays an important role in mediating denervation-induced skeletal muscle atrophy in vivo.
Collapse
MESH Headings
- Animals
- Blotting, Western
- Cells, Cultured
- Endosomal Sorting Complexes Required for Transport/genetics
- Endosomal Sorting Complexes Required for Transport/metabolism
- Female
- Immunohistochemistry
- Male
- Mice
- Mice, Knockout
- Muscle Denervation
- Muscular Atrophy/genetics
- Muscular Atrophy/metabolism
- Myoblasts/cytology
- Myoblasts/metabolism
- Nedd4 Ubiquitin Protein Ligases
- Protein Tyrosine Phosphatases, Non-Receptor/genetics
- Protein Tyrosine Phosphatases, Non-Receptor/metabolism
- Receptor, Fibroblast Growth Factor, Type 1/genetics
- Receptor, Fibroblast Growth Factor, Type 1/metabolism
- Receptor, Notch1/genetics
- Receptor, Notch1/metabolism
- Satellite Cells, Skeletal Muscle/cytology
- Satellite Cells, Skeletal Muscle/metabolism
- Ubiquitin-Protein Ligases/genetics
- Ubiquitin-Protein Ligases/metabolism
Collapse
Affiliation(s)
- Preena Nagpal
- Keenan Research Centre of the LiKaShing Knowledge Institute, St Michaels Hospital, Toronto, Ontario, Canada
- Clinical Science Division, Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Pamela J. Plant
- Keenan Research Centre of the LiKaShing Knowledge Institute, St Michaels Hospital, Toronto, Ontario, Canada
| | - Judy Correa
- Keenan Research Centre of the LiKaShing Knowledge Institute, St Michaels Hospital, Toronto, Ontario, Canada
| | - Alexandra Bain
- Clinical Science Division, Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Michiko Takeda
- Department of Molecular Neurobiology, Max-Planck-Institute of Experimental Medicine, Goettingen, Germany
| | - Hiroshi Kawabe
- Department of Molecular Neurobiology, Max-Planck-Institute of Experimental Medicine, Goettingen, Germany
| | - Daniela Rotin
- Program in Cell Biology, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - James R. Bain
- Department of Surgery, McMaster University, Hamilton, Ontario, Canada
| | - Jane A. E. Batt
- Keenan Research Centre of the LiKaShing Knowledge Institute, St Michaels Hospital, Toronto, Ontario, Canada
- Clinical Science Division, Department of Medicine, University of Toronto, Toronto, Ontario, Canada
- * E-mail:
| |
Collapse
|
23
|
Geldanamycin accelerated peripheral nerve regeneration in comparison to FK-506 in vivo. Neuroscience 2012; 223:114-23. [DOI: 10.1016/j.neuroscience.2012.07.026] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2012] [Revised: 07/10/2012] [Accepted: 07/12/2012] [Indexed: 11/21/2022]
|
24
|
Myotubularin-deficient myoblasts display increased apoptosis, delayed proliferation, and poor cell engraftment. THE AMERICAN JOURNAL OF PATHOLOGY 2012; 181:961-8. [PMID: 22841819 DOI: 10.1016/j.ajpath.2012.05.016] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/01/2011] [Revised: 05/09/2012] [Accepted: 05/22/2012] [Indexed: 12/22/2022]
Abstract
X-linked myotubular myopathy is a severe congenital myopathy caused by deficiency of the lipid phosphatase, myotubularin. Recent studies of human tissue and animal models have discovered structural and physiological abnormalities in myotubularin-deficient muscle, but the impact of myotubularin deficiency on myogenic stem cells within muscles is unclear. In the present study, we evaluated the viability, proliferative capacity, and in vivo engraftment of myogenic cells obtained from severely symptomatic (Mtm1δ4) myotubularin-deficient mice. Mtm1δ4 muscle contains fewer myogenic cells than wild-type (WT) littermates, and the number of myogenic cells decreases with age. The behavior of Mtm1δ4 myoblasts is also abnormal, because they engraft poorly into C57BL/6/Rag1null/mdx5cv mice and display decreased proliferation and increased apoptosis compared with WT myoblasts. Evaluation of Mtm1δ4 animals at 21 and 42 days of life detected fewer satellite cells in Mtm1δ4 muscle compared with WT littermates, and the decrease in satellite cells correlated with progression of disease. In addition, analysis of WT and Mtm1δ4 regeneration after injury detected similar abnormalities of satellite cell function, with fewer satellite cells, fewer dividing cells, and increased apoptotic cells in Mtm1δ4 muscle. These studies demonstrate specific abnormalities in myogenic cell number and behavior that may relate to the progression of disease in myotubularin deficiency, and may also be used to develop in vitro assays by which novel treatment strategies can be assessed.
Collapse
|
25
|
Guo BS, Cheung KK, Yeung SS, Zhang BT, Yeung EW. Electrical stimulation influences satellite cell proliferation and apoptosis in unloading-induced muscle atrophy in mice. PLoS One 2012; 7:e30348. [PMID: 22253929 PMCID: PMC3257250 DOI: 10.1371/journal.pone.0030348] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2011] [Accepted: 12/14/2011] [Indexed: 11/18/2022] Open
Abstract
Muscle atrophy caused by disuse is accompanied by adverse physiological and functional consequences. Satellite cells are the primary source of skeletal muscle regeneration. Satellite cell dysfunction, as a result of impaired proliferative potential and/or increased apoptosis, is thought to be one of the causes contributing to the decreased muscle regeneration capacity in atrophy. We have previously shown that electrical stimulation improved satellite cell dysfunction. Here we test whether electrical stimulation can also enhance satellite cell proliferative potential as well as suppress apoptotic cell death in disuse-induced muscle atrophy. Eight-week-old male BALB/c mice were subjected to a 14-day hindlimb unloading procedure. During that period, one limb (HU-ES) received electrical stimulation (frequency: 20 Hz; duration: 3 h, twice daily) while the contralateral limb served as control (HU). Immunohistochemistry and western blotting techniques were used to characterize specific proteins in cell proliferation and apoptosis. The HU-ES soleus muscles showed significant improvement in muscle mass, cross-sectional area, and peak tetanic force relative to the HU limb (p<0.05). The satellite cell proliferative activity as detected within the BrdU+/Pax7+ population was significantly higher (p<0.05). The apoptotic myonuclei (detected by terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling) and the apoptotic satellite cells (detected by cleaved Poly [ADP-ribose] polymerase co-labeled with Pax7) were reduced (p<0.05) in the HU-ES limb. Furthermore the apoptosis-inducing factor and cleaved caspase-3 were down-regulated while the anti-apoptotic Bcl-2 protein was up-regulated (p<0.05), in the HU-ES limb. These findings suggest that the electrical stimulation paradigm provides an effective stimulus to rescue the loss of myonuclei and satellite cells in disuse muscle atrophy, thus maintaining a viable satellite cell pool for subsequent muscle regeneration. Optimization of stimulation parameters may enhance the outcome of the intervention.
Collapse
Affiliation(s)
- Bao-Sheng Guo
- Muscle Physiology Laboratory, Department of Rehabilitation Sciences, Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong
| | - Kwok-Kuen Cheung
- Muscle Physiology Laboratory, Department of Rehabilitation Sciences, Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong
| | - Simon S. Yeung
- Muscle Physiology Laboratory, Department of Rehabilitation Sciences, Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong
| | - Bao-Ting Zhang
- Muscle Physiology Laboratory, Department of Rehabilitation Sciences, Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong
| | - Ella W. Yeung
- Muscle Physiology Laboratory, Department of Rehabilitation Sciences, Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong
- * E-mail:
| |
Collapse
|
26
|
Liu XH, Yao S, Qiao RF, Levine AC, Kirschenbaum A, Pan J, Wu Y, Qin W, Bauman WA, Cardozo CP. Nandrolone reduces activation of Notch signaling in denervated muscle associated with increased Numb expression. Biochem Biophys Res Commun 2011; 414:165-9. [PMID: 21945932 DOI: 10.1016/j.bbrc.2011.09.048] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2011] [Accepted: 09/11/2011] [Indexed: 11/24/2022]
Abstract
Nandrolone, an anabolic steroid, slows denervation-atrophy in rat muscle. The molecular mechanisms responsible for this effect are not well understood. Androgens and anabolic steroids activate Notch signaling in animal models of aging and thereby mitigate sarcopenia. To explore the molecular mechanisms by which nandrolone prevents denervation-atrophy, we investigated the effects of nandrolone on Notch signaling in denervated rat gastrocnemius muscle. Denervation significantly increased Notch activity reflected by elevated levels of nuclear Notch intracellular domain (NICD) and expression of Hey1 (a Notch target gene). Activation was greatest at 7 and 35 days after denervation but remained present at 56 days after denervation. Activation of Notch in denervated muscle was prevented by nandrolone associated with upregulated expression of Numb mRNA and protein. These data demonstrate that denervation activates Notch signaling, and that nandrolone abrogates this response associated with increased expression of Numb, suggesting a potential mechanism by which nandrolone reduces denervation-atrophy.
Collapse
Affiliation(s)
- Xin-Hua Liu
- Center of Excellence for Medical Consequences of Spinal Cord Injury, James J. Peter VA Medical Center, Bronx, NY 10468, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
The development of satellite cells and their niche in striated muscle complex of anorectal malformations rat embryos. J Surg Res 2011; 168:e71-80. [PMID: 21435657 DOI: 10.1016/j.jss.2011.01.035] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2010] [Revised: 01/09/2011] [Accepted: 01/19/2011] [Indexed: 11/23/2022]
Abstract
BACKGROUND It has been demonstrated that different degrees of pelvic floor muscle (PFM) maldevelop in anorectal malformations (ARMs); yet the development of satellite cells, the myogenic stem cells responsible for muscle growth, repair, and maintenance remains elusive during the embryogenesis of PFM. Striated muscle complex (SMC) is one of the most important components of PFM. The objective of this study was to observe the development pattern of satellite cells and their niche of SMC and investigate its possible role in PFM dysplasia in ARMs. METHODS Immunohistochemistry, cell culture, transmission electron microscopy (TEM), real-time quantitative PCR, and Western blot were performed to trace the dynamic development pattern of satellite cells during the morphogenesis of PFM in ethylenethiourea (ETU)-induced ARMs rat embryos. RESULTS In ARMs rat embryos, earlier presentation and higher number of Pax7-expressing cell were observed in SMC. The expression of Pax7 and vimentin were up-regulated, while the expression of myogenin, vWF, and neurofilament were down-regulated. Ultrastructure analysis of SMC was characterized by increased amount of nuclear heterochromatin of satellite cell nuclei, thickened basal lamina, widened gap between satellite cell and myofiber, and disarrangement of muscle fibers. The satellite cells demonstrated abnormal differentiation after they were isolated and cultured in vitro. CONCLUSIONS Our results suggest that premature origination of satellite cell from myogenic progenitor or precursor may result in the depletion of myogenic precursor and cessation of muscle growth; intrinsic defect in satellite cell structure, and extrinsic impairment of microenvironment compromised the myogenic competence of satellite cell, which might contribute substantially to the hypoplastic SMC in ARMs.
Collapse
|
28
|
Boldrin L, Muntoni F, Morgan JE. Are human and mouse satellite cells really the same? J Histochem Cytochem 2010; 58:941-55. [PMID: 20644208 DOI: 10.1369/jhc.2010.956201] [Citation(s) in RCA: 96] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Satellite cells are quiescent cells located under the basal lamina of skeletal muscle fibers that contribute to muscle growth, maintenance, repair, and regeneration. Mouse satellite cells have been shown to be muscle stem cells that are able to regenerate muscle fibers and self-renew. As human skeletal muscle is also able to regenerate following injury, we assume that the human satellite cell is, like its murine equivalent, a muscle stem cell. In this review, we compare human and mouse satellite cells and highlight their similarities and differences. We discuss gaps in our knowledge of human satellite cells, compared with that of mouse satellite cells, and suggest ways in which we may advance studies on human satellite cells, particularly by finding new markers and attempting to re-create the human satellite cell niche in vitro.
Collapse
Affiliation(s)
- Luisa Boldrin
- Dubowitz Neuromuscular Centre, UCL Institute of Child Health, 30 Guilford Street, London WC1N1EH, United Kingdom.
| | | | | |
Collapse
|
29
|
Jang YC, Lustgarten MS, Liu Y, Muller FL, Bhattacharya A, Liang H, Salmon AB, Brooks SV, Larkin L, Hayworth CR, Richardson A, Van Remmen H. Increased superoxide in vivo accelerates age-associated muscle atrophy through mitochondrial dysfunction and neuromuscular junction degeneration. FASEB J 2010; 24:1376-90. [PMID: 20040516 PMCID: PMC2987499 DOI: 10.1096/fj.09-146308] [Citation(s) in RCA: 227] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2009] [Accepted: 12/03/2009] [Indexed: 11/11/2022]
Abstract
Oxidative stress has been implicated in the etiology of age-related muscle loss (sarcopenia). However, the underlying mechanisms by which oxidative stress contributes to sarcopenia have not been thoroughly investigated. To directly examine the role of chronic oxidative stress in vivo, we used a mouse model that lacks the antioxidant enzyme CuZnSOD (Sod1). Sod1(-/-) mice are characterized by high levels of oxidative damage and an acceleration of sarcopenia. In the present study, we demonstrate that muscle atrophy in Sod1(-/-) mice is accompanied by a progressive decline in mitochondrial bioenergetic function and an elevation of mitochondrial generation of reactive oxygen species. In addition, Sod1(-/-) muscle exhibits a more rapid induction of mitochondrial-mediated apoptosis and loss of myonuclei. Furthermore, aged Sod1(-/-) mice show a striking increase in muscle mitochondrial content near the neuromuscular junctions (NMJs). Despite the increase in content, the function of mitochondria is significantly impaired, with increased denervated NMJs and fragmentation of acetylcholine receptors. As a consequence, contractile force in aged Sod1(-/-) muscles is greatly diminished. Collectively, we show that Sod1(-/-) mice display characteristics of normal aging muscle in an accelerated manner and propose that the superoxide-induced NMJ degeneration and mitochondrial dysfunction are potential mechanisms of sarcopenia.
Collapse
Affiliation(s)
- Youngmok C Jang
- Department of Cellular and Structural Biology, University of Texas Health Science Center at San Antonio, San Antonio, TX 78245, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Ohira Y, Kawano F, Dong Wang X, Nakai N, Ohira T, Okabe H, Naito H, Goto K. Role(s) of Mechanical Load and Satellite Cells in The Regulation of The Size of Soleus Muscle Fiber in Rats. ACTA ACUST UNITED AC 2010. [DOI: 10.2187/bss.24.135] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
31
|
Plant PJ, Bain JR, Correa JE, Woo M, Batt J. Absence of caspase-3 protects against denervation-induced skeletal muscle atrophy. J Appl Physiol (1985) 2009; 107:224-34. [PMID: 19390003 DOI: 10.1152/japplphysiol.90932.2008] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The ubiquitin-proteasome system is a key proteolytic pathway activated during skeletal muscle atrophy. The proteasome, however, cannot degrade intact myofibrils or actinomyosin complexes. In rodent models of diabetes mellitus and uremia, caspase-3 is involved in actinomyosin cleavage, generating fragments that subsequently undergo ubiquitin-proteasome-mediated degradation. Here, we demonstrate that caspase-3 also mediates denervation-induced muscle atrophy. At 2 wk after tibial nerve transection, the denervated gastrocnemius of caspase-3-knockout mice weighed more and demonstrated larger fiber-type-specific cross-sectional area than the denervated gastrocnemius of wild-type mice. However, there was no difference between caspase-3-knockout and wild-type denervated muscles in the magnitude or pattern of actinomyosin degradation, as determined by Western blotting for actin and the 14-kDa actin fragment. Similarly, there was no difference between caspase-3-knockout and wild-type denervated muscles in the magnitude of increase in proteasome activity, total protein ubiquitination, or atrogin-1 and muscle-specific ring finger protein 1 transcript levels. In contrast, there was an increase in TdT-mediated dUTP nick end label-positive nuclei in the denervated muscle of wild-type compared with caspase-3-knockout mice. Apoptotic signaling upstream of caspase-3 remained intact, with equivalent mitochondrial Bax translocation and cytochrome c release and caspase-9 activation in the denervated gastrocnemius muscle of wild-type and caspase-3-knockout mice. In contrast, diminished poly(ADP-ribose) polymerase cleavage in the denervated muscle of caspase-3-knockout compared with wild-type mice revealed that apoptotic signaling downstream of caspase-3 was impaired, suggesting that the absence of caspase-3 protects against denervation-induced muscle atrophy by suppressing apoptosis as opposed to ubiquitin-proteasome-mediated protein degradation.
Collapse
|
32
|
Rhoads RP, Fernyhough ME, Liu X, McFarland DC, Velleman SG, Hausman GJ, Dodson MV. Extrinsic regulation of domestic animal-derived myogenic satellite cells II. Domest Anim Endocrinol 2009; 36:111-26. [PMID: 19261429 DOI: 10.1016/j.domaniend.2008.12.005] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2008] [Revised: 12/18/2008] [Accepted: 12/19/2008] [Indexed: 12/12/2022]
Abstract
The existence of myogenic satellite cells was reported some 47 years ago, and, since that time, satellite cell research has flourished. So much new information is generated (daily) on these cells that it can be difficult for individuals to keep abreast of important issues related to their activation and proliferation, the modulation of the activity of other cell types, the differentiation of the cells to facilitate normal skeletal muscle growth and development, or to the repair of damaged myofibers. The intent of this review is to summarize new information about the extrinsic regulation of myogenic satellite cells and to provide specific mechanisms involved in altering satellite cell physiology. Where possible, examples from agriculturally important animals are used for illustrative purposes.
Collapse
Affiliation(s)
- R P Rhoads
- Department of Animal Sciences, University of Arizona, Tucson, AZ 85719, USA
| | | | | | | | | | | | | |
Collapse
|
33
|
The effect of muscle loading on skeletal muscle regenerative potential: an update of current research findings relating to aging and neuromuscular pathology. Am J Phys Med Rehabil 2009; 88:145-55. [PMID: 19169178 DOI: 10.1097/phm.0b013e3181951fc5] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Skeletal muscle is a dynamic tissue with a remarkable ability to continuously respond to environmental stimuli. Among its adaptive responses is the widely investigated ability of skeletal muscle to regenerate after loading or injury or both. Although significant basic science efforts have been dedicated to better understand the underlying mechanism controlling skeletal muscle regeneration, there has been relatively little impact in the clinical approaches used to treat skeletal muscle injuries and wasting. The purpose of this review article is to provide an overview of the basic biology of satellite cell function in response to muscle loading and to relate these findings in the context of aging and neuromuscular pathology for the rehabilitation medicine specialist.
Collapse
|
34
|
Abstract
Apoptosis has been implicated in mediating denervation-induced muscle wasting. In this study we determined the effect of interference of apoptosis on muscle wasting during denervation by using mice genetically deficient in pro-apoptotic Bax. After denervation, muscle wasting was evident in both wild-type and Bax(-/-) muscles but reduction of muscle weight was attenuated in Bax(-/-) mice. Apoptotic DNA fragmentation increased in wild-type denervated muscles whereas there was no statistical increase in DNA fragmentation in denervated muscles from Bax(-/-) mice. Mitochondrial AIF and Smac/DIABLO releases and Bcl-2, p53 and HSP27 increased whereas XIAP and MnSOD decreased to a similar extent in muscles from wild-type and Bax(-/-) mice following denervation. Mitochondrial cytochrome c release was elevated in denervated muscles from wild-type mice but the increase was suppressed in muscles from Bax(-/-) mice. Increases in caspase-3 and -9 activities and oxidative stress markers H(2)O(2), MDA/4-HAE and nitrotyrosine were all evident in denervated muscles from wild-type mice but these changes were absent in muscles from Bax(-/-) mice. Moreover, ARC increased exclusively in denervated Bax(-/-) muscle. Our data indicate that under conditions of denervation, pro-apoptotic signalling is suppressed and muscle wasting is attenuated when the Bax gene is lacking. These findings suggest that interventions targeting apoptosis may be valuable in ameliorating denervation-associated pathologic muscle wasting in certain neuromuscular disorders that involve partial or full denervation.
Collapse
Affiliation(s)
- P M Siu
- Laboratory of Muscle Biology and Sarcopenia, Division of Exercise Physiology, West Virginia University School of Medicine, Morgantown, West Virginia 26506, USA
| | | |
Collapse
|
35
|
Krajnak K, Waugh S, Miller R, Baker B, Geronilla K, Alway SE, Cutlip RG. Proapoptotic factor Bax is increased in satellite cells in the tibialis anterior muscles of old rats. Muscle Nerve 2007; 34:720-30. [PMID: 16967487 DOI: 10.1002/mus.20656] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Aging impairs the ability of muscle to adapt to exercise or injury. The goal of this study was to determine whether age-related changes in muscle adaptability could be the result of satellite cell apoptosis. Ten days after exposure to an injury protocol, estimates of edema in the exposed tibialis anterior muscles were higher in old (30 months) than young (3 months) rats, and isometric force levels were lower in old rats. Both young and old rats displayed an increase in MyoD labeling in the exposed muscle, indicating that injury induced satellite-cell activation. However, there were more MyoD-labeled cells that coexpressed the proapoptotic factor, Bax, in old than in young rats, suggesting that decrements in muscle recovery may be associated with an increase in satellite-cell apoptosis. Based on these findings we conclude that reducing satellite-cell apoptosis in aged animals may improve muscle recovery after injury.
Collapse
MESH Headings
- Adaptation, Physiological
- Age Factors
- Aging/genetics
- Aging/metabolism
- Aging/pathology
- Animals
- Apoptosis
- Exercise Test
- Immunohistochemistry
- Isometric Contraction/genetics
- Isometric Contraction/physiology
- Male
- Muscle, Skeletal/cytology
- Muscle, Skeletal/injuries
- Muscle, Skeletal/physiology
- MyoD Protein/analysis
- MyoD Protein/metabolism
- Organ Size
- RNA, Messenger/analysis
- Rats
- Rats, Inbred Strains
- Regeneration
- Reverse Transcriptase Polymerase Chain Reaction
- Satellite Cells, Skeletal Muscle/cytology
- Satellite Cells, Skeletal Muscle/metabolism
- Satellite Cells, Skeletal Muscle/physiology
- Transcription, Genetic
- Up-Regulation
- bcl-2-Associated X Protein/analysis
- bcl-2-Associated X Protein/genetics
- bcl-2-Associated X Protein/metabolism
Collapse
Affiliation(s)
- Kristine Krajnak
- Engineering and Controls Technology Branch, National Institute for Occupational Safety and Health, 1095 Willowdale Rd., MS 2027, Morgantown, West Virginia 26505, USA.
| | | | | | | | | | | | | |
Collapse
|
36
|
Jejurikar SS, Henkelman EA, Cederna PS, Marcelo CL, Urbanchek MG, Kuzon WM. Aging increases the susceptibility of skeletal muscle derived satellite cells to apoptosis. Exp Gerontol 2006; 41:828-36. [PMID: 16942852 DOI: 10.1016/j.exger.2006.06.053] [Citation(s) in RCA: 86] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2006] [Revised: 06/07/2006] [Accepted: 06/13/2006] [Indexed: 11/23/2022]
Abstract
The mechanisms causing the impaired regenerative response to injury observed in skeletal muscle of old animals are unknown. Satellite cells, stem cell descendants within adult skeletal muscle, are the primary source of regenerating muscle fibers. Apoptosis may be a mechanism responsible for the depletion of satellite cells in old animals. This work tested the hypothesis that aging increases the susceptibility of satellite cells to apoptosis. Satellite cells were cultured from the extensor digitorum longus muscles of young (3-month-old), adult (9-month-old), and old (31-month-old) Brown Norway rats. Satellite cells were treated for 24h with the pro-apoptotic agents TNF-alpha (20 ng/mL) and Actinomycin D (250 ng/mL). Immunostaining for activated caspases and terminal deoxynucleotydil transferase-mediated dutp nick-end labeling (TUNEL) was performed to identify apoptotic satellite cells. Quantity of the anti-apoptotic protein bcl-2 was determined by Western blot analysis. Satellite cells from old animals demonstrated significantly higher percentages of cells with activated caspases and TUNEL-positive cells, and significantly lower amounts of bcl-2 compared to young and adult animals. These data support the hypothesis that aging increases satellite cell susceptibility to apoptosis. In old muscle, apoptosis may play a causative role in the depletion of satellite cells, impairing the regenerative response to injury.
Collapse
Affiliation(s)
- Sameer S Jejurikar
- Section of Plastic and Reconstructive Surgery, Department of Surgery, University of Michigan Health Systems, Ann Arbor, MI, USA
| | | | | | | | | | | |
Collapse
|
37
|
Dupont-Versteegden EE, Strotman BA, Gurley CM, Gaddy D, Knox M, Fluckey JD, Peterson CA. Nuclear translocation of EndoG at the initiation of disuse muscle atrophy and apoptosis is specific to myonuclei. Am J Physiol Regul Integr Comp Physiol 2006; 291:R1730-40. [PMID: 16873557 DOI: 10.1152/ajpregu.00176.2006] [Citation(s) in RCA: 93] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Skeletal muscle atrophy is associated with an increase in apoptosis, and we showed previously that endonuclease G (EndoG) is localized to nuclei following unloading. The goal of this study was to determine whether the onset of apoptosis in response to disuse was consistent with the hypothesis that EndoG is involved in myofiber nuclear loss. Atrophy was induced by hindlimb suspension for 12 h or 1, 2, 4 and 7 days in 6-mo-old rats. Soleus myofiber cross-sectional area decreased significantly by 2 days, whereas muscle mass and muscle-to-body mass ratio decreased by 4 and 7 days, respectively. By contrast, a significant increase in apoptosis, evidenced by terminal deoxynucleotidyl transferase-mediated dUTP nick-end labeling (TUNEL)-positive nuclei, occurred as early as 12 h after suspension, preceding the elevation in muscle atrophy F-box gene expression. The early increase in apoptosis appeared to be specific to myofiber nuclei, whereas TUNEL-positive interstitial cells did not become significantly elevated until 2 days after suspension. Furthermore, TUNEL-positive myofiber nuclei colocalized with EndoG as early as 12 h after suspension, and no such localization was observed in interstitial cells. Although no significant change in total activated caspase-3, -7, or -12 protein abundance was apparent, activated caspase-3 was expressed in interstitial cells undergoing apoptosis, some of which were endothelial cells. These data indicate that apoptosis is an early, and therefore possibly causative, event in the process of muscle atrophy, and that EndoG nuclear translocation is specific for myofiber nuclear apoptosis, whereas interstitial cells may undergo apoptosis via a more classical, caspase-dependent pathway.
Collapse
|
38
|
Sun H, Liu J, Ding F, Wang X, Liu M, Gu X. Investigation of differentially expressed proteins in rat gastrocnemius muscle during denervation-reinnervation. J Muscle Res Cell Motil 2006; 27:241-50. [PMID: 16752196 DOI: 10.1007/s10974-006-9067-4] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2006] [Accepted: 04/07/2006] [Indexed: 01/22/2023]
Abstract
To have a better insight into the molecular events involved in denervation-induced atrophy and reinnervation-induced regeneration of skeletal muscles, it is important to investigate the changes in expression levels of a great multitude of muscle proteins during the process of denervation-reinnervation. In this study, we employed an experimental model of rat sciatic nerve crush to examine the differentially expressed proteins in the rat gastrocnemius muscle at different time points (0, 1, 2, 3, 4 weeks) after sciatic nerve crush by using two-dimensional gel electrophoresis (2-DE) followed by matrix-assisted laser desorption/ionization-time-of-flight mass spectrometry (MALDI-TOF-MS), collectively referred to as the modern proteomic analysis. The results showed that 16 proteins in the rat gastrocnemius muscle exhibited two distinct types of change pattern in their relative abundance: (1) The relative expression levels of 11 proteins (including alpha actin, myosin heavy chain, etc.) were decreased either within 1 or 2 weeks post-sciatic nerve injury, followed by restoration during the ensuing days until 4 weeks. (2) The other 5 proteins (including alpha enolase, beta enolase, signal peptide peptidase-like 3, etc.) displayed an up-regulation in their relative expression levels within 1 week following sciatic nerve injury, and a subsequent gradual decrease in their relative expression levels until 4 weeks. Moreover, the significance of the changes in expression levels of the 16 proteins during denervation-reinnervation has been selectively discussed.
Collapse
Affiliation(s)
- Hualin Sun
- The Jiangsu Key Laboratory of Neuroregeneration, Nantong University, 19 Qi-Xiu Road, Nantong, Jiangsu Province 226001, P.R. China
| | | | | | | | | | | |
Collapse
|
39
|
Ferreira R, Neuparth MJ, Ascensão A, Magalhães J, Vitorino R, Duarte JA, Amado F. Skeletal muscle atrophy increases cell proliferation in mice gastrocnemius during the first week of hindlimb suspension. Eur J Appl Physiol 2006; 97:340-6. [PMID: 16770471 DOI: 10.1007/s00421-006-0197-6] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/23/2006] [Indexed: 11/28/2022]
Abstract
The comprehension of the cellular mechanisms underlying skeletal muscle atrophy has been the aim of several experimental studies. However, the majority of them focused on alterations of the myocytes induced by different experimental conditions yet disregarding the contribution of other cells such as endothelial cells and fibroblasts. In this sense, 70 Charles River CD1 male mice were randomly assigned to seven groups (n=10 per group): control and 6, 12, 24, 48, 72 h and 1 week with respect to the period of hindlimb suspension. Forty-eight hours before sacrifice, the animals were injected with bromodeoxyuridine (BrdU) in order to identify proliferating cells. Immunohistochemistry and south-western blotting techniques were used to evaluate across the whole gastrocnemius muscle BrdU incorporation into the different proliferating cells. The contribution of the apoptotic response was also measured in order to ascertain whether the balance between cell survival and death was preserved. The results observed during 1 week of unloading-induced atrophy evidenced an intense peak of proliferating activity only after 6 h, mainly due to the duplication of satellite cells. Consequently to this unexpected activation of satellite cells, the addition of nuclei to the fibre syncytium was recognized at 12 h of unloading. After 48 h of weightlessness, the proliferating activity observed was largely due to an interstitial fibrosis. According to the apoptotic index profile observed during the analysed unloading period, this general proliferative activity was balanced by apoptosis, which strongly suggests the existence of a regulatory feedback response between anabolic and catabolic events in unloading-induced skeletal muscle atrophy.
Collapse
Affiliation(s)
- Rita Ferreira
- CIAFEL, Laboratory of Sport Biochemistry, Faculty of Sports Sciences, University of Porto, Rua Dr. Plácido Costa, 91, 4200, Porto, Portugal.
| | | | | | | | | | | | | |
Collapse
|
40
|
Wang XD, Kawano F, Matsuoka Y, Fukunaga K, Terada M, Sudoh M, Ishihara A, Ohira Y. Mechanical load-dependent regulation of satellite cell and fiber size in rat soleus muscle. Am J Physiol Cell Physiol 2006; 290:C981-9. [PMID: 16291821 DOI: 10.1152/ajpcell.00298.2005] [Citation(s) in RCA: 70] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The effects of mechanical unloading and reloading on the properties of rat soleus muscle fibers were investigated in male Wistar Hannover rats. Satellite cells in the fibers of control rats were distributed evenly throughout the fiber length. After 16 days of hindlimb unloading, the number of satellite cells in the central, but not the proximal or distal, region of the fiber was decreased. The number of satellite cells in the central region gradually increased during the 16-day period of reloading. The mean sarcomere length in the central region of the fibers was passively shortened during unloading due to the plantarflexed position at the ankle joint: sarcomere length was maintained at <2.1 μm, which is a critical length for tension development. Myonuclear number and domain size, fiber cross-sectional area, and the total number of mitotically active and quiescent satellite cells of whole muscle fibers were lower than control fibers after 16 days of unloading. These values then returned to control values after 16 days of reloading. These results suggest that satellite cells play an important role in the regulation of muscle fiber properties. The data also indicate that the satellite cell-related regulation of muscle fiber properties is dependent on the level of mechanical loading, which, in turn, is influenced by the mean sarcomere length. However, it is still unclear why the region-specific responses, which were obvious in satellite cells, were not induced in myonuclear number and fiber cross-sectional area.
Collapse
Affiliation(s)
- X D Wang
- Graduate School of Medicine, Osaka University, Suita City, Osaka, Japan
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Kauhanen SC, Ylä-Kotola TM, Leivo IV, Tukiainen E, Asko-Seljavaara SL. Long-term adaptation of human microneurovascular muscle flaps to the paralyzed face: An immunohistochemical study. Microsurgery 2006; 26:557-65. [PMID: 17066408 DOI: 10.1002/micr.20288] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The purpose of this study was to characterize microneurovascular (MNV) muscle transplants immunohistochemically up to 10 years after transfer. Histological data was related to long-term functional outcome. The study comprised 17 patients with a mean age of 41 years suffering from complete unilateral long-lasting facial paralysis. A two-stage procedure was performed between 1986 and 2001. The gracilis, latissimus dorsi, and serratus muscles were used in four, eight, and five patients, respectively. Eighteen biopsy samples were taken from MNV muscle grafts during secondary refinement procedures. In one patient, the tissue samples were collected at two different time points. Immunohistochemistry testing revealed muscle fiber type distribution (anti-myosin fast), proliferating satellite cells (Ki-67), and reinnervation (S-100). Muscle atrophy was assessed histomorphometrically. In a recent study, patient characteristics and the function of the flap were evaluated. Histological data were compared with clinical data and long-term functional outcomes of the patients. In biopsy samples taken 1-10 (mean 31 months) years after MNV muscle transfer, the mean muscle fiber diameter was 38 (range 14-70) microm, indicating a 40% decrease compared with control values. Muscle atrophy was not type-specific and the mean percentage of type II fibers was not altered. Individual variation was, however, considerable. Proliferative activity of satellite cells was seen in 60% of the samples but it tended to decline with an increase in follow-up time. All samples showed intramuscular reinnervation. In statistical analysis severe atrophy correlated with prolonged intraoperative ischemia (P=0.04). The good long-term functional outcome correlated with dominance of fast fibers in muscle grafts (P=0.03). Atrophy tended to be more pronounced in the serratus than in the other muscles (ns). In summary, despite dense muscle reinnervation, morphology of the muscle is not fully restored after muscle transfer. Ischemia time affects muscle morphology. Adaptation of the graft to fast-twitch muscle activity favors better mimic function. The proliferative activity of satellite cells declines with prolonged follow-up time.
Collapse
Affiliation(s)
- Susanna C Kauhanen
- Department of Plastic Surgery, Helsinki University Hospital, Helsinki, Finland.
| | | | | | | | | |
Collapse
|
42
|
Batt J, Bain J, Goncalves J, Michalski B, Plant P, Fahnestock M, Woodgett J. Differential gene expression profiling of short and long term denervated muscle. FASEB J 2005; 20:115-7. [PMID: 16291642 DOI: 10.1096/fj.04-3640fje] [Citation(s) in RCA: 98] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Skeletal muscle function and viability are dependent upon intact innervation. Peripheral nerve injury and muscle denervation cause muscle atrophy. Time to re-innervation is one of the most important determinants of functional outcome. While short-term denervation can result in nearly fully reversible changes in muscle mass, prolonged denervation leads to irreversible muscle impairment from profound atrophy, myocyte death and fibrosis. We performed transcriptional profiling to identify genes that were altered in expression in short-term (1 month) and long-term (3 month) denervated muscle and validated the microarray data by RT-PCR and Western blotting. Genes controlling cell death, metabolism, proteolysis, stress responses and protein synthesis/translation were altered in expression in the denervated muscle. A differential pattern of expression of genes encoding cell cycle regulators and extracellular matrix components was identified that correlated with the development of irreversible post-denervation changes. Genes encoding mediators of protein degradation were differentially expressed between 1 and 3 month denervated muscle suggesting different signaling networks are recruited over time to induce and maintain muscle atrophy. Understanding of the timing and type of pathological processes that are triggered by denervation may allow the design of interventions that delay or protect muscle from loss of nerve function.
Collapse
Affiliation(s)
- Jane Batt
- Clinical Sciences Division, Department of Medicine, University of Toronto, Toronto, Ontario, Canada.
| | | | | | | | | | | | | |
Collapse
|
43
|
Bartoli M, Richard I. Calpains in muscle wasting. Int J Biochem Cell Biol 2005; 37:2115-33. [PMID: 16125114 DOI: 10.1016/j.biocel.2004.12.012] [Citation(s) in RCA: 127] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2004] [Revised: 12/01/2004] [Accepted: 12/31/2004] [Indexed: 01/09/2023]
Abstract
Calpains are intracellular nonlysosomal Ca(2+)-regulated cysteine proteases. They mediate regulatory cleavages of specific substrates in a large number of processes during the differentiation, life and death of the cell. The purpose of this review is to synthesize our current understanding of the participation of calpains in muscle atrophy. Muscle tissue expresses mainly three different calpains: the ubiquitous calpains and calpain 3. The participation of the ubiquitous calpains in the initial degradation of myofibrillar proteins occurring in muscle atrophy as well as in the necrosis process accompanying muscular dystrophies has been well characterized. Inactivating mutations in the calpain 3 gene are responsible for limb-girdle muscular dystrophy type 2A and calpain 3 has been found to be downregulated in different atrophic situations, suggesting that it has to be absent for the atrophy to occur. The fact that similar regulations of calpain activities occur during exercise as well as in atrophy led us to propose that the calpains control cytoskeletal modifications needed for muscle plasticity.
Collapse
Affiliation(s)
- Marc Bartoli
- Généthon, Centre National de la Recherche Scientifique UMR 8115, 1 bis rue de l'Internationale, 91000 Evry, France
| | | |
Collapse
|
44
|
Siu PM, Pistilli EE, Alway SE. Apoptotic responses to hindlimb suspension in gastrocnemius muscles from young adult and aged rats. Am J Physiol Regul Integr Comp Physiol 2005; 289:R1015-26. [PMID: 15919734 DOI: 10.1152/ajpregu.00198.2005] [Citation(s) in RCA: 117] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Although apoptosis has been demonstrated in soleus during hindlimb suspension (HS), it is not known whether apoptosis is also involved in the loss of muscles dominated by mixed fibers. Therefore, we examined the apoptotic responses in gastrocnemius muscles of young adult and aged Fischer 344 x Brown Norway rats after 14 days of HS. The medial gastrocnemius muscle wet weight significantly decreased by 30 and 32%, and muscle wet weight normalized to the animal body weight decreased by 11 and 15% in young adult and aged animals, respectively, after HS. The extent of apoptotic DNA fragmentation increased by 119 and 61% in suspended muscles from young and aged rats, respectively. Bax mRNA increased by 73% in young muscles after HS. Bax and Bcl-2 protein levels were greater in suspended muscles relative to control muscles in both age groups. The level of cytosolic mitochondria-housed apoptotic factor cytochrome c was significantly increased in the mitochondria-free cytosol of suspended muscles from young and aged rats. In contrast, the release/accumulation of AIF, a caspase-independent apoptogenic factor, was exclusively expressed in the suspended muscles from aged rats. Our data also show that aging favors the proapoptotic signaling in skeletal muscle by altering the contents of Bax, Bcl-2, Apaf-1, AIF, caspases, XIAP, Smac/DIABLO, and cytochrome c. Furthermore, these results indicate that apoptosis occurs not only in slow-twitch soleus muscle but also in the mixed-fiber (predominately fast fibered) gastrocnemius muscle. Our data are consistent with the hypothesis that apoptotic signaling differs in young adult and aged gastrocnemius muscles during HS.
Collapse
Affiliation(s)
- Parco M Siu
- Division of Exercise Physiology, School of Medicine, Robert C. Byrd Health Sciences Center, West Virginia University, Morgantown WV 26506-9227, USA
| | | | | |
Collapse
|
45
|
Siu PM, Alway SE. Id2 and p53 participate in apoptosis during unloading-induced muscle atrophy. Am J Physiol Cell Physiol 2005; 288:C1058-73. [PMID: 15601750 DOI: 10.1152/ajpcell.00495.2004] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Apoptotic signaling was examined in the patagialis (PAT) muscles of young adult and old quail. One wing was loaded for 14 days to induce hypertrophy and then unloaded for 7 or 14 days to induce muscle atrophy. Although the nuclear Id2 protein content was not different between unloaded and control muscles in either age group, cytoplasmic Id2 protein content of unloaded muscles was higher than that in contralateral control muscles after 7 days of unloading in young quails. Nuclear and cytoplasmic p53 contents and the p53 nuclear index of the unloaded muscles were higher than those in control muscles after 7 days of unloading in young quails, whereas in aged quails, the p53 and Id2 contents and p53 nuclear index of the unloaded muscles were not altered by unloading. Immunofluorescent staining indicated that myonuclei and activated satellite cell nuclei contributed to the increased number of p53-positive nuclei. Conversely, unloading in either young adult or aged PAT muscles did not alter c-Myc protein content. Although Cu-Zn-SOD content was not different in unloaded and control muscles, Mn-SOD content increased in PAT muscles after 7 days of unloading in young quails, suggesting that unloading induced an oxidative disturbance in these muscles. Moderate correlational relationships existed among Id2, p53, c-Myc, SOD, apoptosis-regulatory factors, and TdT-mediated dUTP nick end labeling index. These data indicate that Id2 and p53 are involved in the apoptotic responses during unloading-induced muscle atrophy after hypertrophy in young adult birds. Furthermore, our data suggest that there is an aging-dependent regulation of Id2 and p53 during unloading of previously hypertrophied muscles.
Collapse
Affiliation(s)
- Parco M Siu
- Division of Exercise Physiology, West Virginia Univ. School of Medicine, Robert C. Byrd Health Science Center, Morgantown, WV 26506-9227, USA
| | | |
Collapse
|
46
|
Abstract
Apoptosis has been implicated in the regulation of denervation-induced muscle atrophy. However, the activation of apoptotic signal transduction during muscle denervation has not been fully elucidated. The present study examined the apoptotic responses to denervation in rat gastrocnemius muscle. Following 14 days of denervation, the extent of apoptotic DNA fragmentation as determined by a cytosolic nucleosome ELISA was increased by 100% in the gastrocnemius muscle. RT-PCR and immunoblot analyses indicated that Bax was dramatically upregulated while Bcl-2 was modestly increased; however, the Bax/Bcl-2 ratio was significantly increased in denervated muscles relative to control muscles. Analyses of ELISA and immunoblots from mitochondria-free cytosol extracts showed a significant increase in mitochondria-associated apoptotic factors, including cytochrome c, Smac/DIABLO and apoptosis-inducing factor (AIF). In addition to the upregulation of caspase-3 and -9 mRNA, pro-/cleaved caspase protein and proteolytic activity levels, the X-linked inhibitor of apoptosis (XIAP) protein level was downregulated. The cleaved product of poly(ADP-ribose) polymerase (PARP) was detected in muscle samples following denervation. Although we did not find a difference in the inhibitor of DNA binding/differentiation-2 (Id2) and c-Myc protein contents between the denervated and control muscles, the protein content of tumour suppressor p53 was significantly increased in both the nuclear and the cytosolic fractions with denervation. Moreover, denervation increased the protein content of HSP70, whereas the MnSOD (a mitochondrial isoform of superoxide dismutase) protein content was diminished, which indicated that denervation might have induced cellular and/or oxidative stress. Our data show that mitochondria-associated apoptotic signalling is upregulated during muscle denervation. We interpret these findings to indicate that apoptosis has a physiologically important role in regulating denervation-induced muscle atrophy.
Collapse
Affiliation(s)
- Parco M Siu
- Laboratory of Muscle Biology an Sarcopenia, Division of Exercise Physiology, West Virginia University School of Medicine, Morgantown WV 26506-9227, USA
| | | |
Collapse
|
47
|
Abstract
Muscle-fiber loss is a characteristic of many progressive neuromuscular disorders. Over the past decade, identification of a growing number of apoptosis-associated factors and events in pathological skeletal muscle provided increasing evidence that apoptotic cell-death mechanisms account significantly for muscle-fiber atrophy and loss in a wide spectrum of neuromuscular disorders. It became obvious that there is not one specific pathway for muscle fibers to undergo apoptotic degradation. In contrast, certain neuromuscular diseases seem to involve characteristic expression patterns of apoptosis-related factors and pathways. Furthermore, there are some characteristics of muscle-fiber apoptosis that rely on the muscle fiber itself as an extremely specified cell type. Multinucleated muscle fibers with successive muscle-fiber segments controlled by individual nuclei display some specifics different from apoptosis of mononucleated cells. This review focuses on the expression patterns of apoptosis-associated factors in different primary and secondary neuromuscular disorders and gives a synopsis of current knowledge.
Collapse
Affiliation(s)
- Dominique S Tews
- Edinger-Institute, Johann Wolfgang Goethe University Hospital, Deutschordenstrasse 46, D-60528 Frankfurt am Main, Germany.
| |
Collapse
|
48
|
Tews DS, Behrhof W, Schindler S. Expression patterns of initiator and effector caspases in denervated human skeletal muscle. Muscle Nerve 2005; 31:175-81. [PMID: 15625686 DOI: 10.1002/mus.20253] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
There is evidence that apoptotic cell death contributes to the loss of denervated muscle fibers. In 17 patients with neurogenic muscular atrophy, we studied the expression of the apoptosis mediators APAF-1/caspase-9 and degrading caspases-2, -3, and -7 by immunohistochemical and western blot analyses. Muscle with neurogenic atrophy showed distinct upregulation of caspase-9 and -7 and no expression for APAF-1 (apoptosis protease-activating factor-1) and caspase-2 and -3. Expression of caspase-7 was restricted to atrophic fibers, but caspase-9 was also found in normal-sized muscle fibers where its expression was often confined to single fiber segments. These findings indicate that upregulated expression of caspase-9 can initiate the proteolytic cascade involving the downstream executioner caspase-7, which mediates degradation of denervated muscle fibers. However, apoptotic events may be restricted to single muscle-fiber segments, where apoptotic cell degradation contributes to the long-term process of atrophy. Pharmacological inhibition of caspases may be a therapeutic strategy in diminishing muscle atrophy.
Collapse
Affiliation(s)
- Dominique S Tews
- Neurological (Edinger) Institute, Johann Wolfgang Goethe University Hospital, Deutschordenstrasse 46, D-60528 Frankfurt am Main, Germany.
| | | | | |
Collapse
|
49
|
Mitchell PO, Pavlath GK. Skeletal muscle atrophy leads to loss and dysfunction of muscle precursor cells. Am J Physiol Cell Physiol 2004; 287:C1753-62. [PMID: 15329339 DOI: 10.1152/ajpcell.00292.2004] [Citation(s) in RCA: 91] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Atrophy of skeletal muscle leads to decreases in myofiber size and nuclear number; however, the effects of atrophic conditions on muscle precursor cells (MPC) are largely unknown. MPC lie outside myofibers and represent the main source of additional myonuclei necessary for muscle growth and repair. In the present study, we examined the properties of MPC after hindlimb suspension (HS)-induced atrophy and subsequent recovery of the mouse hindlimb muscles. We demonstrated that the number of MPC in atrophied muscles was decreased. RT-PCR analysis of cells isolated from atrophied muscles indicated that several mRNA characteristic of the myogenic program in MPC were absent. Cells isolated from atrophied muscles failed to properly proliferate and undergo differentiation into multinucleated myotubes. Thus atrophy led to a decrease in MPC and caused dysfunction in those MPC that remained. Upon regrowth of the atrophied muscles, these deleterious effects were reversed. Our data suggest that preventing loss or dysfunction of MPC may be a new pharmacological target during muscle atrophy.
Collapse
Affiliation(s)
- Patrick O Mitchell
- Department of Pharmacology, Emory University School of Medicine, 5024 O. W. Rollins Research Center, Atlanta, GA 30322, USA
| | | |
Collapse
|
50
|
Siu PM, Pistilli EE, Butler DC, Alway SE. Aging influences cellular and molecular responses of apoptosis to skeletal muscle unloading. Am J Physiol Cell Physiol 2004; 288:C338-49. [PMID: 15483226 DOI: 10.1152/ajpcell.00239.2004] [Citation(s) in RCA: 106] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
The influence of aging on skeletal myocyte apoptosis is not well understood. In this study we examined apoptosis and apoptotic regulatory factor responses to muscle atrophy induced via limb unloading following loading-induced hypertrophy. Muscle hypertrophy was induced by attaching a weight to one wing of young and aged Japanese quails for 14 days. Removing the weight for 7 or 14 days after the initial 14 days of loading induced muscle atrophy. The contralateral wing served as the intra-animal control. A time-released bromodeoxyuridine (BrdU) pellet was implanted subcutaneously with wing weighting to identify activated satellite cells/muscle precursor cells throughout the experimental period. Bcl-2 mRNA and protein levels decreased after 7 days of unloading, but they were unchanged after 14 days of unloading in young muscles. Bcl-2 protein level but not mRNA level decreased after 7 days of unloading in muscles of aged birds. Seven days of unloading increased the mRNA level of Bax in muscles from both young and aged birds. Fourteen days of unloading increased mRNA and protein levels of Bcl-2, decreased protein levels of Bax, and decreased nuclear apoptosis-inducing factor (AIF) protein level in muscles of aged birds. BrdU-positive nuclei were found in all unloaded muscles from both age groups, but the number of BrdU-positive nuclei relative to the total nuclei decreased after 14 days of unloading compared with 7 days of unloading. The TdT-mediated dUTP nick end labeling (TUNEL) index was higher after 7 days of unloading in both young and aged muscles and after 14 days of unloading in aged muscles. Immunofluorescent staining revealed that almost all of the TUNEL-positive nuclei were also BrdU immunopositive, suggesting that activated satellite cell nuclei (both fused and nonfused) underwent nuclear apoptosis during unloading. There were significant correlations among levels of Bcl-2, Bax, and AIF and TUNEL index. Our data are consistent with the hypothesis that apoptosis regulates, at least in part, unloading-induced muscle atrophy and loss of activated satellite cell nuclei in previously loaded muscles. Moreover, these data suggest that aging influences the apoptotic responses to prolonged unloading following hypertrophy in skeletal myocytes.
Collapse
Affiliation(s)
- Parco M Siu
- Division of Exercise Physiology, West Virginia University School of Medicine, Morgantown, West Virginia 26506-9227, USA
| | | | | | | |
Collapse
|