1
|
Collins BC, Shapiro JB, Scheib MM, Musci RV, Verma M, Kardon G. Three-dimensional imaging studies in mice identify cellular dynamics of skeletal muscle regeneration. Dev Cell 2024; 59:1457-1474.e5. [PMID: 38569550 PMCID: PMC11153043 DOI: 10.1016/j.devcel.2024.03.017] [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: 04/25/2023] [Revised: 12/06/2023] [Accepted: 03/08/2024] [Indexed: 04/05/2024]
Abstract
The function of many organs, including skeletal muscle, depends on their three-dimensional structure. Muscle regeneration therefore requires not only reestablishment of myofibers but also restoration of tissue architecture. Resident muscle stem cells (SCs) are essential for regeneration, but how SCs regenerate muscle architecture is largely unknown. We address this problem using genetic labeling of mouse SCs and whole-mount imaging to reconstruct, in three dimensions, muscle regeneration. Unexpectedly, we found that myofibers form via two distinct phases of fusion and the residual basement membrane of necrotic myofibers is critical for promoting fusion and orienting regenerated myofibers. Furthermore, the centralized myonuclei characteristic of regenerated myofibers are associated with myofibrillogenesis and endure months post injury. Finally, we elucidate two cellular mechanisms for the formation of branched myofibers, a pathology characteristic of diseased muscle. We provide a synthesis of the cellular events of regeneration and show that these differ from those used during development.
Collapse
Affiliation(s)
- Brittany C Collins
- Department of Human Genetics, University of Utah, Salt Lake City, UT, USA
| | - Jacob B Shapiro
- Department of Human Genetics, University of Utah, Salt Lake City, UT, USA
| | - Mya M Scheib
- Department of Human Genetics, University of Utah, Salt Lake City, UT, USA
| | - Robert V Musci
- Department of Human Genetics, University of Utah, Salt Lake City, UT, USA
| | - Mayank Verma
- Department of Pediatrics, Division of Neurology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Gabrielle Kardon
- Department of Human Genetics, University of Utah, Salt Lake City, UT, USA.
| |
Collapse
|
2
|
Granic A, Suetterlin K, Shavlakadze T, Grounds M, Sayer A. Hallmarks of ageing in human skeletal muscle and implications for understanding the pathophysiology of sarcopenia in women and men. Clin Sci (Lond) 2023; 137:1721-1751. [PMID: 37986616 PMCID: PMC10665130 DOI: 10.1042/cs20230319] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 11/01/2023] [Accepted: 11/14/2023] [Indexed: 11/22/2023]
Abstract
Ageing is a complex biological process associated with increased morbidity and mortality. Nine classic, interdependent hallmarks of ageing have been proposed involving genetic and biochemical pathways that collectively influence ageing trajectories and susceptibility to pathology in humans. Ageing skeletal muscle undergoes profound morphological and physiological changes associated with loss of strength, mass, and function, a condition known as sarcopenia. The aetiology of sarcopenia is complex and whilst research in this area is growing rapidly, there is a relative paucity of human studies, particularly in older women. Here, we evaluate how the nine classic hallmarks of ageing: genomic instability, telomere attrition, epigenetic alterations, loss of proteostasis, deregulated nutrient sensing, mitochondrial dysfunction, cellular senescence, stem cell exhaustion, and altered intercellular communication contribute to skeletal muscle ageing and the pathophysiology of sarcopenia. We also highlight five novel hallmarks of particular significance to skeletal muscle ageing: inflammation, neural dysfunction, extracellular matrix dysfunction, reduced vascular perfusion, and ionic dyshomeostasis, and discuss how the classic and novel hallmarks are interconnected. Their clinical relevance and translational potential are also considered.
Collapse
Affiliation(s)
- Antoneta Granic
- AGE Research Group, Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, U.K
- NIHR Newcastle Biomedical Research Centre, Newcastle University and Newcastle Upon Tyne Hospitals NHS Foundation Trust, Newcastle Upon Tyne, U.K
| | - Karen Suetterlin
- AGE Research Group, Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, U.K
- NIHR Newcastle Biomedical Research Centre, Newcastle University and Newcastle Upon Tyne Hospitals NHS Foundation Trust, Newcastle Upon Tyne, U.K
- John Walton Muscular Dystrophy Research Centre, Institute of Genetic Medicine, Newcastle University, Centre for Life, Newcastle upon Tyne, U.K
| | - Tea Shavlakadze
- Regeneron Pharmaceuticals Inc., Tarrytown, New York, NY, U.S.A
| | - Miranda D. Grounds
- Department of Anatomy, Physiology and Human Biology, School of Human Sciences, the University of Western Australia, Perth, WA 6009, Australia
| | - Avan A. Sayer
- AGE Research Group, Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, U.K
- NIHR Newcastle Biomedical Research Centre, Newcastle University and Newcastle Upon Tyne Hospitals NHS Foundation Trust, Newcastle Upon Tyne, U.K
| |
Collapse
|
3
|
Harriot AD, Altair Morris T, Vanegas C, Kallenbach J, Pinto K, Joca HC, Moutin MJ, Shi G, Ursitti JA, Grosberg A, Ward CW. Detyrosinated microtubule arrays drive myofibrillar malformations in mdx muscle fibers. Front Cell Dev Biol 2023; 11:1209542. [PMID: 37691825 PMCID: PMC10485621 DOI: 10.3389/fcell.2023.1209542] [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: 04/20/2023] [Accepted: 08/09/2023] [Indexed: 09/12/2023] Open
Abstract
Altered myofibrillar structure is a consequence of dystrophic pathology that impairs skeletal muscle contractile function and increases susceptibility to contraction injury. In murine Duchenne muscular dystrophy (mdx), myofibrillar alterations are abundant in advanced pathology (>4 months), an age where we formerly established densified microtubule (MT) arrays enriched in detyrosinated (deTyr) tubulin as negative disease modifiers impacting cell mechanics and mechanotransduction. Given the essential role of deTyr-enriched MT arrays in myofibrillar growth, maintenance, and repair, we examined the increased abundance of these arrays as a potential mechanism for these myofibrillar alterations. Here we find an increase in deTyr-tubulin as an early event in dystrophic pathology (4 weeks) with no evidence myofibrillar alterations. At 16 weeks, we show deTyr-enriched MT arrays significantly densified and co-localized to areas of myofibrillar malformation. Profiling the enzyme complexes responsible for deTyr-tubulin, we identify vasohibin 2 (VASH2) and small vasohibin binding protein (SVBP) significantly elevated in the mdx muscle at 4 weeks. Using the genetic increase in VASH2/SVBP expression in 4 weeks wild-type mice we find densified deTyr-enriched MT arrays that co-segregate with myofibrillar malformations similar to those in the 16 weeks mdx. Given that no changes in sarcomere organization were identified in fibers expressing sfGFP as a control, we conclude that disease-dependent densification of deTyr-enriched MT arrays underscores the altered myofibrillar structure in dystrophic skeletal muscle fibers.
Collapse
Affiliation(s)
- Anicca D. Harriot
- Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Tessa Altair Morris
- Center for Complex Biological Systems, Edwards Lifesciences Foundation Cardiovascular Innovation and Research Center, and the NSF-Simons Center for Multiscale Cell Fate Research, University of California, Irvine, Irvine, CA, United States
| | - Camilo Vanegas
- Department of Orthopedics, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Jacob Kallenbach
- Department of Orthopedics, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Kaylie Pinto
- Department of Molecular Medicine, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Humberto C. Joca
- Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Marie-Jo Moutin
- INSERM U1216 Centre National de la Recherche Scientifique, Grenoble Institut Neurosciences, University Grenoble Alpes, Grenoble, France
| | - Guoli Shi
- Department of Orthopedics, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Jeanine A. Ursitti
- Department of Orthopedics, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Anna Grosberg
- Center for Complex Biological Systems, Edwards Lifesciences Foundation Cardiovascular Innovation and Research Center, and the NSF-Simons Center for Multiscale Cell Fate Research, University of California, Irvine, Irvine, CA, United States
- Department of Biomedical Engineering, Sue and Bill Gross Stem Cell Research, University of California, Irvine, Irvine, CA, United States
- Department of Chemical and Biomolecular Engineering, Sue and Bill Gross Stem Cell Research, University of California, Irvine, Irvine, CA, United States
| | - Christopher W. Ward
- Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, MD, United States
- Department of Orthopedics, University of Maryland School of Medicine, Baltimore, MD, United States
| |
Collapse
|
4
|
Pizza FX, Buckley KH. Regenerating Myofibers after an Acute Muscle Injury: What Do We Really Know about Them? Int J Mol Sci 2023; 24:12545. [PMID: 37628725 PMCID: PMC10454182 DOI: 10.3390/ijms241612545] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 08/02/2023] [Accepted: 08/04/2023] [Indexed: 08/27/2023] Open
Abstract
Injury to skeletal muscle through trauma, physical activity, or disease initiates a process called muscle regeneration. When injured myofibers undergo necrosis, muscle regeneration gives rise to myofibers that have myonuclei in a central position, which contrasts the normal, peripheral position of myonuclei. Myofibers with central myonuclei are called regenerating myofibers and are the hallmark feature of muscle regeneration. An important and underappreciated aspect of muscle regeneration is the maturation of regenerating myofibers into a normal sized myofiber with peripheral myonuclei. Strikingly, very little is known about processes that govern regenerating myofiber maturation after muscle injury. As knowledge of myofiber formation and maturation during embryonic, fetal, and postnatal development has served as a foundation for understanding muscle regeneration, this narrative review discusses similarities and differences in myofiber maturation during muscle development and regeneration. Specifically, we compare and contrast myonuclear positioning, myonuclear accretion, myofiber hypertrophy, and myofiber morphology during muscle development and regeneration. We also discuss regenerating myofibers in the context of different types of myofiber necrosis (complete and segmental) after muscle trauma and injurious contractions. The overall goal of the review is to provide a framework for identifying cellular and molecular processes of myofiber maturation that are unique to muscle regeneration.
Collapse
Affiliation(s)
- Francis X. Pizza
- Department of Exercise and Rehabilitation Sciences, University of Toledo, Toledo, OH 43606, USA
| | - Kole H. Buckley
- Division of Gastroenterology and Hepatology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA;
| |
Collapse
|
5
|
Use of a novel technique to assess impact of age-related denervation on mouse soleus muscle function. Biogerontology 2023; 24:377-390. [PMID: 36790689 PMCID: PMC10147802 DOI: 10.1007/s10522-023-10021-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Accepted: 01/26/2023] [Indexed: 02/16/2023]
Abstract
Denervation contributes to loss of force-generating capacity in aged skeletal muscles, but problems with quantification of denervated fibers mean the precise impact of denervation on muscle function remains unclear. This study therefore looked to develop a reliable assay for identifying denervated muscle fibers, and used this to explore the impact of denervation on age-related force-generation in mouse skeletal muscle. Thirteen young (6-month-old) and 10 old (24-months-old) C57Bl/6 J female mice were utilized. Anaesthetized mice were infused with the fluorescent deoxyglucose analog 2[N-(7-nitrobenz-2-oxa-1,2-diaxol-4-yl)amino]-2-deoxyglucose (2-NBDG) and the tibial nerve was repeatedly stimulated to label active skeletal muscle fibers by activity-dependent uptake of 2-NBDG. Data on muscle force generation were acquired as part of the stimulation routine. Labeled muscles were removed, snap frozen, sectioned, and slide mounted. Sections were imaged to show accumulation of 2-NBDG in activated fibers and lack of 2-NBDG accumulation in quiescent (denervated) fibers, then processed using immunohistochemistry to allow collection of data on fiber number and morphology. Soleus muscles from older mice had nine times as many denervated fibers as those from young mice (average n = 36 vs 4, old vs young). Older muscles developed significantly more passive force and less specific force, but denervation only partly accounted for age-related deficits in specific force. Further investigations are required to definitively identify contributors to the decrease in force generation that remain unaccounted for.
Collapse
|
6
|
Kim HJ, Kim SW, Lee SH, Jung DW, Williams DR. Inhibiting 5-lipoxygenase prevents skeletal muscle atrophy by targeting organogenesis signalling and insulin-like growth factor-1. J Cachexia Sarcopenia Muscle 2022; 13:3062-3077. [PMID: 36221153 PMCID: PMC9745465 DOI: 10.1002/jcsm.13092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 08/05/2022] [Accepted: 09/02/2022] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Skeletal muscle atrophy can occur in response to numerous factors, such as ageing and certain medications, and produces a major socio-economic burden. At present, there are no approved drugs for treating skeletal muscle atrophy. Arachidonate 5-lipoxygenase (Alox5) is a drug target for a number of diseases. However, pharmacological targeting of Alox5, and its role in skeletal muscle atrophy, is unclear. METHODS The potential effects of gene knockdown and pharmacological targeting of Alox5 on skeletal muscle atrophy were investigated using cell-based models, animal models and human skeletal muscle primary cells. Malotilate, a clinically safe drug developed for enhancing liver regeneration and Alox5 inhibitor, was investigated as a repurposing candidate. Mechanism(s) of action in skeletal muscle atrophy was assessed by measuring the expression level or activation status of key regulatory pathways and validated using gene knockdown and RNA sequencing. RESULTS Myotubes treated with the atrophy-inducing glucocorticoid, dexamethasone, were protected from catabolic responses by treatment with malotilate (+41.29%, P < 0.01). Similar anti-atrophy effects were achieved by gene knockdown of Alox5 (+30.4%, P < 0.05). Malotilate produced anti-atrophy effects without affecting the myogenic differentiation programme. In an in vivo model of skeletal muscle atrophy, malotilate treatment preserved muscle force/strength (grip strength: +35.72%, latency to fall: +553.1%, P < 0.05), increased mass and fibre cross-sectional area (quadriceps: +23.72%, soleus: +33.3%, P < 0.01) and down-regulated atrogene expression (Atrogin-1: -61.58%, Murf-1: -66.06%, P < 0.01). Similar, beneficial effects of malotilate treatment were observed in an ageing muscle model, which also showed the preservation of fast-twitch fibres (Type 2a: +56.48%, Type 2b: +37.32%, P < 0.01). Leukotriene B4, a product of Alox5 activity with inflammatory and catabolic functions, was found to be elevated in skeletal muscle undergoing atrophy (quadriceps: +224.4%, P < 0.001). Cellular transcriptome analysis showed that targeting Alox5 up-regulated biological processes regulating organogenesis and increased the expression of insulin-like growth factor-1, a key anti-atrophy hormone (+226.5%, P < 0.05). Interestingly, these effects were restricted to the atrophy condition and not observed in normal skeletal muscle cultures with Alox5 inhibition. Human myotubes were also protected from atrophy by pharmacological targeting of Alox5 (+23.68%, P < 0.05). CONCLUSIONS These results shed new light on novel drug targets and mechanisms underpinning skeletal muscle atrophy. Alox5 is a regulator and drug target for muscle atrophy, and malotilate is an attractive compound for repurposing studies to treat this disease.
Collapse
Affiliation(s)
- Hyun-Jun Kim
- New Drug Targets Laboratory, School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju, South Korea
| | - Seon-Wook Kim
- New Drug Targets Laboratory, School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju, South Korea
| | - Sang-Hoon Lee
- New Drug Targets Laboratory, School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju, South Korea
| | - Da-Woon Jung
- New Drug Targets Laboratory, School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju, South Korea
| | - Darren R Williams
- New Drug Targets Laboratory, School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju, South Korea
| |
Collapse
|
7
|
Hyperglycemia Negatively Affects IPSC-Derived Myoblast Proliferation and Skeletal Muscle Regeneration and Function. Cells 2022; 11:cells11223674. [PMID: 36429100 PMCID: PMC9688533 DOI: 10.3390/cells11223674] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 10/17/2022] [Accepted: 10/28/2022] [Indexed: 11/22/2022] Open
Abstract
Diabetic myopathy is a co-morbidity diagnosed in most diabetes mellitus patients, yet its pathogenesis is still understudied, which hinders the development of effective therapies. This project aimed to investigate the effect of hyperglycemia on human myoblast physiology, devoid of other complicating factors, by utilizing human myoblasts derived from induced pluripotent stem cells (iPSCs), in a defined in vitro system. IPSC-derived myoblasts were expanded under three glucose conditions: low (5 mM), medium (17.5 mM) or high (25 mM). While hyperglycemic myoblasts demonstrated upregulation of Glut4 relative to the euglycemic control, myoblast proliferation demonstrated a glucose dose-dependent impedance. Further cellular analysis revealed a retarded cell cycle progression trapped at the S phase and G2/M phase and an impaired mitochondrial function in hyperglycemic myoblasts. Terminal differentiation of these hyperglycemic myoblasts resulted in significantly hypertrophic and highly branched myotubes with disturbed myosin heavy chain arrangement. Lastly, functional assessment of these myofibers derived from hyperglycemic myoblasts demonstrated comparatively increased fatigability. Collectively, the hyperglycemic myoblasts demonstrated deficient muscle regeneration capability and functionality, which falls in line with the sarcopenia symptoms observed in diabetic myopathy patients. This human-based iPSC-derived skeletal muscle hyperglycemic model provides a valuable platform for mechanistic investigation of diabetic myopathy and therapeutic development.
Collapse
|
8
|
Lloyd EM, Hepburn MS, Li J, Mowla A, Hwang Y, Choi YS, Grounds MD, Kennedy BF. Three-dimensional mechanical characterization of murine skeletal muscle using quantitative micro-elastography. BIOMEDICAL OPTICS EXPRESS 2022; 13:5879-5899. [PMID: 36733728 PMCID: PMC9872891 DOI: 10.1364/boe.471062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 08/29/2022] [Accepted: 09/02/2022] [Indexed: 06/18/2023]
Abstract
Skeletal muscle function is governed by both the mechanical and structural properties of its constituent tissues, which are both modified by disease. Characterizing the mechanical properties of skeletal muscle tissue at an intermediate scale, i.e., between that of cells and organs, can provide insight into diseases such as muscular dystrophies. In this study, we use quantitative micro-elastography (QME) to characterize the micro-scale elasticity of ex vivo murine skeletal muscle in three-dimensions in whole muscles. To address the challenge of achieving high QME image quality with samples featuring uneven surfaces and geometry, we encapsulate the muscles in transparent hydrogels with flat surfaces. Using this method, we study aging and disease in quadriceps tissue by comparing normal wild-type (C57BL/6J) mice with dysferlin-deficient BLAJ mice, a model for the muscular dystrophy dysferlinopathy, at 3, 10, and 24 months of age (sample size of three per group). We observe a 77% decrease in elasticity at 24 months in dysferlin-deficient quadriceps compared to wild-type quadriceps.
Collapse
Affiliation(s)
- Erin M. Lloyd
- Department of Anatomy, Physiology and Human Biology, School of Human Sciences, The University of Western Australia, 35 Stirling Highway, Perth, Western Australia, 6009, Australia
- These authors contributed equally to this work
| | - Matt S. Hepburn
- BRITElab, Harry Perkins Institute of Medical Research, QEII Medical Centre, Nedlands, Western Australia, 6009, Australia and Centre for Medical Research, The University of Western Australia, Perth, Western Australia, 6009, Australia
- Department of Electrical, Electronic & Computer Engineering, School of Engineering, The University of Western Australia, 35, Stirling Highway, Perth, Western Australia, 6009, Australia
- These authors contributed equally to this work
| | - Jiayue Li
- BRITElab, Harry Perkins Institute of Medical Research, QEII Medical Centre, Nedlands, Western Australia, 6009, Australia and Centre for Medical Research, The University of Western Australia, Perth, Western Australia, 6009, Australia
- Department of Electrical, Electronic & Computer Engineering, School of Engineering, The University of Western Australia, 35, Stirling Highway, Perth, Western Australia, 6009, Australia
- Australian Research Council Centre for Personalised Therapeutics Technologies, Australia
| | - Alireza Mowla
- BRITElab, Harry Perkins Institute of Medical Research, QEII Medical Centre, Nedlands, Western Australia, 6009, Australia and Centre for Medical Research, The University of Western Australia, Perth, Western Australia, 6009, Australia
- Department of Electrical, Electronic & Computer Engineering, School of Engineering, The University of Western Australia, 35, Stirling Highway, Perth, Western Australia, 6009, Australia
| | - Yongsung Hwang
- Soonchunhyang Institute of Medi-Bio Science, Soonchunhyang University, Cheonan-si, Chungcheongnam-do, 31151, Republic of Korea
| | - Yu Suk Choi
- Department of Anatomy, Physiology and Human Biology, School of Human Sciences, The University of Western Australia, 35 Stirling Highway, Perth, Western Australia, 6009, Australia
| | - Miranda D. Grounds
- Department of Anatomy, Physiology and Human Biology, School of Human Sciences, The University of Western Australia, 35 Stirling Highway, Perth, Western Australia, 6009, Australia
| | - Brendan F. Kennedy
- BRITElab, Harry Perkins Institute of Medical Research, QEII Medical Centre, Nedlands, Western Australia, 6009, Australia and Centre for Medical Research, The University of Western Australia, Perth, Western Australia, 6009, Australia
- Department of Electrical, Electronic & Computer Engineering, School of Engineering, The University of Western Australia, 35, Stirling Highway, Perth, Western Australia, 6009, Australia
- Australian Research Council Centre for Personalised Therapeutics Technologies, Australia
| |
Collapse
|
9
|
Babaeijandaghi F, Cheng R, Kajabadi N, Soliman H, Chang CK, Smandych J, Tung LW, Long R, Ghassemi A, Rossi FMV. Metabolic reprogramming of skeletal muscle by resident macrophages points to CSF1R inhibitors as muscular dystrophy therapeutics. Sci Transl Med 2022; 14:eabg7504. [PMID: 35767650 DOI: 10.1126/scitranslmed.abg7504] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
The role of tissue-resident macrophages during tissue regeneration or fibrosis is not well understood, mainly due to the lack of a specific marker for their identification. Here, we identified three populations of skeletal muscle-resident myelomonocytic cells: a population of macrophages positive for lymphatic vessel endothelial receptor 1 (LYVE1) and T cell membrane protein 4 (TIM4 or TIMD4), a population of LYVE1-TIM4- macrophages, and a population of cells likely representing dendritic cells that were positive for CD11C and major histocompatibility complex class II (MHCII). Using a combination of parabiosis and lineage-tracing experiments, we found that, at steady state, TIM4- macrophages were replenished from the blood, whereas TIM4+ macrophages locally self-renewed [self-renewing resident macrophages (SRRMs)]. We further showed that Timd4 could be reliably used to distinguish SRRMs from damage-induced infiltrating macrophages. Using a colony-stimulating factor 1 receptor (CSF1R) inhibition/withdrawal approach to specifically deplete SRRMs, we found that SRRMs provided a nonredundant function in clearing damage-induced apoptotic cells early after extensive acute injury. In contrast, in chronic mild injury as seen in a mouse model of Duchenne muscular dystrophy, depletion of both TIM4-- and TIM4+-resident macrophage populations through long-term CSF1R inhibition changed muscle fiber composition from damage-sensitive glycolytic fibers toward damage-resistant glycolytic-oxidative fibers, thereby protecting muscle against contraction-induced injury both ex vivo and in vivo. This work reveals a previously unidentified role for resident macrophages in modulating tissue metabolism and may have therapeutic potential given the ongoing clinical testing of CSF1R inhibitors.
Collapse
Affiliation(s)
- Farshad Babaeijandaghi
- Biomedical Research Centre, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Ryan Cheng
- Biomedical Research Centre, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Nasim Kajabadi
- Biomedical Research Centre, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Hesham Soliman
- Biomedical Research Centre, University of British Columbia, Vancouver, BC V6T 1Z3, Canada.,Department of Pharmacology and Toxicology, Faculty of Pharmacy, Minia University, Minia 61519, Egypt.,Aspect Biosystems, 1781 W 75th Ave, Vancouver, BC V6P 6P2, Canada
| | - Chih-Kai Chang
- Biomedical Research Centre, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Josh Smandych
- Biomedical Research Centre, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Lin Wei Tung
- Biomedical Research Centre, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Reece Long
- Biomedical Research Centre, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Amirhossein Ghassemi
- Biomedical Research Centre, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Fabio M V Rossi
- Biomedical Research Centre, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| |
Collapse
|
10
|
Kiriaev L, Houweling PJ, North KN, Head SI. Loss of α-actinin-3 confers protection from eccentric contraction damage in fast-twitch EDL muscles from aged mdx dystrophic mice by reducing pathological fibre branching. Hum Mol Genet 2022; 31:1417-1429. [PMID: 34761268 PMCID: PMC9071495 DOI: 10.1093/hmg/ddab326] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 10/27/2021] [Accepted: 11/02/2021] [Indexed: 11/14/2022] Open
Abstract
The common null polymorphism (R577X) in the ACTN3 gene is present in over 1.5 billion people worldwide and results in the absence of the protein α-actinin-3 from the Z-discs of fast-twitch skeletal muscle fibres. We have previously reported that this polymorphism is a modifier of dystrophin-deficient Duchenne Muscular Dystrophy. To investigate the mechanism underlying this, we use a double knockout (dk)Actn3KO/mdx (dKO) mouse model, which lacks both dystrophin and sarcomere α-actinin-3. We used dKO mice and mdx dystrophic mice at 12 months (aged) to investigate the correlation between morphological changes to the fast-twitch dKO EDL and the reduction in force deficit produced by an in vitro eccentric contraction protocol. In the aged dKO mouse, we found a marked reduction in fibre branching complexity that correlated with protection from eccentric contraction induced force deficit. Complex branches in the aged dKO EDL fibres (28%) were substantially reduced compared to aged mdx EDL fibres (68%), and this correlates with a graded force loss over three eccentric contractions for dKO muscles (~36% after first contraction, ~66% overall) compared to an abrupt drop in mdx upon the first eccentric contraction (~75% after first contraction, ~89% after three contractions). In dKO, protection from eccentric contraction damage was linked with a doubling of SERCA1 pump density the EDL. We propose that the increased oxidative metabolism of fast-twitch glycolytic fibres characteristic of the null polymorphism (R577X) and increase in SR Ca2+ pump proteins reduces muscle fibre branching and decreases susceptibility to eccentric injury in the dystrophinopathies.
Collapse
Affiliation(s)
- Leonit Kiriaev
- School of Medicine, Western Sydney University, Sydney, NSW 2560, Australia
| | - Peter J Houweling
- Murdoch Children’s Research Institute, Melbourne, Victoria 3052, Australia
- Department of Paediatrics, University of Melbourne, Melbourne, Victoria 3052, Australia
| | - Kathryn N North
- Murdoch Children’s Research Institute, Melbourne, Victoria 3052, Australia
- Department of Paediatrics, University of Melbourne, Melbourne, Victoria 3052, Australia
| | - Stewart I Head
- School of Medicine, Western Sydney University, Sydney, NSW 2560, Australia
- Murdoch Children’s Research Institute, Melbourne, Victoria 3052, Australia
| |
Collapse
|
11
|
Adachi Y, Masuda M, Sakakibara I, Uchida T, Niida Y, Mori Y, Kamei Y, Okumura Y, Ohminami H, Ohnishi K, Yamanaka-Okumura H, Nikawa T, Taketani Y. All-trans retinoic acid changes muscle fiber type via increasing GADD34 dependent on MAPK signal. Life Sci Alliance 2022; 5:5/7/e202101345. [PMID: 35318262 PMCID: PMC8960774 DOI: 10.26508/lsa.202101345] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 03/09/2022] [Accepted: 03/11/2022] [Indexed: 11/25/2022] Open
Abstract
ATRA increases GADD34 expression by decreasing the expression of Six1, which down-regulates the transcriptional activity with TLE3 and increasing mRNA stability through blocking the interaction between TTP and ARE on GADD34 mRNA, resulting in muscle fiber type change. All-trans retinoic acid (ATRA) increases the sensitivity to unfolded protein response in differentiating leukemic blasts. The downstream transcriptional factor of PERK, a major arm of unfolded protein response, regulates muscle differentiation. However, the role of growth arrest and DNA damage-inducible protein 34 (GADD34), one of the downstream factors of PERK, and the effects of ATRA on GADD34 expression in muscle remain unclear. In this study, we identified ATRA increased the GADD34 expression independent of the PERK signal in the gastrocnemius muscle of mice. ATRA up-regulated GADD34 expression through the transcriptional activation of GADD34 gene via inhibiting the interaction of homeobox Six1 and transcription co-repressor TLE3 with the MEF3-binding site on the GADD34 gene promoter in skeletal muscle. ATRA also inhibited the interaction of TTP, which induces mRNA degradation, with AU-rich element on GADD34 mRNA via p-38 MAPK, resulting in the instability of GADD34 mRNA. Overexpressed GADD34 in C2C12 cells changes the type of myosin heavy chain in myotubes. These results suggest ATRA increases GADD34 expression via transcriptional and post-transcriptional regulation, which changes muscle fiber type.
Collapse
Affiliation(s)
- Yuichiro Adachi
- Department of Clinical Nutrition and Food Management, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Japan
| | - Masashi Masuda
- Department of Clinical Nutrition and Food Management, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Japan
| | - Iori Sakakibara
- Department of Nutritional Physiology, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Japan
| | - Takayuki Uchida
- Department of Nutritional Physiology, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Japan
| | - Yuki Niida
- Department of Clinical Nutrition and Food Management, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Japan
| | - Yuki Mori
- Department of Clinical Nutrition and Food Management, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Japan
| | - Yuki Kamei
- Department of Clinical Nutrition and Food Management, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Japan
| | - Yosuke Okumura
- Department of Clinical Nutrition and Food Management, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Japan
| | - Hirokazu Ohminami
- Department of Clinical Nutrition and Food Management, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Japan
| | - Kohta Ohnishi
- Department of Clinical Nutrition and Food Management, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Japan
| | - Hisami Yamanaka-Okumura
- Department of Clinical Nutrition and Food Management, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Japan
| | - Takeshi Nikawa
- Department of Nutritional Physiology, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Japan
| | - Yutaka Taketani
- Department of Clinical Nutrition and Food Management, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Japan
| |
Collapse
|
12
|
Kiriaev L, Kueh S, Morley JW, North KN, Houweling PJ, Head SI. Lifespan Analysis of Dystrophic mdx Fast-Twitch Muscle Morphology and Its Impact on Contractile Function. Front Physiol 2021; 12:771499. [PMID: 34950049 PMCID: PMC8689589 DOI: 10.3389/fphys.2021.771499] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Accepted: 11/08/2021] [Indexed: 11/13/2022] Open
Abstract
Duchenne muscular dystrophy is caused by the absence of the protein dystrophin from skeletal muscle and is characterized by progressive cycles of necrosis/regeneration. Using the dystrophin deficient mdx mouse model, we studied the morphological and contractile chronology of dystrophic skeletal muscle pathology in fast-twitch Extensor Digitorum Longus muscles from animals 4–22 months of age containing 100% regenerated muscle fibers. Catastrophically, the older age groups lost ∼80% of their maximum force after one eccentric contraction (EC) of 20% strain with the greatest loss of ∼92% recorded in senescent 22-month-old mdx mice. In old age groups, there was minimal force recovery ∼24% after 120 min, correlated with a dramatic increase in the number and complexity of branched fibers. This data supports our two-phase model where a “tipping point” is reached when branched fibers rupture irrevocably on EC. These findings have important implications for pre-clinical drug studies and genetic rescue strategies.
Collapse
Affiliation(s)
- Leonit Kiriaev
- Myogenica Laboratory, School of Medicine, Western Sydney University, Sydney, NSW, Australia
- *Correspondence: Leonit Kiriaev,
| | - Sindy Kueh
- Myogenica Laboratory, School of Medicine, Western Sydney University, Sydney, NSW, Australia
| | - John W. Morley
- Myogenica Laboratory, School of Medicine, Western Sydney University, Sydney, NSW, Australia
| | - Kathryn N. North
- Muscle Research Group, Murdoch Children’s Research Institute, Melbourne, VIC, Australia
| | - Peter J. Houweling
- Muscle Research Group, Murdoch Children’s Research Institute, Melbourne, VIC, Australia
| | - Stewart I. Head
- Myogenica Laboratory, School of Medicine, Western Sydney University, Sydney, NSW, Australia
- Muscle Research Group, Murdoch Children’s Research Institute, Melbourne, VIC, Australia
| |
Collapse
|
13
|
Age-related structural changes show that loss of fibers is not a significant contributor to muscle atrophy in old mice. Exp Gerontol 2021; 156:111618. [PMID: 34737004 DOI: 10.1016/j.exger.2021.111618] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 10/10/2021] [Accepted: 10/29/2021] [Indexed: 12/25/2022]
Abstract
Age-related loss of skeletal muscle mass is widely considered a consequence of both fiber atrophy and fiber death. Evidence for fiber death derives largely from an age-related reduction in fiber numbers in muscle cross-sections, however it is unclear how age-related alterations in muscle morphology affect accuracy of such counts. To explore this we performed an examination of muscle and tendon length, muscle mass and girth, and pennation angle, in addition to histological section fiber counts of parallel-fibered (sternomastoid), fusiform (biceps brachii), and pennate (tibialis anterior, extensor digitorum longus, soleus) muscles from 31 mice aged 6-32 months. Age-related decline in mass and girth occurred in soleus (p = 0.026; p = 0.040), tibialis anterior (p = 0.004; p = 0.039), and extensor digitorum longus (p = 0.040; p = 0.022) muscles, for which location of maximal girth also changed. Tendon length and pennation angle remained consistent across the lifespan in all except soleus which showed elongation of both proximal and distal tendons coupled with alterations in pennation angle. Age-related decreases in fiber number were observed in transversely sectioned soleus and extensor digitorum longus muscles however when age-related changes in morphology were accounted for via oblique sectioning the age-related decrease in fiber number was eliminated. Findings show loss of fibers is not a significant contributor to age-related muscle wasting in mice, and that age-related changes in connective tissue selectively impact muscle structure. Fiber shortening is a likely contributor to loss of mass and change in function in muscles of old mice.
Collapse
|
14
|
Kiriaev L, Kueh S, Morley JW, Houweling PJ, Chan S, North KN, Head SI. Dystrophin-negative slow-twitch soleus muscles are not susceptible to eccentric contraction induced injury over the lifespan of the mdx mouse. Am J Physiol Cell Physiol 2021; 321:C704-C720. [PMID: 34432537 DOI: 10.1152/ajpcell.00122.2021] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Accepted: 08/09/2021] [Indexed: 11/22/2022]
Abstract
Duchenne muscular dystrophy (DMD) is the second most common fatal genetic disease in humans and is characterized by the absence of a functional copy of the protein dystrophin from skeletal muscle. In dystrophin-negative humans and rodents, regenerated skeletal muscle fibers show abnormal branching. The number of fibers with branches and the complexity of branching increases with each cycle of degeneration/regeneration. Previously, using the mdx mouse model of DMD, we have proposed that once the number and complexity of branched fibers present in dystrophic fast-twitch EDL muscle surpasses a stable level, we term the "tipping point," the branches, in and of themselves, mechanically weaken the muscle by rupturing when subjected to high forces during eccentric contractions. Here, we use the slow-twitch soleus muscle from the dystrophic mdx mouse to study prediseased "periambulatory" dystrophy at 2-3 wk, the peak regenerative "adult" phase at 6-9 wk, and "old" at 58-112 wk. Using isolated mdx soleus muscles, we examined contractile function and response to eccentric contraction correlated with the amount and complexity of regenerated branched fibers. The intact muscle was enzymatically dispersed into individual fibers in order to count fiber branching and some muscles were optically cleared to allow laser scanning confocal microscopy. We demonstrate throughout the lifespan of the mdx mouse that dystrophic slow-twitch soleus muscle is no more susceptible to eccentric contraction-induced injury than age-matched littermate controls and that this is correlated with a reduction in the number and complexity of branched fibers compared with fast-twitch dystrophic EDL muscles.
Collapse
MESH Headings
- Age Factors
- Animals
- Disease Models, Animal
- Dystrophin/deficiency
- Dystrophin/genetics
- Kinetics
- Male
- Mice, Inbred mdx
- Muscle Contraction
- Muscle Fibers, Fast-Twitch/metabolism
- Muscle Fibers, Fast-Twitch/pathology
- Muscle Fibers, Slow-Twitch/metabolism
- Muscle Fibers, Slow-Twitch/pathology
- Muscle Strength
- Muscular Dystrophy, Duchenne/genetics
- Muscular Dystrophy, Duchenne/metabolism
- Muscular Dystrophy, Duchenne/pathology
- Muscular Dystrophy, Duchenne/physiopathology
- Mutation
- Mice
Collapse
Affiliation(s)
- Leonit Kiriaev
- School of Medicine, Western Sydney University, Sydney, New South Wales, Australia
- School of Medical Sciences, University of New South Wales, Sydney, New South Wales, Australia
| | - Sindy Kueh
- School of Medicine, Western Sydney University, Sydney, New South Wales, Australia
- School of Medical Sciences, University of New South Wales, Sydney, New South Wales, Australia
| | - John W Morley
- School of Medicine, Western Sydney University, Sydney, New South Wales, Australia
- School of Medical Sciences, University of New South Wales, Sydney, New South Wales, Australia
| | - Peter J Houweling
- Murdoch Children's Research Institute, Melbourne, Victoria, Australia
| | - Stephen Chan
- School of Medicine, Western Sydney University, Sydney, New South Wales, Australia
- School of Medical Sciences, University of New South Wales, Sydney, New South Wales, Australia
| | - Kathryn N North
- Murdoch Children's Research Institute, Melbourne, Victoria, Australia
| | - Stewart I Head
- School of Medicine, Western Sydney University, Sydney, New South Wales, Australia
- Murdoch Children's Research Institute, Melbourne, Victoria, Australia
- School of Medical Sciences, University of New South Wales, Sydney, New South Wales, Australia
| |
Collapse
|
15
|
Saclier M, Ben Larbi S, My Ly H, Moulin E, Mounier R, Chazaud B, Juban G. Interplay between myofibers and pro-inflammatory macrophages controls muscle damage in mdx mice. J Cell Sci 2021; 134:272022. [PMID: 34471933 DOI: 10.1242/jcs.258429] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Accepted: 07/30/2021] [Indexed: 12/18/2022] Open
Abstract
Duchenne muscular dystrophy is a genetic muscle disease characterized by chronic inflammation and fibrosis mediated by a pro-fibrotic macrophage population expressing pro-inflammatory markers. Our aim was to characterize cellular events leading to the alteration of macrophage properties and to modulate macrophage inflammatory status using the gaseous mediator hydrogen sulfide (H2S). Using co-culture experiments, we first showed that myofibers derived from mdx mice strongly skewed the polarization of resting macrophages towards a pro-inflammatory phenotype. Treatment of mdx mice with NaHS, an H2S donor, reduced the number of pro-inflammatory macrophages in skeletal muscle, which was associated with a decreased number of nuclei per fiber, as well as reduced myofiber branching and fibrosis. Finally, we established the metabolic sensor AMP-activated protein kinase (AMPK) as a critical NaHS target in muscle macrophages. These results identify an interplay between myofibers and macrophages where dystrophic myofibers contribute to the maintenance of a highly inflammatory environment sustaining a pro-inflammatory macrophage status, which in turn favors myofiber damage, myofiber branching and establishment of fibrosis. Our results also highlight the use of H2S donors as a potential therapeutic strategy to improve the dystrophic muscle phenotype by dampening chronic inflammation. This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
- Marielle Saclier
- Department of Biosciences, University of Milan, 20133 Milan, Italy
| | - Sabrina Ben Larbi
- Institut NeuroMyoGène, Université Claude Bernard Lyon 1, CNRS UMR 5310, INSERM U1217, Université de Lyon, 69008 Lyon, France
| | - Ha My Ly
- Institut NeuroMyoGène, Université Claude Bernard Lyon 1, CNRS UMR 5310, INSERM U1217, Université de Lyon, 69008 Lyon, France
| | - Eugénie Moulin
- Institut NeuroMyoGène, Université Claude Bernard Lyon 1, CNRS UMR 5310, INSERM U1217, Université de Lyon, 69008 Lyon, France
| | - Rémi Mounier
- Institut NeuroMyoGène, Université Claude Bernard Lyon 1, CNRS UMR 5310, INSERM U1217, Université de Lyon, 69008 Lyon, France
| | - Bénédicte Chazaud
- Institut NeuroMyoGène, Université Claude Bernard Lyon 1, CNRS UMR 5310, INSERM U1217, Université de Lyon, 69008 Lyon, France
| | - Gaëtan Juban
- Institut NeuroMyoGène, Université Claude Bernard Lyon 1, CNRS UMR 5310, INSERM U1217, Université de Lyon, 69008 Lyon, France
| |
Collapse
|
16
|
Partridge TA. Enhancing Interrogation of Skeletal Muscle Samples for Informative Quantitative Data. J Neuromuscul Dis 2021; 8:S257-S269. [PMID: 34511511 PMCID: PMC8673506 DOI: 10.3233/jnd-210736] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Careful quantitative analysis of histological preparations of muscle samples is crucial to accurate investigation of myopathies in man and of interpretation of data from animals subjected to experimental or potentially therapeutic treatments. Protocols for measuring cell numbers are subject to problems arising from biases associated with preparative and analytical techniques. Prominent among these is the effect of polarized structure of skeletal muscle on sampling bias. It is also common in this tissue to collect data as ratios to convenient reference dominators, the fundamental bases of which are ill-defined, or unrecognized or not accurately assessable. Use of such 'floating' denominators raises a barrier to estimation of the absolute values that assume practical importance in medical research, where accurate comparison between different scenarios in different species is essential to the aim of translating preclinical research findings in animal models to clinical utility in Homo sapiens.This review identifies some of the underappreciated problems with current morphometric practice, some of which are exacerbated in skeletal muscle, and evaluates the extent of their intrusiveness into the of building an objective, accurate, picture of the structure of the muscle sample. It also contains recommendations for eliminating or at least minimizing these problems. Principal among these, would be the use of stereological procedures to avoid the substantial counting biases arising from inter-procedure differences in object size and section thickness.Attention is also drawn to the distortions of interpretation arising from use of undefined or inappropriate denominators.
Collapse
Affiliation(s)
- Terence A Partridge
- Professor of Integrative Systemic Biology, George Washington University, Washington DC.,Honorary Professor, Institute of Child Health, University College London
| |
Collapse
|
17
|
Ben Larbi S, Saclier M, Fessard A, Juban G, Chazaud B. Histological Analysis of Tibialis Anterior Muscle of DMDmdx4Cv Mice from 1 to 24 Months. J Neuromuscul Dis 2021; 8:513-524. [PMID: 33843691 DOI: 10.3233/jnd-200562] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
BACKGROUND The mdx-C57/B6 mouse model does not show the clinical signs of Duchenne muscular dystrophy (DMD), although muscles exhibit hallmarks of permanent regeneration and alterations in muscle function. The DMDmdx4Cv strain exhibits very few revertant dystrophin positive myofibers, making that model suitable for studies on gene and cell therapies. OBJECTIVE The study appraises the histological evolution of the Tibialis Anterior muscle of WT and DMD mdx4Cv mutant from 1 to 24 months. METHODS Histological analysis included a series of immunostainings of muscle sections for assessing tissue features (fibrosis, lipid deposition, necrosis) and cellular characteristics (size of myofibers, number and distribution of myonuclei, number of satellite cells, vessels, macrophages). RESULTS None of the investigated cell types (satellite cells, endothelial cells, macrophages) showed variations in their density within the tissue in both WT and DMD mdx4Cv muscle. However, analyzing their number per myofiber showed that in DMD mdx4Cv, myofiber capillarization was increased from 1 to 6 months as compared with WT muscle, then dropped from 12 months. Macrophage number did not vary in WT muscle and peaked at 6 months in DMD mdx4Cv muscle. The number of satellite cells per myofiber did not vary in WT muscle while it remained high in DMD mdx4Cv muscle, starting to decrease from 12 months and being significantly lower at 24 months of age. Myofiber size was not different in DMD mdx4Cv from WT except at 24 months, when it strongly decreased in DMD mdx4Cv muscle. Necrosis and lipid deposition were rare in DMD mdx4Cv muscle. Fibrosis did not increase with age in DMD mdx4Cv muscle and was higher than in WT at 6 and 12 months of age. CONCLUSIONS As a whole, the results show a strong decrease of the myofiber size at 24 months, and an increased capillarization until 6 months of age in DMD mdx4Cv as compared with the WT. Thus, DMD mdx4Cv mice poorly recapitulates histological DMD features, and its use should take into account the age of the animals according to the purpose of the investigation.
Collapse
Affiliation(s)
- Sabrina Ben Larbi
- Institut NeuroMyoGène, Université Claude Bernard Lyon 1, CNRS UMR 2310, INSERM U1217, Université Lyon, Villeurbanne, France
| | | | - Aurélie Fessard
- Institut NeuroMyoGène, Université Claude Bernard Lyon 1, CNRS UMR 2310, INSERM U1217, Université Lyon, Villeurbanne, France
| | - Gaëtan Juban
- Institut NeuroMyoGène, Université Claude Bernard Lyon 1, CNRS UMR 2310, INSERM U1217, Université Lyon, Villeurbanne, France
| | - Bénédicte Chazaud
- Institut NeuroMyoGène, Université Claude Bernard Lyon 1, CNRS UMR 2310, INSERM U1217, Université Lyon, Villeurbanne, France
| |
Collapse
|
18
|
Bertin B, Renaud Y, Jagla T, Lavergne G, Dondi C, Da Ponte JP, Junion G, Jagla K. Gelsolin and dCryAB act downstream of muscle identity genes and contribute to preventing muscle splitting and branching in Drosophila. Sci Rep 2021; 11:13197. [PMID: 34162956 PMCID: PMC8222376 DOI: 10.1038/s41598-021-92506-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Accepted: 06/03/2021] [Indexed: 11/30/2022] Open
Abstract
A combinatorial code of identity transcription factors (iTFs) specifies the diversity of muscle types in Drosophila. We previously showed that two iTFs, Lms and Ap, play critical role in the identity of a subset of larval body wall muscles, the lateral transverse (LT) muscles. Intriguingly, a small portion of ap and lms mutants displays an increased number of LT muscles, a phenotype that recalls pathological split muscle fibers in human. However, genes acting downstream of Ap and Lms to prevent these aberrant muscle feature are not known. Here, we applied a cell type specific translational profiling (TRAP) to identify gene expression signatures underlying identity of muscle subsets including the LT muscles. We found that Gelsolin (Gel) and dCryAB, both encoding actin-interacting proteins, displayed LT muscle prevailing expression positively regulated by, the LT iTFs. Loss of dCryAB function resulted in LTs with irregular shape and occasional branched ends also observed in ap and lms mutant contexts. In contrast, enlarged and then split LTs with a greater number of myonuclei formed in Gel mutants while Gel gain of function resulted in unfused myoblasts, collectively indicating that Gel regulates LTs size and prevents splitting by limiting myoblast fusion. Thus, dCryAB and Gel act downstream of Lms and Ap and contribute to preventing LT muscle branching and splitting. Our findings offer first clues to still unknown mechanisms of pathological muscle splitting commonly detected in human dystrophic muscles and causing muscle weakness.
Collapse
Affiliation(s)
- Benjamin Bertin
- GReD Institute - INSERM U1103, CNRS UMR6293, Université Clermont Auvergne, 28, place Henri-Dunant, 63000, Clermont-Ferrand, France
| | - Yoan Renaud
- GReD Institute - INSERM U1103, CNRS UMR6293, Université Clermont Auvergne, 28, place Henri-Dunant, 63000, Clermont-Ferrand, France
| | - Teresa Jagla
- GReD Institute - INSERM U1103, CNRS UMR6293, Université Clermont Auvergne, 28, place Henri-Dunant, 63000, Clermont-Ferrand, France
| | - Guillaume Lavergne
- GReD Institute - INSERM U1103, CNRS UMR6293, Université Clermont Auvergne, 28, place Henri-Dunant, 63000, Clermont-Ferrand, France
| | - Cristiana Dondi
- GReD Institute - INSERM U1103, CNRS UMR6293, Université Clermont Auvergne, 28, place Henri-Dunant, 63000, Clermont-Ferrand, France
| | - Jean-Philippe Da Ponte
- GReD Institute - INSERM U1103, CNRS UMR6293, Université Clermont Auvergne, 28, place Henri-Dunant, 63000, Clermont-Ferrand, France
| | - Guillaume Junion
- GReD Institute - INSERM U1103, CNRS UMR6293, Université Clermont Auvergne, 28, place Henri-Dunant, 63000, Clermont-Ferrand, France.
| | - Krzysztof Jagla
- GReD Institute - INSERM U1103, CNRS UMR6293, Université Clermont Auvergne, 28, place Henri-Dunant, 63000, Clermont-Ferrand, France.
| |
Collapse
|
19
|
Girardi F, Taleb A, Ebrahimi M, Datye A, Gamage DG, Peccate C, Giordani L, Millay DP, Gilbert PM, Cadot B, Le Grand F. TGFβ signaling curbs cell fusion and muscle regeneration. Nat Commun 2021; 12:750. [PMID: 33531466 PMCID: PMC7854756 DOI: 10.1038/s41467-020-20289-8] [Citation(s) in RCA: 56] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Accepted: 11/21/2020] [Indexed: 12/12/2022] Open
Abstract
Muscle cell fusion is a multistep process involving cell migration, adhesion, membrane remodeling and actin-nucleation pathways to generate multinucleated myotubes. However, molecular brakes restraining cell-cell fusion events have remained elusive. Here we show that transforming growth factor beta (TGFβ) pathway is active in adult muscle cells throughout fusion. We find TGFβ signaling reduces cell fusion, regardless of the cells' ability to move and establish cell-cell contacts. In contrast, inhibition of TGFβ signaling enhances cell fusion and promotes branching between myotubes in mouse and human. Exogenous addition of TGFβ protein in vivo during muscle regeneration results in a loss of muscle function while inhibition of TGFβR2 induces the formation of giant myofibers. Transcriptome analyses and functional assays reveal that TGFβ controls the expression of actin-related genes to reduce cell spreading. TGFβ signaling is therefore requisite to limit mammalian myoblast fusion, determining myonuclei numbers and myofiber size.
Collapse
Affiliation(s)
- Francesco Girardi
- Sorbonne Université, INSERM UMRS974, Association Institut de Myologie, Centre de Recherche en Myologie, 75013, Paris, France
| | - Anissa Taleb
- Sorbonne Université, INSERM UMRS974, Association Institut de Myologie, Centre de Recherche en Myologie, 75013, Paris, France
| | - Majid Ebrahimi
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON, M5S3G9, Canada
- Donnelly Centre for Cellular and Biomolecular Research, Toronto, ON, M5S3E1, Canada
| | - Asiman Datye
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON, M5S3G9, Canada
- Donnelly Centre for Cellular and Biomolecular Research, Toronto, ON, M5S3E1, Canada
| | - Dilani G Gamage
- Division of Molecular Cardiovascular Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA
| | - Cécile Peccate
- Sorbonne Université, INSERM UMRS974, Association Institut de Myologie, Centre de Recherche en Myologie, 75013, Paris, France
| | - Lorenzo Giordani
- Sorbonne Université, INSERM UMRS974, Association Institut de Myologie, Centre de Recherche en Myologie, 75013, Paris, France
| | - Douglas P Millay
- Division of Molecular Cardiovascular Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, 45229, USA
| | - Penney M Gilbert
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON, M5S3G9, Canada
- Donnelly Centre for Cellular and Biomolecular Research, Toronto, ON, M5S3E1, Canada
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON, M5S3G5, Canada
| | - Bruno Cadot
- Sorbonne Université, INSERM UMRS974, Association Institut de Myologie, Centre de Recherche en Myologie, 75013, Paris, France
| | - Fabien Le Grand
- Sorbonne Université, INSERM UMRS974, Association Institut de Myologie, Centre de Recherche en Myologie, 75013, Paris, France.
- Institut NeuroMyoGène, Université Claude Bernard Lyon 1, CNRS UMR 5310, INSERM U1217, 69008, Lyon, France.
| |
Collapse
|
20
|
Lifetime analysis of mdx skeletal muscle reveals a progressive pathology that leads to myofiber loss. Sci Rep 2020; 10:17248. [PMID: 33057110 PMCID: PMC7560899 DOI: 10.1038/s41598-020-74192-9] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Accepted: 09/23/2020] [Indexed: 02/06/2023] Open
Abstract
The muscular dystrophy X-linked mouse (mdx) is the most commonly used preclinical model for Duchenne muscular dystrophy. Although disease progression in the mouse does not perfectly model the human disease, it shares many pathological features. Early characterizations of the model reported severe pathology through early adulthood followed by disease stabilization. As a result, research in the mdx mouse has largely focused on early adulthood. The overarching goal of this study is to improve the understanding of the mdx mouse model by tracking pathological features of the disease throughout life. We performed a thorough characterization of myofiber pathology in mdx mice from 2 weeks to 2 years of age. We report that individual mdx muscle fibers undergo progressive hypertrophy that continues through the lifespan. Despite massive hypertrophy on the myofiber level, we report no hypertrophy on the muscle level. These seemingly contradictory findings are explained by previously underappreciated myofiber loss in mdx mice. We conclude that due to myofiber loss, in combination with the progressive nature of other pathological features, aged mdx muscle tissue provides reliable benchmarks for disease progression that may be valuable in testing the efficacy of therapeutics for Duchenne muscular dystrophy.
Collapse
|
21
|
Garcia-Pelagio KP, Pratt SJP, Lovering RM. Effects of myofiber isolation technique on sarcolemma biomechanics. Biotechniques 2020; 69:388-391. [PMID: 33000629 PMCID: PMC7686532 DOI: 10.2144/btn-2020-0087] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Isolated myofibers are commonly used to understand the function of skeletal muscle in vivo. This can involve single isolated myofibers obtained from dissection or from enzymatic dissociation. Isolation via dissection allows control of sarcomere length and preserves tendon attachment but is labor-intensive, time-consuming and yields few viable myofibers. In contrast, enzymatic dissociation is fast and facile, produces hundreds of myofibers, and more importantly reduces the number of muscles/animals needed for studies. Biomechanical properties of the sarcolemma have been studied using myofibers from the extensor digitorum longus, but this has been limited to dissected myofibers, making data collection slow and difficult. We have modified this tool to perform biomechanical measurements of the sarcolemma in dissociated myofibers from the flexor digitorum brevis.
Collapse
Affiliation(s)
- Karla P Garcia-Pelagio
- Departmento de Física, Facultad de Ciencias, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Stephen JP Pratt
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Richard M Lovering
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD 21201, USA.,Department of Orthopedics, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| |
Collapse
|
22
|
Martin RA, Buckley KH, Mankowski DC, Riley BM, Sidwell AN, Douglas SL, Worth RG, Pizza FX. Myogenic Cell Expression of Intercellular Adhesion Molecule-1 Contributes to Muscle Regeneration after Injury. THE AMERICAN JOURNAL OF PATHOLOGY 2020; 190:2039-2055. [PMID: 32650005 DOI: 10.1016/j.ajpath.2020.06.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Revised: 06/10/2020] [Accepted: 06/15/2020] [Indexed: 10/23/2022]
Abstract
This study investigated intercellular adhesion molecule-1 (ICAM-1), a membrane protein that mediates cell-to-cell adhesion and communication, as a mechanism through which the inflammatory response facilitates muscle regeneration after injury. Toxin-induced muscle injury to tibialis anterior muscles of wild-type mice caused ICAM-1 to be expressed by a population of satellite cells/myoblasts and myofibers. Myogenic cell expression of ICAM-1 contributed to the restoration of muscle structure after injury, as regenerating myofibers were more abundant and myofiber size was larger for wild-type compared with Icam1-/- mice during 28 days of recovery. Contrastingly, restoration of muscle function after injury was similar between the genotypes. ICAM-1 facilitated the restoration of muscle structure after injury through mechanisms involving the regulation of myofiber branching, protein synthesis, and the organization of nuclei within myofibers after myogenic cell fusion. These findings provide support for a paradigm in which ICAM-1 expressed by myogenic cells after muscle injury augments their adhesive and fusogenic properties, which, in turn, facilitates regenerative and hypertrophic processes that restore structure to injured muscle.
Collapse
Affiliation(s)
- Ryan A Martin
- School of Exercise and Rehabilitation Sciences, The University of Toledo, Toledo, Ohio
| | - Kole H Buckley
- School of Exercise and Rehabilitation Sciences, The University of Toledo, Toledo, Ohio
| | - Drew C Mankowski
- School of Exercise and Rehabilitation Sciences, The University of Toledo, Toledo, Ohio
| | - Benjamin M Riley
- School of Exercise and Rehabilitation Sciences, The University of Toledo, Toledo, Ohio
| | - Alena N Sidwell
- School of Exercise and Rehabilitation Sciences, The University of Toledo, Toledo, Ohio
| | - Stephanie L Douglas
- School of Exercise and Rehabilitation Sciences, The University of Toledo, Toledo, Ohio
| | - Randall G Worth
- Department of Medical Microbiology and Immunology, The University of Toledo, Toledo, Ohio
| | - Francis X Pizza
- School of Exercise and Rehabilitation Sciences, The University of Toledo, Toledo, Ohio.
| |
Collapse
|
23
|
Jorgenson KW, Phillips SM, Hornberger TA. Identifying the Structural Adaptations that Drive the Mechanical Load-Induced Growth of Skeletal Muscle: A Scoping Review. Cells 2020; 9:cells9071658. [PMID: 32660165 PMCID: PMC7408414 DOI: 10.3390/cells9071658] [Citation(s) in RCA: 71] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Revised: 07/06/2020] [Accepted: 07/07/2020] [Indexed: 12/21/2022] Open
Abstract
The maintenance of skeletal muscle mass plays a critical role in health and quality of life. One of the most potent regulators of skeletal muscle mass is mechanical loading, and numerous studies have led to a reasonably clear understanding of the macroscopic and microscopic changes that occur when the mechanical environment is altered. For instance, an increase in mechanical loading induces a growth response that is mediated, at least in part, by an increase in the cross-sectional area of the myofibers (i.e., myofiber hypertrophy). However, very little is known about the ultrastructural adaptations that drive this response. Even the most basic questions, such as whether mechanical load-induced myofiber hypertrophy is mediated by an increase in the size of the pre-existing myofibrils and/or an increase in the number myofibrils, have not been resolved. In this review, we thoroughly summarize what is currently known about the macroscopic, microscopic and ultrastructural changes that drive mechanical load-induced growth and highlight the critical gaps in knowledge that need to be filled.
Collapse
Affiliation(s)
- Kent W. Jorgenson
- School of Veterinary Medicine and the Department of Comparative Biosciences, University of Wisconsin, Madison, WI 53706, USA;
| | - Stuart M. Phillips
- Department of Kinesiology, McMaster University, Hamilton, ON L8S 4K1, Canada;
| | - Troy A. Hornberger
- School of Veterinary Medicine and the Department of Comparative Biosciences, University of Wisconsin, Madison, WI 53706, USA;
- Correspondence:
| |
Collapse
|
24
|
Implications of increased S100β and Tau5 proteins in dystrophic nerves of two mdx mouse models for Duchenne muscular dystrophy. Mol Cell Neurosci 2020; 105:103484. [DOI: 10.1016/j.mcn.2020.103484] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Accepted: 03/25/2020] [Indexed: 12/31/2022] Open
|
25
|
Porcari P, Hall MG, Clark CA, Greally E, Straub V, Blamire AM. Time-dependent diffusion MRI as a probe of microstructural changes in a mouse model of Duchenne muscular dystrophy. NMR IN BIOMEDICINE 2020; 33:e4276. [PMID: 32101354 DOI: 10.1002/nbm.4276] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Revised: 01/05/2020] [Accepted: 01/30/2020] [Indexed: 06/10/2023]
Abstract
Dystrophic muscles show a high variability of fibre sizes and altered sarcolemmal integrity, which are typically assessed by histology. Time-dependent diffusion MRI is sensitive to tissue microstructure and its investigation through age-related changes in dystrophic and healthy muscles may help the understanding of the onset and progression of Duchenne muscular dystrophy (DMD). We investigated the capability of time-dependent diffusion MRI to quantify age and disease-related changes in hind-limb muscle microstructure between dystrophic (mdx) and wild-type (WT) mice of three age groups (7.5, 22 and 44 weeks). Diffusion time-dependent apparent diffusion coefficients (ADCs) of the gastrocnemius and tibialis anterior muscles were determined versus age and diffusion-gradient orientation at six diffusion times (Δ; range: 25-350 ms). Mean muscle ADCs were compared between groups and ages, and correlated with T2 , using Student's t test, one-way analysis of variance and Pearson correlation, respectively. Muscle fibre sizes and sarcolemmal integrity were evaluated by histology and compared with diffusion measurements. Hind-limb muscle ADC showed characteristic restricted diffusion behaviour in both mdx and WT animals with decreasing ADC values at longer Δ. Significant differences in ADC were observed at long Δ values (≥ 250 ms; p < 0.05, comparison between groups; p < 0.01, comparison between ages) with ADC increased by 5-15% in dystrophic muscles, indicative of reduced diffusion restriction. No significant correlation was found between T2 and ADC. Additionally, muscle fibre size distributions showed higher variability and lower mean fibre size in mdx than WT animals (p < 0.001). The extensive Evans Blue Dye uptake shown in dystrophic muscles revealed substantial sarcolemmal damage, suggesting diffusion measurements as more consistent with altered permeability rather than changes in muscle fibre sizes. This study shows the potential of diffusion MRI to non-invasively discriminate between dystrophic and healthy muscles with enhanced sensitivity when using long Δ.
Collapse
Affiliation(s)
- Paola Porcari
- Institute of Genetic Medicine and Centre for In Vivo Imaging, Newcastle University, Newcastle upon Tyne, UK
| | - Matt G Hall
- Developmental Imaging and Biophysics Section, UCL GOS Institute of Child Health, London, UK
| | - Chris A Clark
- Developmental Imaging and Biophysics Section, UCL GOS Institute of Child Health, London, UK
| | - Elizabeth Greally
- John Walton Muscular Dystrophy Research Centre, Institute of Genetic Medicine, Newcastle University, Newcastle upon Tyne, UK
| | - Volker Straub
- John Walton Muscular Dystrophy Research Centre, Institute of Genetic Medicine, Newcastle University, Newcastle upon Tyne, UK
| | - Andrew M Blamire
- Institute of Cellular Medicine and Centre for In Vivo Imaging, Newcastle University, Newcastle upon Tyne, UK
| |
Collapse
|
26
|
Barzilai-Tutsch H, Genin O, Pines M, Halevy O. Early pathological signs in young dysf -/- mice are improved by halofuginone. Neuromuscul Disord 2020; 30:472-482. [PMID: 32451154 DOI: 10.1016/j.nmd.2020.04.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Revised: 03/14/2020] [Accepted: 04/03/2020] [Indexed: 01/09/2023]
Abstract
Dysferlinopathies are a non-lethal group of late-onset muscular dystrophies. Here, we evaluated the fusion ability of primary myoblasts from young dysf-/- mice and the muscle histopathology prior to, and during early stages of disease onset. The ability of primary myoblasts of 5-week-old dysf-/- mice to form large myotubes was delayed compared to their wild-type counterparts, as evaluated by scanning electron microscopy. However, their fusion activity, as reflected by the presence of actin filaments connecting several cells, was enhanced by the antifibrotic drug halofuginone. Early dystrophic signs were already apparent in 4-week-old dysf-/- mice; their collagen level was double that in wild-type mice and continued to rise until 5 months of age. Continuous treatment with halofuginone from 4 weeks to 5 months of age reduced muscle fibrosis in a phosphorylated-Smad3 inhibition-related manner. Halofuginone also enhanced myofiber hypertrophy, reduced the percentage of centrally nucleated myofibers, and increased muscle performance. Together, the data suggest an inhibitory effect of halofuginone on the muscle histopathology at very early stages of dysferlinopathy, and enhancement of muscle performance. These results offer new opportunities for early pharmaceutical treatment in dysferlinopathies with favorable outcomes at later stages of life.
Collapse
Affiliation(s)
- Hila Barzilai-Tutsch
- Department of Animal Sciences, The Hebrew University of Jerusalem, P.O. Box 12, Rehovot 76100, Israel
| | - Olga Genin
- Institute of Animal Science, the Volcani Center, Bet Dagan 52505, Israel
| | - Mark Pines
- Institute of Animal Science, the Volcani Center, Bet Dagan 52505, Israel
| | - Orna Halevy
- Department of Animal Sciences, The Hebrew University of Jerusalem, P.O. Box 12, Rehovot 76100, Israel.
| |
Collapse
|
27
|
Abstract
Skeletal muscle fibres are multinucleated cells that contain postmitotic nuclei (i.e. they are no longer able to divide) and perform muscle contraction. They are formed by fusion of muscle precursor cells, and grow into elongating myofibres by the addition of further precursor cells, called satellite cells, which are also responsible for regeneration following injury. Skeletal muscle regeneration occurs in most muscular dystrophies in response to necrosis of muscle fibres. However, the complex environment within dystrophic skeletal muscle, which includes inflammatory cells, fibroblasts and fibro-adipogenic cells, together with the genetic background of the in vivo model and the muscle being studied, complicates the interpretation of laboratory studies on muscular dystrophies. Many genes are expressed in satellite cells and in other tissues, which makes it difficult to determine the molecular cause of various types of muscular dystrophies. Here, and in the accompanying poster, we discuss our current knowledge of the cellular mechanisms that govern the growth and regeneration of skeletal muscle, and highlight the defects in satellite cell function that give rise to muscular dystrophies. Summary: The mechanisms of skeletal muscle development, growth and regeneration are described. We discuss whether these processes are dysregulated in inherited muscle diseases and identify pathways that may represent therapeutic targets.
Collapse
Affiliation(s)
- Jennifer Morgan
- Dubowitz Neuromuscular Centre, UCL Great Ormond Street Institute of Child Health, 30 Guilford Street, London WC1N 1EH, UK .,National Institute for Health Research, Great Ormond Street Institute of Child Health Biomedical Research Centre, University College London, London WC1N 1EH, UK
| | - Terence Partridge
- Dubowitz Neuromuscular Centre, UCL Great Ormond Street Institute of Child Health, 30 Guilford Street, London WC1N 1EH, UK.,National Institute for Health Research, Great Ormond Street Institute of Child Health Biomedical Research Centre, University College London, London WC1N 1EH, UK.,Center for Genetic Medicine Research, Children's National Medical Center, 111 Michigan Ave NW, Washington, DC 20010, USA
| |
Collapse
|
28
|
Murach KA, Dungan CM, Peterson CA, McCarthy JJ. Muscle Fiber Splitting Is a Physiological Response to Extreme Loading in Animals. Exerc Sport Sci Rev 2019; 47:108-115. [PMID: 30640746 DOI: 10.1249/jes.0000000000000181] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Skeletal muscle fiber branching and splitting typically is associated with damage and regeneration and is considered pathological when observed during loading-induced hypertrophy. We hypothesize that fiber splitting is a nonpathological component of extreme loading and hypertrophy, which is primarily supported by evidence in animals, and propose that the mechanisms and consequences of fiber splitting deserve further exploration.
Collapse
Affiliation(s)
- Kevin A Murach
- Center for Muscle Biology.,Department of Rehabilitation Sciences, and
| | - Cory M Dungan
- Center for Muscle Biology.,Department of Rehabilitation Sciences, and
| | | | - John J McCarthy
- Center for Muscle Biology.,Department of Physiology, University of Kentucky, Lexington, KY
| |
Collapse
|
29
|
Kiriaev L, Kueh S, Morley JW, North KN, Houweling PJ, Head SI. Branched fibers from old fast-twitch dystrophic muscles are the sites of terminal damage in muscular dystrophy. Am J Physiol Cell Physiol 2018; 314:C662-C674. [PMID: 29412689 DOI: 10.1152/ajpcell.00161.2017] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
A striking pathological feature of dystrophinopathies is the presence of morphologically abnormal branched skeletal muscle fibers. The deterioration of muscle contractile function in Duchenne muscular dystrophy is accompanied by both an increase in number and complexity of these branched fibers. We propose that when number and complexity of branched fibers reaches a critical threshold, or "tipping point," the branches in and of themselves are the site of contraction-induced rupture. In the present study, we use the dystrophic mdx mouse and littermate controls to study the prediseased dystrophic fast-twitch extensor digitorum longus (EDL) muscle at 2-3 wk, the peak myonecrotic phase at 6-9 wk, and finally, "old," at 58-112 wk. Using a combination of isolated muscle function contractile measurements coupled with single-fiber imaging and confocal microscope imaging of cleared whole muscles, we identified a distinct pathophysiology, acute fiber rupture at branch nodes, which occurs in "old" fast-twitch EDL muscle approaching the end stage of the dystrophinopathy muscle disease, where the EDL muscles are entirely composed of complexed branched fibers. This evidence supports our concept of "tipping point" where the number and extent of fiber branching reach a level where the branching itself terminally compromises muscle function, irrespective of the absence of dystrophin.
Collapse
Affiliation(s)
- Leonit Kiriaev
- School of Medical Sciences, University of New South Wales , Sidney, New South Wales , Australia
| | - Sindy Kueh
- School of Medical Sciences, University of New South Wales , Sidney, New South Wales , Australia.,School of Medicine, Western Sydney University, Penrith, New South Wales , Australia
| | - John W Morley
- School of Medicine, Western Sydney University, Penrith, New South Wales , Australia
| | - Kathryn N North
- Murdoch Children's Research Institute , Melbourne, Victoria , Australia
| | - Peter J Houweling
- Murdoch Children's Research Institute , Melbourne, Victoria , Australia
| | - Stewart I Head
- School of Medical Sciences, University of New South Wales , Sidney, New South Wales , Australia.,School of Medicine, Western Sydney University, Penrith, New South Wales , Australia
| |
Collapse
|
30
|
Cutler AA, Jackson JB, Corbett AH, Pavlath GK. Non-equivalence of nuclear import among nuclei in multinucleated skeletal muscle cells. J Cell Sci 2018; 131:jcs.207670. [PMID: 29361530 DOI: 10.1242/jcs.207670] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2017] [Accepted: 12/21/2017] [Indexed: 01/01/2023] Open
Abstract
Skeletal muscle is primarily composed of large myofibers containing thousands of post-mitotic nuclei distributed throughout a common cytoplasm. Protein production and localization in specialized myofiber regions is crucial for muscle function. Myonuclei differ in transcriptional activity and protein accumulation, but how these differences among nuclei sharing a cytoplasm are achieved is unknown. Regulated nuclear import of proteins is one potential mechanism for regulating transcription spatially and temporally in individual myonuclei. The best-characterized nuclear localization signal (NLS) in proteins is the classical NLS (cNLS), but many other NLS motifs exist. We examined cNLS and non-cNLS reporter protein import using multinucleated muscle cells generated in vitro, revealing that cNLS and non-cNLS nuclear import differs among nuclei in the same cell. Investigation of cNLS nuclear import rates in isolated myofibers ex vivo confirmed differences in nuclear import rates among myonuclei. Analyzing nuclear import throughout myogenesis revealed that cNLS and non-cNLS import varies during differentiation. Taken together, our results suggest that both spatial and temporal regulation of nuclear import pathways are important in muscle cell differentiation and protein regionalization in myofibers.
Collapse
Affiliation(s)
- Alicia A Cutler
- Department of Pharmacology, Emory University, Atlanta, GA 30322, USA.,Graduate Program in Biochemistry, Cell and Developmental Biology, Emory University, Atlanta, GA 30322, USA
| | | | - Anita H Corbett
- Department of Biology, Emory University, Atlanta, GA 30322, USA
| | - Grace K Pavlath
- Department of Pharmacology, Emory University, Atlanta, GA 30322, USA
| |
Collapse
|
31
|
Mackey AL, Kjaer M. The breaking and making of healthy adult human skeletal muscle in vivo. Skelet Muscle 2017; 7:24. [PMID: 29115986 PMCID: PMC5688812 DOI: 10.1186/s13395-017-0142-x] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Accepted: 10/23/2017] [Indexed: 01/11/2023] Open
Abstract
Background While muscle regeneration has been extensively studied in animal and cell culture models, in vivo myogenesis in adult human skeletal muscle has not been investigated in detail. Methods Using forced lengthening contractions induced by electrical stimulation, we induced myofibre injury in young healthy males. Muscle biopsies were collected from the injured leg 7 and 30 days after muscle injury and from the uninjured leg as a control. Immuno-stained single muscle fibres and muscle cross sections were studied by wide-field and confocal microscopy. Samples were also studied at the ultra-structural level by electron microscopy. Results Microscopy of single muscle fibres in 3 dimensions revealed a repeating pattern of necrotic and regenerating zones along the length of the same myofibre, characterised by extensive macrophage infiltration alongside differentiating myogenic progenitor cells and myotubes: the hallmarks of myogenesis. The myofibre basement membrane was preserved during these processes and interestingly was seen at a later stage as a second basement membrane surrounding the regenerating fibres. Conclusions This is the first work to document in vivo myogenesis in humans in detail and highlights the importance of the basement membrane in the process of regeneration. In addition, it provides insight into parallels between the regeneration of adult skeletal muscle in mouse and man, confirming that this model may be a useful tool in investigating myofibre and matrix formation, as well as specific cell types, during regeneration from the perspective of human muscle.
Collapse
Affiliation(s)
- Abigail L Mackey
- Institute of Sports Medicine Copenhagen, Department of Orthopaedic Surgery M, Bispebjerg Hospital, Copenhagen, Denmark. .,Center for Healthy Aging, Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| | - Michael Kjaer
- Institute of Sports Medicine Copenhagen, Department of Orthopaedic Surgery M, Bispebjerg Hospital, Copenhagen, Denmark.,Center for Healthy Aging, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
32
|
Banerjee A, Vest KE, Pavlath GK, Corbett AH. Nuclear poly(A) binding protein 1 (PABPN1) and Matrin3 interact in muscle cells and regulate RNA processing. Nucleic Acids Res 2017; 45:10706-10725. [PMID: 28977530 PMCID: PMC5737383 DOI: 10.1093/nar/gkx786] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Accepted: 08/27/2017] [Indexed: 01/01/2023] Open
Abstract
The polyadenylate binding protein 1 (PABPN1) is a ubiquitously expressed RNA binding protein vital for multiple steps in RNA metabolism. Although PABPN1 plays a critical role in the regulation of RNA processing, mutation of the gene encoding this ubiquitously expressed RNA binding protein causes a specific form of muscular dystrophy termed oculopharyngeal muscular dystrophy (OPMD). Despite the tissue-specific pathology that occurs in this disease, only recently have studies of PABPN1 begun to explore the role of this protein in skeletal muscle. We have used co-immunoprecipitation and mass spectrometry to identify proteins that interact with PABPN1 in mouse skeletal muscles. Among the interacting proteins we identified Matrin 3 (MATR3) as a novel protein interactor of PABPN1. The MATR3 gene is mutated in a form of distal myopathy and amyotrophic lateral sclerosis (ALS). We demonstrate, that like PABPN1, MATR3 is critical for myogenesis. Furthermore, MATR3 controls critical aspects of RNA processing including alternative polyadenylation and intron retention. We provide evidence that MATR3 also binds and regulates the levels of long non-coding RNA (lncRNA) Neat1 and together with PABPN1 is required for normal paraspeckle function. We demonstrate that PABPN1 and MATR3 are required for paraspeckles, as well as for adenosine to inosine (A to I) RNA editing of Ctn RNA in muscle cells. We provide a functional link between PABPN1 and MATR3 through regulation of a common lncRNA target with downstream impact on paraspeckle morphology and function. We extend our analysis to a mouse model of OPMD and demonstrate altered paraspeckle morphology in the presence of endogenous levels of alanine-expanded PABPN1. In this study, we report protein-binding partners of PABPN1, which could provide insight into novel functions of PABPN1 in skeletal muscle and identify proteins that could be sequestered with alanine-expanded PABPN1 in the nuclear aggregates found in OPMD.
Collapse
Affiliation(s)
- Ayan Banerjee
- Department of Biology, Emory University, Atlanta, GA 30322, USA
| | - Katherine E Vest
- Department of Pharmacology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Grace K Pavlath
- Department of Pharmacology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Anita H Corbett
- Department of Biology, Emory University, Atlanta, GA 30322, USA
| |
Collapse
|
33
|
Spatial and age-related changes in the microstructure of dystrophic and healthy diaphragms. PLoS One 2017; 12:e0183853. [PMID: 28877195 PMCID: PMC5587283 DOI: 10.1371/journal.pone.0183853] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2016] [Accepted: 08/11/2017] [Indexed: 12/21/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is a progressive degenerative disease that results in fibrosis and atrophy of muscles. The main cause of death associated with DMD is failure of the diaphragm. The diaphragm is a dome-shaped muscle with a fiber microstructure that differs across regions of the muscle. However, no studies to our knowledge have examined spatial variations of muscle fibers in dystrophic diaphragm or how aging affects those variations in DMD. In this study, diaphragms were obtained from mdx and healthy mice at ages three, seven, and ten months in the dorsal, midcostal, and ventral regions. Through immunostaining and confocal imaging, we quantified sarcomere length, interstitial space between fibers, fiber branching, fiber cross sectional area (CSA), and fiber regeneration measured by centrally located nuclei. Because DMD is associated with chronic inflammation, we also investigated the number of macrophages in diaphragm muscle cross-sections. We saw regional differences in the number of regenerating fibers and macrophages during the progression of DMD in the mdx diaphragm. Additionally, the number of regenerating fibers increased with age, while CSA and the number of branching fibers decreased. Dystrophic diaphragms had shorter sarcomere lengths than age-matched controls. Our results suggest that the dystrophic diaphragm in the mdx mouse is structurally heterogeneous and remodels non-uniformly over time. Understanding regional changes in dystrophic diaphragms over time will facilitate the development of targeted therapies to prevent or minimize respiratory failure in DMD patients.
Collapse
|
34
|
Cutler AA, Dammer EB, Doung DM, Seyfried NT, Corbett AH, Pavlath GK. Biochemical isolation of myonuclei employed to define changes to the myonuclear proteome that occur with aging. Aging Cell 2017; 16:738-749. [PMID: 28544616 PMCID: PMC5506426 DOI: 10.1111/acel.12604] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/21/2017] [Indexed: 01/03/2023] Open
Abstract
Skeletal muscle aging is accompanied by loss of muscle mass and strength. Examining changes in myonuclear proteins with age would provide insight into molecular processes which regulate these profound changes in muscle physiology. However, muscle tissue is highly adapted for contraction and thus comprised largely of contractile proteins making the nuclear proteins difficult to identify from whole muscle samples. By developing a method to purify myonuclei from whole skeletal muscle, we were able to collect myonuclei for analysis by flow cytometry, biochemistry, and mass spectrometry. Nuclear purification dramatically increased the number and intensity of nuclear proteins detected by mass spectrometry compared to whole tissue. We exploited this increased proteomic depth to investigate age-related changes to the myonuclear proteome. Nuclear levels of 54 of 779 identified proteins (7%) changed significantly with age; these proteins were primarily involved in chromatin maintenance and RNA processing. To determine whether the changes we detected were specific to myonuclei or were common to nuclei of excitatory tissues, we compared aging in myonuclei to aging in brain nuclei. Although several of the same processes were affected by aging in both brain and muscle nuclei, the specific proteins involved in these alterations differed between the two tissues. Isolating myonuclei allowed a deeper view into the myonuclear proteome than previously possible facilitating identification of novel age-related changes in skeletal muscle. Our technique will enable future studies into a heretofore underrepresented compartment of skeletal muscle.
Collapse
Affiliation(s)
- Alicia A. Cutler
- Department of Pharmacology; Emory University; Atlanta GA 30322 USA
- Graduate Program in Biochemistry, Cell and Developmental Biology; Emory University; Atlanta GA 30322 USA
| | - Eric B. Dammer
- Department of Biochemistry; Emory University; Atlanta GA 30322 USA
| | - Duc M. Doung
- Department of Biochemistry; Emory University; Atlanta GA 30322 USA
| | | | | | - Grace K. Pavlath
- Department of Pharmacology; Emory University; Atlanta GA 30322 USA
| |
Collapse
|
35
|
Choo HJ, Canner JP, Vest KE, Thompson Z, Pavlath GK. A tale of two niches: differential functions for VCAM-1 in satellite cells under basal and injured conditions. Am J Physiol Cell Physiol 2017; 313:C392-C404. [PMID: 28701357 DOI: 10.1152/ajpcell.00119.2017] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Revised: 06/27/2017] [Accepted: 07/06/2017] [Indexed: 11/22/2022]
Abstract
Cell-cell adhesion molecules play key roles in maintaining quiescence or promoting activation of various stem cells in their niche. Muscle stem cells called satellite cells (SC) are critical for skeletal muscle regeneration after injury, but little is known about the role of adhesion molecules in regulating the behavior of these stem cells. Vascular cell adhesion molecule-1 (VCAM-1) is a cell-cell adhesion protein expressed on quiescent and activated SC whose function is unknown in this context. We deleted Vcam1 from SC using an inducible Cre recombinase in young mice. In the injured niche, Vcam1-/- SC underwent premature lineage progression to a more differentiated state as well as apoptosis leading to a transient delay in myofiber growth during regeneration. Apoptosis was also increased in Vcam1-/- SC in vitro concomitant with decreased levels of phosphorylated Akt, a prosurvival signal activated by VCAM-1 signaling in other cell types. During muscle regeneration, we observed an influx of immune cells expressing α4 integrin, a component of the major, high-affinity VCAM-1 ligand, α4β1 integrin. Furthermore, α4 integrin mRNA and protein were induced in SC 2 days after injury. These results suggest that SC interact with other SC as well as immune cells through α4β1 integrin in the injured niche to promote expansion of SC. In the uninjured niche, multiple cell types also expressed α4 integrin. However, only basal fusion of Vcam1-/- SC with myofibers was decreased, contributing to decreased myofiber growth. These studies define differential roles for VCAM-1 in SC depending on the state of their niche.
Collapse
Affiliation(s)
- Hyo-Jung Choo
- Department of Pharmacology, Emory University, Atlanta, Georgia; and.,Department of Cell Biology, Emory University, Atlanta, Georgia
| | - James P Canner
- Department of Pharmacology, Emory University, Atlanta, Georgia; and
| | - Katherine E Vest
- Department of Pharmacology, Emory University, Atlanta, Georgia; and
| | - Zachary Thompson
- Department of Pharmacology, Emory University, Atlanta, Georgia; and
| | - Grace K Pavlath
- Department of Pharmacology, Emory University, Atlanta, Georgia; and
| |
Collapse
|
36
|
Rader EP, Naimo MA, Ensey J, Baker BA. Agonist muscle adaptation accompanied by antagonist muscle atrophy in the hindlimb of mice following stretch-shortening contraction training. BMC Musculoskelet Disord 2017; 18:60. [PMID: 28148306 PMCID: PMC5288976 DOI: 10.1186/s12891-017-1397-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2016] [Accepted: 01/11/2017] [Indexed: 02/26/2023] Open
Abstract
Background The vast majority of dynamometer-based animal models for investigation of the response to chronic muscle contraction exposure has been limited to analysis of isometric, lengthening, or shortening contractions in isolation. An exception to this has been the utilization of a rat model to study stretch-shortening contractions (SSCs), a sequence of consecutive isometric, lengthening, and shortening contractions common during daily activity and resistance-type exercise. However, the availability of diverse genetic strains of rats is limited. Therefore, the purpose of the present study was to develop a dynamometer-based SSC training protocol to induce increased muscle mass and performance in plantarflexor muscles of mice. Methods Young (3 months old) C57BL/6 mice were subjected to 1 month of plantarflexion SSC training. Hindlimb muscles were analyzed for muscle mass, quantitative morphology, myogenesis/myopathy relevant gene expression, and fiber type distribution. Results The main aim of the research was achieved when training induced a 2-fold increase in plantarflexion peak torque output and a 19% increase in muscle mass for the agonist plantaris (PLT) muscle. In establishing this model, several outcomes emerged which raised the value of the model past that of being a mere recapitulation of the rat model. An increase in the number of muscle fibers per transverse muscle section accounted for the PLT muscle mass gain while the antagonist tibialis anterior (TA) muscle atrophied by 30% with preferential atrophy of type IIb and IIx fibers. These alterations were accompanied by distinct gene expression profiles. Conclusions The findings confirm the development of a stretch-shortening contraction training model for the PLT muscle of mice and demonstrate that increased cross-sectional fiber number can occur following high-intensity SSC training. Furthermore, the TA muscle atrophy provides direct evidence for the concept of muscle imbalance in phasic non-weight bearing muscles, a concept largely characterized based on clinical observation of patients. The susceptibility to this imbalance is demonstrated to be selective for the type IIb and IIx muscle fiber types. Overall, the study highlights the importance of considering muscle fiber number modulation and the effect of training on surrounding muscles in exercise comprised of SSCs. Electronic supplementary material The online version of this article (doi:10.1186/s12891-017-1397-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Erik P Rader
- Centers for Disease Control and Prevention, National Institute for Occupational Safety and Health, MS L3014, 1095 Willowdale Rd, Morgantown, West Virginia, 26505, USA.
| | - Marshall A Naimo
- Centers for Disease Control and Prevention, National Institute for Occupational Safety and Health, MS L3014, 1095 Willowdale Rd, Morgantown, West Virginia, 26505, USA.,West Virginia University School of Medicine, Division of Exercise Physiology, Morgantown, West Virginia, USA
| | - James Ensey
- Centers for Disease Control and Prevention, National Institute for Occupational Safety and Health, MS L3014, 1095 Willowdale Rd, Morgantown, West Virginia, 26505, USA
| | - Brent A Baker
- Centers for Disease Control and Prevention, National Institute for Occupational Safety and Health, MS L3014, 1095 Willowdale Rd, Morgantown, West Virginia, 26505, USA
| |
Collapse
|
37
|
Iyer SR, Shah SB, Valencia AP, Schneider MF, Hernández-Ochoa EO, Stains JP, Blemker SS, Lovering RM. Altered nuclear dynamics in MDX myofibers. J Appl Physiol (1985) 2016; 122:470-481. [PMID: 27979987 DOI: 10.1152/japplphysiol.00857.2016] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2016] [Revised: 12/02/2016] [Accepted: 12/06/2016] [Indexed: 01/17/2023] Open
Abstract
Duchenne muscular dystrophy (DMD) is a genetic disorder in which the absence of dystrophin leads to progressive muscle degeneration and weakness. Although the genetic basis is known, the pathophysiology of dystrophic skeletal muscle remains unclear. We examined nuclear movement in wild-type (WT) and muscular dystrophy mouse model for DMD (MDX) (dystrophin-null) mouse myofibers. We also examined expression of proteins in the linkers of nucleoskeleton and cytoskeleton (LINC) complex, as well as nuclear transcriptional activity via histone H3 acetylation and polyadenylate-binding nuclear protein-1. Because movement of nuclei is not only LINC dependent but also microtubule dependent, we analyzed microtubule density and organization in WT and MDX myofibers, including the application of a unique 3D tool to assess microtubule core structure. Nuclei in MDX myofibers were more mobile than in WT myofibers for both distance traveled and velocity. MDX muscle shows reduced expression and labeling intensity of nesprin-1, a LINC protein that attaches the nucleus to the microtubule and actin cytoskeleton. MDX nuclei also showed altered transcriptional activity. Previous studies established that microtubule structure at the cortex is disrupted in MDX myofibers; our analyses extend these findings by showing that microtubule structure in the core is also disrupted. In addition, we studied malformed MDX myofibers to better understand the role of altered myofiber morphology vs. microtubule architecture in the underlying susceptibility to injury seen in dystrophic muscles. We incorporated morphological and microtubule architectural concepts into a simplified finite element mathematical model of myofiber mechanics, which suggests a greater contribution of myofiber morphology than microtubule structure to muscle biomechanical performance.NEW & NOTEWORTHY Microtubules provide the means for nuclear movement but show altered organization in the muscular dystrophy mouse model (MDX) (dystrophin-null) muscle. Here, MDX myofibers show increased nuclear movement, altered transcriptional activity, and altered linkers of nucleoskeleton and cytoskeleton complex expression compared with healthy myofibers. Microtubule architecture was incorporated in finite element modeling of passive stretch, revealing a role of fiber malformation, commonly found in MDX muscle. The results suggest that alterations in microtubule architecture in MDX muscle affect nuclear movement, which is essential for muscle function.
Collapse
Affiliation(s)
- Shama R Iyer
- Department of Orthopaedics, University of Maryland School of Medicine, Baltimore, Maryland
| | - Sameer B Shah
- Departments of Orthopaedic Surgery and Bioengineering, University of California San Diego, La Jolla, California
| | - Ana P Valencia
- Department of Orthopaedics, University of Maryland School of Medicine, Baltimore, Maryland
| | - Martin F Schneider
- Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, Maryland
| | - Erick O Hernández-Ochoa
- Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, Maryland
| | - Joseph P Stains
- Department of Orthopaedics, University of Maryland School of Medicine, Baltimore, Maryland
| | - Silvia S Blemker
- Department of Biomedical Engineering and Mechanical and Aerospace Engineering, University of Virginia, Charlottesville, Virginia; and
| | - Richard M Lovering
- Department of Orthopaedics, University of Maryland School of Medicine, Baltimore, Maryland; .,Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland
| |
Collapse
|
38
|
Lu-Nguyen N, Malerba A, Popplewell L, Schnell F, Hanson G, Dickson G. Systemic Antisense Therapeutics for Dystrophin and Myostatin Exon Splice Modulation Improve Muscle Pathology of Adult mdx Mice. MOLECULAR THERAPY. NUCLEIC ACIDS 2016; 6:15-28. [PMID: 28325281 PMCID: PMC5363451 DOI: 10.1016/j.omtn.2016.11.009] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Revised: 11/21/2016] [Accepted: 11/21/2016] [Indexed: 01/10/2023]
Abstract
Antisense-mediated exon skipping is a promising approach for the treatment of Duchenne muscular dystrophy (DMD), a rare life-threatening genetic disease due to dystrophin deficiency. Such an approach can restore the disrupted reading frame of dystrophin pre-mRNA, generating a truncated form of the protein. Alternatively, antisense therapy can be used to induce destructive exon skipping of myostatin pre-mRNA, knocking down myostatin expression to enhance muscle strength and reduce fibrosis. We have reported previously that intramuscular or intraperitoneal antisense administration inducing dual exon skipping of dystrophin and myostatin pre-mRNAs was beneficial in mdx mice, a mouse model of DMD, although therapeutic effects were muscle type restricted, possibly due to the delivery routes used. Here, following systemic intravascular antisense treatment, muscle strength and body activity of treated adult mdx mice increased to the levels of healthy controls. Importantly, hallmarks of muscular dystrophy were greatly improved in mice receiving the combined exon-skipping therapy, as compared to those receiving dystrophin antisense therapy alone. Our results support the translation of antisense therapy for dystrophin restoration and myostatin inhibition into the clinical setting for DMD.
Collapse
Affiliation(s)
- Ngoc Lu-Nguyen
- School of Biological Sciences, Royal Holloway-University of London, Egham, Surrey TW20 0EX, UK
| | - Alberto Malerba
- School of Biological Sciences, Royal Holloway-University of London, Egham, Surrey TW20 0EX, UK
| | - Linda Popplewell
- School of Biological Sciences, Royal Holloway-University of London, Egham, Surrey TW20 0EX, UK
| | - Fred Schnell
- Sarepta Therapeutics Inc., 215 First Street, Cambridge, MA 02142, USA
| | - Gunnar Hanson
- Sarepta Therapeutics Inc., 215 First Street, Cambridge, MA 02142, USA
| | - George Dickson
- School of Biological Sciences, Royal Holloway-University of London, Egham, Surrey TW20 0EX, UK.
| |
Collapse
|
39
|
Mazzotti AL, Coletti D. The Need for a Consensus on the Locution "Central Nuclei" in Striated Muscle Myopathies. Front Physiol 2016; 7:577. [PMID: 27932999 PMCID: PMC5120524 DOI: 10.3389/fphys.2016.00577] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Accepted: 11/10/2016] [Indexed: 01/08/2023] Open
Affiliation(s)
- Anna L Mazzotti
- Assointerpreti, Italian Association of Conference InterpretersRome, Italy; MPS Public SpeakingRome, Italy
| | - Dario Coletti
- Biology of Adaptation and Aging, CNRS, UMR 8256, INSERM ERL U1164, Institut de Biologie Paris-Seine, Université Pierre et Marie CurieParis, France; Department of Anatomical, Histological, Forensic Sciences and Orthopedics, Sapienza University of RomeRome, Italy; Interuniversity Institute of MyologyRome, Italy
| |
Collapse
|
40
|
Pichavant C, Burkholder TJ, Pavlath GK. Decrease of myofiber branching via muscle-specific expression of the olfactory receptor mOR23 in dystrophic muscle leads to protection against mechanical stress. Skelet Muscle 2016; 6:2. [PMID: 26798450 PMCID: PMC4721043 DOI: 10.1186/s13395-016-0077-7] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2015] [Accepted: 01/05/2016] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Abnormal branched myofibers within skeletal muscles are commonly found in diverse animal models of muscular dystrophy as well as in patients. Branched myofibers from dystrophic mice are more susceptible to break than unbranched myofibers suggesting that muscles containing a high percentage of these myofibers are more prone to injury. Previous studies showed ubiquitous over-expression of mouse olfactory receptor 23 (mOR23), a G protein-coupled receptor, in wild type mice decreased myofiber branching. Whether mOR23 over-expression specifically in skeletal muscle cells is sufficient to mitigate myofiber branching in dystrophic muscle is unknown. METHODS We created a novel transgenic mouse over-expressing mOR23 specifically in muscle cells and then bred with dystrophic (mdx) mice. Myofiber branching was analyzed in these two transgenic mice and membrane integrity was assessed by Evans blue dye fluorescence. RESULTS mOR23 over-expression in muscle led to a decrease of myofiber branching after muscle regeneration in non-dystrophic mouse muscles and reduced the severity of myofiber branching in mdx mouse muscles. Muscles from mdx mouse over-expressing mOR23 significantly exhibited less damage to eccentric contractions than control mdx muscles. CONCLUSIONS The decrease of myofiber branching in mdx mouse muscles over-expressing mOR23 reduced the amount of membrane damage induced by mechanical stress. These results suggest that modifying myofiber branching in dystrophic patients, while not preventing degeneration, could be beneficial for mitigating some of the effects of the disease process.
Collapse
Affiliation(s)
- Christophe Pichavant
- Department of Pharmacology, Emory University, Atlanta, GA USA ; Present address: Department of Genetics, Stanford University, Stanford, CA USA
| | - Thomas J Burkholder
- School of Applied Physiology, Georgia Institute of Technology, Atlanta, GA USA
| | - Grace K Pavlath
- Department of Pharmacology, Emory University, Atlanta, GA USA ; 1510 Clifton Road, Room 5024, Atlanta, GA 30322 USA
| |
Collapse
|
41
|
Makarevich PI, Boldyreva MA, Gluhanyuk EV, Efimenko AY, Dergilev KV, Shevchenko EK, Sharonov GV, Gallinger JO, Rodina PA, Sarkisyan SS, Hu YC, Parfyonova YV. Enhanced angiogenesis in ischemic skeletal muscle after transplantation of cell sheets from baculovirus-transduced adipose-derived stromal cells expressing VEGF165. Stem Cell Res Ther 2015; 6:204. [PMID: 26503601 PMCID: PMC4620646 DOI: 10.1186/s13287-015-0199-6] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2015] [Revised: 09/28/2015] [Accepted: 10/06/2015] [Indexed: 12/15/2022] Open
Abstract
Introduction Cell therapy using adipose-derived stromal cells (ADSC) is an intensively developing approach to promote angiogenesis and regeneration. Administration technique is crucial and among others minimal constructs - cell sheets (CS) have certain advantages. Delivery of CS allows transplantation of cells along with matrix proteins to facilitate engraftment. Cells’ therapeutic potential can be also increased by expression of proangiogenic factors by viral transduction. In this work we report on therapeutic efficacy of CS from mouse ADSC transduced to express human vascular endothelial growth factor 165 a/a isoform (VEGF165), which showed potency to restore perfusion and protect tissue in a model of limb ischemia. Methods Mouse ADSC (mADSC) isolated from C57 male mice were expanded for CS formation (106cells per CS). Constructs were transduced to express human VEGF165 by baculoviral (BV) system. CS were transplanted subcutaneously to mice with surgically induced limb ischemia and followed by laser Doppler perfusion measurements. At endpoint animals were sacrificed and skeletal muscle was evaluated for necrosis and vessel density; CS with underlying muscle was stained for apoptosis, proliferation, monocytes and blood vessels. Results Using BV system and sodium butyrate treatment we expressed human VEGF165 in mADSC (production of VEGF165 reached ≈ 25-27 ng/ml/105 cells) and optimized conditions to ensure cells’ viability after transduction. Implantation of mock-transduced CS resulted in significant improvement of limb perfusion, increased capillary density and necrosis reduction at 2 weeks post-surgery compared to untreated animals. Additional improvement of blood flow and angiogenesis was observed after transplantation of VEGF165-expressing CS indicating enhanced therapeutic potential of genetically modified constructs. Moreover, we found delivery of mADSC as CS to be superior to equivalent dose of suspended cells in terms of perfusion and angiogenesis. Histology analysis of extracted CS detected limited proliferation and approximately 10 % prevalence of apoptosis in transplanted mADSC. Significant vascularization of CS and infiltration by monocytes were found in both – BV-transduced and control CS indicating graft and host interaction after transplantation. Conclusions Delivery of ADSC by subcutaneous transplantation of CS is effective for stimulation of angiogenesis and tissue protection in limb ischemia with a potential for efficacy improvement by BV transduction to express VEGF165. Electronic supplementary material The online version of this article (doi:10.1186/s13287-015-0199-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Pavel I Makarevich
- Laboratory of Angiogenesis, Russian Cardiology Research and Production Complex, Moscow, 121552, Russia. .,Laboratory of Regenerative Medicine, Medical Research and Educational Centre of Moscow State University, Moscow, 119192, Russia.
| | - Maria A Boldyreva
- Laboratory of Angiogenesis, Russian Cardiology Research and Production Complex, Moscow, 121552, Russia.
| | - Evgeny V Gluhanyuk
- Laboratory of Angiogenesis, Russian Cardiology Research and Production Complex, Moscow, 121552, Russia. .,Faculty of Medicine, M.V. Lomonosov Moscow State University, Moscow, 119192, Russia.
| | - Anastasia Yu Efimenko
- Laboratory of Regenerative Medicine, Medical Research and Educational Centre of Moscow State University, Moscow, 119192, Russia. .,Faculty of Medicine, M.V. Lomonosov Moscow State University, Moscow, 119192, Russia.
| | - Konstantin V Dergilev
- Laboratory of Angiogenesis, Russian Cardiology Research and Production Complex, Moscow, 121552, Russia.
| | - Evgeny K Shevchenko
- Laboratory of Angiogenesis, Russian Cardiology Research and Production Complex, Moscow, 121552, Russia.
| | - Georgy V Sharonov
- Faculty of Medicine, M.V. Lomonosov Moscow State University, Moscow, 119192, Russia.
| | - Julia O Gallinger
- Faculty of Medicine, M.V. Lomonosov Moscow State University, Moscow, 119192, Russia.
| | - Polina A Rodina
- Faculty of Medicine, M.V. Lomonosov Moscow State University, Moscow, 119192, Russia.
| | - Stepan S Sarkisyan
- Faculty of Medicine, M.V. Lomonosov Moscow State University, Moscow, 119192, Russia.
| | - Yu-Chen Hu
- Department of Chemical Engineering, National Tsing Hua University, Hsinchu, 300, Taiwan.
| | - Yelena V Parfyonova
- Laboratory of Angiogenesis, Russian Cardiology Research and Production Complex, Moscow, 121552, Russia. .,Faculty of Medicine, M.V. Lomonosov Moscow State University, Moscow, 119192, Russia.
| |
Collapse
|
42
|
Grounds MD, Pinniger GJ. What is the mechanism for in vivo loss of skeletal muscle function in elderly women? Acta Physiol (Oxf) 2015; 215:9-12. [PMID: 26132503 DOI: 10.1111/apha.12547] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- M. D. Grounds
- School of Anatomy, Physiology and Human Biology; The University of Western Australia; Perth WA Australia
| | - G. J. Pinniger
- School of Anatomy, Physiology and Human Biology; The University of Western Australia; Perth WA Australia
| |
Collapse
|
43
|
Ackermann MA, Ward CW, Gurnett C, Kontrogianni-Konstantopoulos A. Myosin Binding Protein-C Slow Phosphorylation is Altered in Duchenne Dystrophy and Arthrogryposis Myopathy in Fast-Twitch Skeletal Muscles. Sci Rep 2015; 5:13235. [PMID: 26287277 PMCID: PMC4642557 DOI: 10.1038/srep13235] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2015] [Accepted: 05/05/2015] [Indexed: 11/09/2022] Open
Abstract
Myosin Binding Protein-C slow (sMyBP-C), encoded by MYBPC1, comprises a family of regulatory proteins of skeletal muscles that are phosphorylated by PKA and PKC. MYBPC1 missense mutations are linked to the development of Distal Arthrogryposis-1 (DA-1). Although structure-function details for this myopathy are evolving, function is undoubtedly driven by sequence variations and post-translational modifications in sMyBP-C. Herein, we examined the phosphorylation profile of sMyBP-C in mouse and human fast-twitch skeletal muscles. We used Flexor Digitorum Brevis (FDB) isolated from young (~2-months old) and old (~14-months old) wild type and mdx mice, and human Abductor Hallucis (AH) and gastrocnemious muscles carrying the DA-1 mutations. Our results indicate both constitutive and differential phosphorylation of sMyBP-C in aged and diseased muscles. We report a 7-35% reduction in the phosphorylation levels of select sites in old wild type and young or old mdx FDB mouse muscles, compared to young wild type tissue. Similarly, we observe a 30-70% decrease in the phosphorylation levels of all PKA and PKC phospho-sites in the DA-1 AH, but not gastrocnemius, muscle. Overall, our studies show that the phosphorylation pattern of sMyBP-C is differentially regulated in response to age and disease, suggesting that phosphorylation plays important roles in these processes.
Collapse
Affiliation(s)
- Maegen A Ackermann
- University of Maryland, School of Medicine, Department of Biochemistry and Molecular Biology, Baltimore, MD, USA
| | | | - Christina Gurnett
- Washington University, School of Medicine, Department of Neurology, St. Louis, MO, USA
| | | |
Collapse
|
44
|
Duddy W, Duguez S, Johnston H, Cohen TV, Phadke A, Gordish-Dressman H, Nagaraju K, Gnocchi V, Low S, Partridge T. Muscular dystrophy in the mdx mouse is a severe myopathy compounded by hypotrophy, hypertrophy and hyperplasia. Skelet Muscle 2015; 5:16. [PMID: 25987977 PMCID: PMC4434871 DOI: 10.1186/s13395-015-0041-y] [Citation(s) in RCA: 95] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2015] [Accepted: 04/16/2015] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Preclinical testing of potential therapies for Duchenne muscular dystrophy (DMD) is conducted predominantly of the mdx mouse. But lack of a detailed quantitative description of the pathology of this animal limits our ability to evaluate the effectiveness of putative therapies or their relevance to DMD. METHODS Accordingly, we have measured the main cellular components of muscle growth and regeneration over the period of postnatal growth and early pathology in mdx and wild-type (WT) mice; phalloidin binding is used as a measure of fibre size, myonuclear counts and BrdU labelling as records of myogenic activity. RESULTS We confirm a two-phase postnatal growth pattern in WT muscle: first, increase in myonuclear number over weeks 1 to 3, then expansion of myonuclear domain. Mdx muscle growth lags behind that of WT prior to overt signs of pathology. Fibres are smaller, with fewer myonuclei and smaller myonuclear domains. Moreover, satellite cells are more readily detached from mdx than WT muscle fibres. At 3 weeks, mdx muscles enter a phase of florid myonecrosis, accompanied by concurrent regeneration of an intensity that results in complete replacement of pre-existing muscle over the succeeding 3 to 4 weeks. Both WT and mdx muscles attain maximum size by 12 to 14 weeks, mdx muscle fibres being up to 50% larger than those of WT as they become increasingly branched. Mdx muscle fibres also become hypernucleated, containing twice as many myonuclei per sarcoplasmic volume, as those of WT, the excess corresponding to the number of centrally placed myonuclei. CONCLUSIONS The best-known consequence of lack of dystrophin that is common to DMD and the mdx mouse is the conspicuous necrosis and regeneration of muscle fibres. We present protocols for measuring this in terms both of loss of muscle nuclei previously labelled with BrdU and of the intensity of myonuclear labelling with BrdU administered during the regeneration period. Both measurements can be used to assess the efficacy of putative antinecrotic agents. We also show that lack of dystrophin is associated with a number of previously unsuspected abnormalities of muscle fibre structure and function that do not appear to be directly associated with myonecrosis.
Collapse
Affiliation(s)
- William Duddy
- Center for Genetic Medicine Research, Children's National Medical Center, 111 Michigan Avenue NW, Washington DC, 20010 USA ; Myology Center of Research, Institut de Myologie Pitié-Salpétrière - Bâtiment Babinski, 75651 Paris Cedex 13, France
| | - Stephanie Duguez
- Center for Genetic Medicine Research, Children's National Medical Center, 111 Michigan Avenue NW, Washington DC, 20010 USA ; Myology Center of Research, Institut de Myologie Pitié-Salpétrière - Bâtiment Babinski, 75651 Paris Cedex 13, France
| | - Helen Johnston
- Center for Genetic Medicine Research, Children's National Medical Center, 111 Michigan Avenue NW, Washington DC, 20010 USA
| | - Tatiana V Cohen
- Center for Genetic Medicine Research, Children's National Medical Center, 111 Michigan Avenue NW, Washington DC, 20010 USA ; Center for Genetic Muscle Disorders, Kennedy Krieger Institute, 801 N. Broadway, Baltimore, MD 21205 USA
| | - Aditi Phadke
- Center for Genetic Medicine Research, Children's National Medical Center, 111 Michigan Avenue NW, Washington DC, 20010 USA
| | - Heather Gordish-Dressman
- Center for Genetic Medicine Research, Children's National Medical Center, 111 Michigan Avenue NW, Washington DC, 20010 USA
| | - Kanneboyina Nagaraju
- Center for Genetic Medicine Research, Children's National Medical Center, 111 Michigan Avenue NW, Washington DC, 20010 USA
| | - Viola Gnocchi
- Center for Genetic Medicine Research, Children's National Medical Center, 111 Michigan Avenue NW, Washington DC, 20010 USA
| | - SiewHui Low
- Department of Embryology, Carnegie Institution for Science, Baltimore, MD 21218 USA
| | - Terence Partridge
- Center for Genetic Medicine Research, Children's National Medical Center, 111 Michigan Avenue NW, Washington DC, 20010 USA
| |
Collapse
|
45
|
Soffe Z, Radley-Crabb HG, McMahon C, Grounds MD, Shavlakadze T. Effects of loaded voluntary wheel exercise on performance and muscle hypertrophy in young and old male C57Bl/6J mice. Scand J Med Sci Sports 2015; 26:172-88. [PMID: 25653015 DOI: 10.1111/sms.12416] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/23/2014] [Indexed: 01/05/2023]
Abstract
This study compared the capacity of young and old male C57Bl/6J mice to exercise with increasing resistance over 10 weeks, and its impact on muscle mass. Young mice (aged 15-25 weeks) were subjected to low (LR) and high (HR) resistance exercise, whereas only LR was used for old mice (107-117 weeks). Weekly patterns of voluntary wheel activity, food consumption and body weights were measured. Running patterns changed over time and with age, with two peaks of activity detected for young, but only one for old mice: speed and distance run was also less for old mice. The mass for six limb muscles was measured at the end of the experiment. The most pronounced increase in mass in response to exercise was for the soleus in young and old mice, and also quadriceps and gastrocnemius in young mice. Soleus and quadriceps muscles were analyzed histologically for myofiber number and size. A striking feature was the many small myofibers in response to exercise in young (but not old) soleus, whereas these were not present after exercise in young or old quadriceps. Overall, there was a striking difference in response to exercise between muscles and this was influenced by age.
Collapse
Affiliation(s)
- Z Soffe
- School of Anatomy, Physiology and Human Biology, the University of Western Australia, Nedlands, Western Australia, Australia
| | - H G Radley-Crabb
- School of Anatomy, Physiology and Human Biology, the University of Western Australia, Nedlands, Western Australia, Australia.,School of Biomedical Sciences, CHIRI Biosciences Research Precinct, Curtin University, Bentley, Western Australia, Australia
| | - C McMahon
- Developmental Biology Group, Agresearch Ltd, Hamilton, New Zealand
| | - M D Grounds
- School of Anatomy, Physiology and Human Biology, the University of Western Australia, Nedlands, Western Australia, Australia
| | - T Shavlakadze
- School of Anatomy, Physiology and Human Biology, the University of Western Australia, Nedlands, Western Australia, Australia.,Developmental Biology Group, Agresearch Ltd, Hamilton, New Zealand
| |
Collapse
|
46
|
Abstract
Muscle cells have an elaborate plasma membrane and t-tubule system that has been evolutionarily refined to maximize electrical conductivity for synchronous muscle contraction. However, this elaborate plasma membrane network has intrinsic vulnerabilities to stretch-induced membrane injury, and thus requires ongoing maintenance and repair. Herein we discuss the types of membrane injuries encountered by myofibers in healthy muscle and in muscular dystrophy. We review the different mechanisms by which muscle fibers in patients with muscular dystrophy are rendered more susceptible to injury, and we summarize the latest developments in our understanding of how the muscular dystrophy protein dysferlin mediates satellite-cell independent membrane repair.
Collapse
Affiliation(s)
- Sandra T Cooper
- Institute for Neuroscience and Muscle Research, Kids Research Institute, The Children's Hospital at Westmead, Sydney, New South Wales, Australia Discipline of Paediatrics and Child Health, Faculty of Medicine, University of Sydney, New South Wales, Australia
| | - Stewart I Head
- School of Medical Sciences, University of New South Wales, Sydney, New South Wales, Australia
| |
Collapse
|
47
|
Boldrin L, Zammit PS, Morgan JE. Satellite cells from dystrophic muscle retain regenerative capacity. Stem Cell Res 2014; 14:20-9. [PMID: 25460248 PMCID: PMC4305370 DOI: 10.1016/j.scr.2014.10.007] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2014] [Revised: 10/10/2014] [Accepted: 10/23/2014] [Indexed: 12/17/2022] Open
Abstract
Duchenne muscular dystrophy is an inherited disorder that is characterized by progressive skeletal muscle weakness and wasting, with a failure of muscle maintenance/repair mediated by satellite cells (muscle stem cells). The function of skeletal muscle stem cells resident in dystrophic muscle may be perturbed by being in an increasing pathogenic environment, coupled with constant demands for repairing muscle. To investigate the contribution of satellite cell exhaustion to this process, we tested the functionality of satellite cells isolated from the mdx mouse model of Duchenne muscular dystrophy. We found that satellite cells derived from young mdx mice contributed efficiently to muscle regeneration within our in vivo mouse model. To then test the effects of long-term residence in a dystrophic environment, satellite cells were isolated from aged mdx muscle. Surprisingly, they were as functional as those derived from young or aged wild type donors. Removing satellite cells from a dystrophic milieu reveals that their regenerative capacity remains both intact and similar to satellite cells derived from healthy muscle, indicating that the host environment is critical for controlling satellite cell function. Grafted mdx satellite cells regenerate muscle as well as wild-type satellite cells. Aged mdx myofibers bear more satellite cells than aged wild type fibers. mdx satellite cells retain their ability to activate. Aged mdx satellite cells are robustly regenerative in vivo.
Collapse
MESH Headings
- Aging
- Animals
- Cells, Cultured
- Disease Models, Animal
- Mice
- Mice, Inbred C57BL
- Mice, Inbred mdx
- Mice, Nude
- Muscles/pathology
- Muscles/physiology
- Muscular Dystrophy, Duchenne/metabolism
- Muscular Dystrophy, Duchenne/pathology
- Muscular Dystrophy, Duchenne/therapy
- Regeneration
- Satellite Cells, Skeletal Muscle/cytology
- Satellite Cells, Skeletal Muscle/metabolism
- Satellite Cells, Skeletal Muscle/transplantation
Collapse
Affiliation(s)
- Luisa Boldrin
- University College London, Institute of Child Health, The Dubowitz Neuromuscular Centre, Molecular Neurosciences Section, Developmental Neurosciences Programme, 30 Guilford Street, London WC1N 1EH, United Kingdom.
| | - Peter S Zammit
- King's College London, The Randall Division of Cell and Molecular Biophysics, New Hunt's House, London SE1 1UL, United Kingdom
| | - Jennifer E Morgan
- University College London, Institute of Child Health, The Dubowitz Neuromuscular Centre, Molecular Neurosciences Section, Developmental Neurosciences Programme, 30 Guilford Street, London WC1N 1EH, United Kingdom.
| |
Collapse
|
48
|
The need to more precisely define aspects of skeletal muscle regeneration. Int J Biochem Cell Biol 2014; 56:56-65. [PMID: 25242742 DOI: 10.1016/j.biocel.2014.09.010] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2014] [Revised: 09/04/2014] [Accepted: 09/08/2014] [Indexed: 12/11/2022]
Abstract
A more precise definition of the term 'skeletal muscle regeneration' is required to reduce confusion and misconceptions. In this paper the term is used only for events that follow myofibre necrosis, to result in myogenesis and new muscle formation: other key events include early inflammation and revascularisation, and later fibrosis and re-innervation. The term 'muscle regeneration' is sometimes used casually for situations that do not involve myonecrosis; such as restoration of muscle mass by hypertrophy after atrophy, and other forms of damage to muscle tissue components. These situations are excluded from the definition in this paper which is focussed on mammalian muscles with the long-term aim of clinical translation to enhance new muscle formation after acute or chronic injury or during surgery to replace whole muscles. The paper briefly outlines the cellular events involved in myogenesis during development and post-natal muscle growth, discusses the role of satellite cells in mature normal muscles, and the likely incidence of myofibre necrosis/regeneration in healthy ageing mammals (even when subjected to exercise). The importance of the various components of regeneration is outlined to emphasise that problems in each of these aspects can influence overall new muscle formation; thus care is needed for correct interpretation of altered kinetics. Various markers used to identify regenerating myofibres are critically discussed and, since these can all occur in other conditions, caution is required for accurate interpretation of these cellular events. Finally, clinical situations are outlined where there is a need to enhance skeletal muscle regeneration: these include acute and chronic injuries or transplantation with bioengineering to form new muscles, therapeutic approaches to muscular dystrophies, and comment on proposed stem cell therapies to reduce age-related loss of muscle mass and function. This article is part of a directed issue entitled: Regenerative Medicine: the challenge of translation.
Collapse
|
49
|
Grounds MD. Therapies for sarcopenia and regeneration of old skeletal muscles: more a case of old tissue architecture than old stem cells. BIOARCHITECTURE 2014; 4:81-7. [PMID: 25101758 DOI: 10.4161/bioa.29668] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Age related loss of skeletal muscle mass and function (sarcopenia) reduces independence and the quality of life for individuals, and leads to falls and fractures with escalating health costs for the rapidly aging human population. Thus there is much interest in developing interventions to reduce sarcopenia. One area that has attracted recent attention is the proposed use of myogenic stem cells to improve regeneration of old muscles. This mini-review challenges the fundamental need for myogenic stem cell therapy for sarcopenia. It presents evidence that demonstrates the excellent capacity of myogenic stem cells from very old rodent and human muscles to form new muscles after experimental myofiber necrosis. The many factors required for successful muscle regeneration are considered with a strong focus on integration of components of old muscle bioarchitecture. The fundamental role of satellite cells in homeostasis of normal aging muscles and the incidence of endogenous regeneration in old muscles is questioned. These issues, combined with problems for clinical myogenic stem cell therapies for severe muscle diseases, raise fundamental concerns about the justification for myogenic stem cell therapy for sarcopenia.
Collapse
Affiliation(s)
- Miranda D Grounds
- School of Anatomy, Physiology and Human Biology; University of Western Australia; Crawley, Australia
| |
Collapse
|