1
|
Xiang X, Huang L, Luo W, Qin L, Bian M, Chen W, Han G, Wang N, Mo G, Zhang C, Zhang Y, Yang H, Lu S, Zhang J, Fu T. Neuromuscular electrical stimulation alleviates stroke-related sarcopenia by promoting satellite cells myogenic differentiation via AMPK-ULK1-Autophagy axis. J Orthop Translat 2025; 52:249-264. [PMID: 40342549 PMCID: PMC12059223 DOI: 10.1016/j.jot.2025.03.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Revised: 03/11/2025] [Accepted: 03/31/2025] [Indexed: 05/11/2025] Open
Abstract
Background Stroke-related sarcopenia can result in muscle mass loss and muscle fibers abnormality, significantly affecting muscle function. The clinical management of stroke-related sarcopenia still requires further research and investigation. This study aims to explore a promising therapy to restore muscle function and promote muscle regeneration in stroke-related sarcopenia, providing a new theory for stroke-related sarcopenia treatment. Methods Stroke-related sarcopenia rat model was established by using permanent middle cerebral artery occlusion (pMCAO) rat and treated with neuromuscular electrical stimulation (NMES). Electrical stimulation (ES) treatment in vitro was mimicked to test the effect of NMES on muscle regeneration in rat skeletal muscle satellite cells (MuSCs). Catwalk, H&E and Masson's trichrome staining, immunofluorescence, transcriptomic analysis, transmission electron microscopy, MuSCs transfection, autophagy flux detection, quantitative real-time PCR analysis, Co-Immunoprecipitation and Western Blot were used to investigate the role of NMES and its mechanism in stroke-related sarcopenia in vivo. Results After NMES treatment, muscle mass and myogenic differentiation were significantly increased in stroke-related sarcopenia rats. The NMES group had more stable gait, neater footprints, higher muscle wet weight, more voluminous morphology and more regenerated muscle fibers. Additionally, ES treatment induced myogenic differentiation in rat MuSCs in vitro. Transcriptomic analysis also showed that "AMPK signaling pathway" was enriched and genes upregulated in ES-treated cells, revealing ES treatment could activate the autophagy in an AMPK-ULK1-dependent mechanism in MuSCs. Besides, it was also founded that infusion of AMPK or ULK1 inhibitor, knockdown of AMPK or ULK1 in MuSCs could block the effect of myotube formation of ES. Conclusion NMES not only restores muscle function but also enhances myogenic activity and muscle regeneration via AMPK-ULK1 autophagy in stroke-related sarcopenia rats. Our study provides a promising strategy for the treatment of stroke-related sarcopenia. The translational potential of this article This study first demonstrates that NMES alleviates stroke-related sarcopenia by promoting MuSCs differentiation through AMPK-ULK1-autophagy axis. The findings reveal a novel therapeutic mechanism, suggesting that NMES can restore muscle function and enhance regeneration in stroke patients. By combining NMES with MuSCs-based therapies, this approach offers a promising strategy for clinical rehabilitation, potentially improving muscle mass and function in stroke survivors. The translational potential lies in its applicability to non-invasive, cost-effective treatments for sarcopenia, enhancing patients' quality of life.
Collapse
Affiliation(s)
- Xingdong Xiang
- Department of Rehabilitation Medicine, Zhongshan Hospital Fudan University, Shanghai, 200032, China
| | - Lei Huang
- Department of Orthopedic Surgery, Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, China
| | - Wenchen Luo
- Department of Anesthesiology, Zhongshan Hospital Fudan University, Shanghai, 200032, China
| | - Lieyang Qin
- Department of Cardiovascular Surgery, Zhongshan Hospital Fudan University, Shanghai, 200032, China
| | - Mengxuan Bian
- Department of Orthopedic Surgery, Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, China
| | - Weisin Chen
- Department of Orthopedic Surgery, Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, China
| | - Guanjie Han
- Department of Orthopedic Surgery, Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, China
| | - Ning Wang
- Department of Orthopedic Surgery, Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, China
| | - Guokang Mo
- Department of Orthopedic Surgery, Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, China
| | - Cheng Zhang
- Department of Orthopedic Surgery, Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, China
| | - Yongxing Zhang
- Department of Orthopedic Surgery, Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, China
| | - Huilin Yang
- Department of Orthopedic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, 215123, China
| | - Shunyi Lu
- Department of Orthopedic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, 215123, China
| | - Jian Zhang
- Department of Rehabilitation Medicine, Zhongshan Hospital Fudan University, Shanghai, 200032, China
| | - Tengfei Fu
- Department of Rehabilitation Medicine, Zhongshan Hospital Fudan University, Shanghai, 200032, China
| |
Collapse
|
2
|
Hernández‐Camacho JD, Vicente‐García C, Ardila‐García L, Padilla‐Campos A, López‐Lluch G, Santos‐Ocaña C, Zammit PS, Carvajal JJ, Navas P, Fernández‐Ayala DJ. Prenatal and progressive coenzyme Q 10 administration to mitigate muscle dysfunction in mitochondrial disease. J Cachexia Sarcopenia Muscle 2024; 15:2402-2416. [PMID: 39354863 PMCID: PMC11634497 DOI: 10.1002/jcsm.13574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Revised: 07/22/2024] [Accepted: 07/29/2024] [Indexed: 10/03/2024] Open
Abstract
BACKGROUND ADCK genes encode aarF domain-containing mitochondrial kinases involved in coenzyme Q (CoQ) biosynthesis and regulation. Haploinsufficiency of ADCK2 in humans leads to adult-onset physical incapacity with reduced mitochondrial CoQ levels in skeletal muscle, resulting in mitochondrial myopathy and alterations in fatty acid β-oxidation. The sole current treatment for CoQ deficiencies is oral administration of CoQ10, which causes only partial recovery with postnatal treatment, underscoring the importance of early diagnosis for successful intervention. METHODS We used Adck2 heterozygous mice to examine the influence of this gene on muscle structure, function and regeneration throughout development, growth and ageing. This investigation involved techniques including immunohistochemistry, analysis of CoQ levels, mitochondrial respiratory content, muscle transcriptome analysis and functional tests. RESULTS We demonstrated that Adck2 heterozygous mice exhibit defects from embryonic development, particularly in skeletal muscle (1102 genes deregulated). Adck2 heterozygous embryos were 7% smaller in size and displayed signs of delayed development. Prenatal administration of CoQ10 could mitigate these embryonic defects. Heterozygous Adck2 mice also showed a decrease in myogenic cell differentiation, with more severe consequences in 'aged' mice (41.63% smaller) (P < 0.01). Consequently, heterozygous Adck2 mice displayed accelerated muscle wasting associated with ageing in muscle structure (P < 0.05), muscle function (less grip strength capacity) (P < 0.001) and muscle mitochondrial respiration (P < 0.001). Furthermore, progressive CoQ10 administration conferred protective effects on mitochondrial function (P < 0.0001) and skeletal muscle (P < 0.05). CONCLUSIONS Our work uncovered novel aspects of CoQ deficiencies, revealing defects during embryonic development in mammals for the first time. Additionally, we identified the gradual establishment and progression of the deleterious Adck2 mouse phenotype. Importantly, CoQ10 supplementation demonstrated a protective effect when initiated during development.
Collapse
Affiliation(s)
- Juan Diego Hernández‐Camacho
- Centro Andaluz de Biología del Desarrollo—CSICUniversidad Pablo de OlavideSevilleSpain
- CIBERERInstituto de Salud Carlos IIIMadridSpain
| | | | - Lorena Ardila‐García
- Centro Andaluz de Biología del Desarrollo—CSICUniversidad Pablo de OlavideSevilleSpain
| | - Ana Padilla‐Campos
- Centro Andaluz de Biología del Desarrollo—CSICUniversidad Pablo de OlavideSevilleSpain
| | - Guillermo López‐Lluch
- Centro Andaluz de Biología del Desarrollo—CSICUniversidad Pablo de OlavideSevilleSpain
- CIBERERInstituto de Salud Carlos IIIMadridSpain
| | - Carlos Santos‐Ocaña
- Centro Andaluz de Biología del Desarrollo—CSICUniversidad Pablo de OlavideSevilleSpain
- CIBERERInstituto de Salud Carlos IIIMadridSpain
| | - Peter S. Zammit
- Randall Centre for Cell and Molecular BiophysicsKing's College LondonLondonUK
| | - Jaime J. Carvajal
- Centro Andaluz de Biología del Desarrollo—CSICUniversidad Pablo de OlavideSevilleSpain
| | - Plácido Navas
- Centro Andaluz de Biología del Desarrollo—CSICUniversidad Pablo de OlavideSevilleSpain
- CIBERERInstituto de Salud Carlos IIIMadridSpain
| | - Daniel J.M. Fernández‐Ayala
- Centro Andaluz de Biología del Desarrollo—CSICUniversidad Pablo de OlavideSevilleSpain
- CIBERERInstituto de Salud Carlos IIIMadridSpain
| |
Collapse
|
3
|
Zhu GZ, Zhao K, Li HZ, Wu DZ, Chen YB, Han D, Gao JW, Chen XY, Yu YP, Huang ZW, Tu C, Zhong ZM. Melatonin ameliorates age-related sarcopenia by inhibiting fibrogenic conversion of satellite cell. Mol Med 2024; 30:238. [PMID: 39614149 DOI: 10.1186/s10020-024-00998-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Accepted: 11/13/2024] [Indexed: 12/01/2024] Open
Abstract
The fibrogenic conversion of satellite cells contributes to the atrophy and fibrosis of skeletal muscle, playing a significant role in the pathogenesis of age-related sarcopenia. Melatonin, a hormone secreted by the pineal gland, exhibits anti-aging and anti-fibrotic effects in various conditions. However, the effect of melatonin on satellite cell fate and age-related sarcopenia remains under-explored. Here, we report that melatonin treatment mitigated the loss of muscle mass and strength in aged mice, replenished the satellite cell pool and curtailed muscle fibrosis. When primary SCs were cultured in vitro and subjected to aging induction via D-galactose, they exhibited a diminished myogenic potential and a conversion from myogenic to fibrogenic lineage. Notably, melatonin treatment effectively restored the myogenic potential and inhibited this lineage conversion. Furthermore, melatonin attenuated the expression of the fibrogenic cytokine, transforming growth factor-β1, and reduced the phosphorylation of its downstream targets Smad2/3 both in vivo and in vitro. In summary, our findings show melatonin's capacity to counteract muscle decline and inhibit fibrogenic conversion in aging SCs and highlight its potential therapeutic value for age-related sarcopenia.
Collapse
Affiliation(s)
- Guo-Zheng Zhu
- Division of Spine Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, 1838 North Guangzhou Ave, Guangzhou, 510515, People's Republic of China
- Department of Orthopaedics, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Kai Zhao
- Department of Orthopaedics, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Hong-Zhou Li
- Division of Spine Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, 1838 North Guangzhou Ave, Guangzhou, 510515, People's Republic of China
| | - Di-Zheng Wu
- Division of Spine Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, 1838 North Guangzhou Ave, Guangzhou, 510515, People's Republic of China
| | - Yun-Biao Chen
- Division of Spine Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, 1838 North Guangzhou Ave, Guangzhou, 510515, People's Republic of China
| | - Dong Han
- Division of Spine Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, 1838 North Guangzhou Ave, Guangzhou, 510515, People's Republic of China
| | - Jia-Wen Gao
- Division of Spine Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, 1838 North Guangzhou Ave, Guangzhou, 510515, People's Republic of China
| | - Xing-Yu Chen
- Division of Spine Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, 1838 North Guangzhou Ave, Guangzhou, 510515, People's Republic of China
| | - Yong-Peng Yu
- Division of Spine Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, 1838 North Guangzhou Ave, Guangzhou, 510515, People's Republic of China
| | - Zhi-Wei Huang
- Division of Spine Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, 1838 North Guangzhou Ave, Guangzhou, 510515, People's Republic of China
| | - Chen Tu
- Division of Spine Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, 1838 North Guangzhou Ave, Guangzhou, 510515, People's Republic of China
| | - Zhao-Ming Zhong
- Division of Spine Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, 1838 North Guangzhou Ave, Guangzhou, 510515, People's Republic of China.
| |
Collapse
|
4
|
Han D, Chen YB, Zhao K, Li HZ, Chen XY, Zhu GZ, Tu C, Gao JW, Zhuang JS, Wu ZY, Zhong ZM. Tanshinone IIA alleviates inflammation-induced skeletal muscle atrophy by regulating mitochondrial dysfunction. Arch Biochem Biophys 2024; 762:110215. [PMID: 39547552 DOI: 10.1016/j.abb.2024.110215] [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: 06/14/2024] [Revised: 11/11/2024] [Accepted: 11/13/2024] [Indexed: 11/17/2024]
Abstract
Skeletal muscle atrophy, characterized by loss of muscle mass and function, is often linked to systemic inflammation. Tanshinone IIA (Tan IIA), a major active constituent of Salvia miltiorrhiza, has anti-inflammatory and antioxidant properties. However, the effect of Tan IIA on inflammation-induced skeletal muscle atrophy remains unclear. Here, a mice model of the inflammatory muscle atrophy was established using lipopolysaccharide (LPS). Tan IIA intervention significantly increased muscle mass and strength, improved muscle fiber size, and maintained the integrity of skeletal muscle mitochondrial morphology in LPS-treated mice. Myotubes derived from myosatellite cells (MUSCs) were exposed to LPS in vitro. Tan IIA treatment inhibited LPS-induced muscle protein degradation and increased myotube diameter. Notably, Tan IIA attenuated LPS-induced inflammatory response and hyperactive mitophagy both in vivo and in vitro. In addition, Tan IIA treatment effectively diminished oxidative stress, inhibited the accumulation of mitochondrial reactive oxygen species (mtROS), and attenuated mitochondrial fission in LPS-treated myotubes. Reducing mtROS production helped to inhibit LPS-induced excessive mitophagy and myotubes atrophy. Together, our results reveal that Tan IIA can protect against inflammation-induced skeletal muscle atrophy by regulating mitochondrial dysfunction, presenting innovative potential therapeutics for skeletal muscle atrophy.
Collapse
Affiliation(s)
- Dong Han
- Division of Spine Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yun-Biao Chen
- Division of Spine Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Kai Zhao
- Department of Orthopaedics, The First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Hong-Zhou Li
- Division of Spine Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xing-Yu Chen
- Division of Spine Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Guo-Zheng Zhu
- Division of Spine Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Chen Tu
- Division of Spine Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jia-Wen Gao
- Division of Spine Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jing-Shen Zhuang
- Division of Spine Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Zhi-Yong Wu
- Department of Traditional Chinese Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Zhao-Ming Zhong
- Division of Spine Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, China.
| |
Collapse
|
5
|
Khan HA, Van Hateren N, Borycki AG. Light-Sheet Microscopy Enables Three-Dimensional Fluorescence Imaging and Live Imaging of Satellite Cells on Skeletal Muscle Fibers. Methods Mol Biol 2024. [PMID: 38997538 DOI: 10.1007/7651_2024_552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/14/2024]
Abstract
The ex vivo myofiber culture system has proven to be a useful methodology to explore the biology and behavior of satellite cells within their niche environment. However, a limitation of this system is that myofibers and their associated satellite cells are commonly examined using conventional fluorescence microscopy, which renders a three-dimensional system into two-dimensional imaging, leading to the loss of precious information or misleading interpretation of observations. Here, we report on the use of light-sheet fluorescence microscopy to generate three-dimensional and live imaging of satellite cells on myofibers. Light-sheet microscopy offers high imaging speed and good spatial resolution with minimal photo-bleaching, allowing live imaging and three-dimensional acquisition of skeletal muscle fiber specimen. The potentials of this technology are wide, ranging from the visualization of satellite cell behavior such as cell division and cell migration to imaging the sub-cellular localization of proteins or organelles.
Collapse
Affiliation(s)
- Hira Asif Khan
- School of Biosciences, University of Sheffield, Firth Court, Western Bank, Sheffield, UK
- Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, UK
| | - Nick Van Hateren
- The Wolfson Light Microscopy Facility, University of Sheffield, Firth Court, Western Bank, Sheffield, UK
| | - Anne-Gaëlle Borycki
- School of Biosciences, University of Sheffield, Firth Court, Western Bank, Sheffield, UK.
| |
Collapse
|
6
|
Meinhold M, Verbrugge S, Shi A, Schönfelder M, Becker L, Jaspers RT, Zammit PS, Wackerhage H. Yap/Taz activity is associated with increased expression of phosphoglycerate dehydrogenase that supports myoblast proliferation. Cell Tissue Res 2024; 395:271-283. [PMID: 38183459 PMCID: PMC10904560 DOI: 10.1007/s00441-023-03851-w] [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: 05/17/2023] [Accepted: 11/24/2023] [Indexed: 01/08/2024]
Abstract
In skeletal muscle, the Hippo effector Yap promotes satellite cell, myoblast, and rhabdomyoblast proliferation but prevents myogenic differentiation into multinucleated muscle fibres. We previously noted that Yap drives expression of the first enzyme of the serine biosynthesis pathway, phosphoglycerate dehydrogenase (Phgdh). Here, we examined the regulation and function of Phgdh in satellite cells and myoblasts and found that Phgdh protein increased during satellite cell activation. Analysis of published data reveal that Phgdh mRNA in mouse tibialis anterior muscle was highly expressed at day 3 of regeneration after cardiotoxin injection, when markers of proliferation are also robustly expressed and in the first week of synergist-ablated muscle. Finally, siRNA-mediated knockdown of PHGDH significantly reduced myoblast numbers and the proliferation rate. Collectively, our data suggest that Phgdh is a proliferation-enhancing metabolic enzyme that is induced when quiescent satellite cells become activated.
Collapse
Affiliation(s)
- Marius Meinhold
- School of Medicine and Health, Technical University of Munich, Connollystrasse 32, 80809, Munich, Germany.
| | - Sander Verbrugge
- School of Medicine and Health, Technical University of Munich, Connollystrasse 32, 80809, Munich, Germany
| | - Andi Shi
- Laboratory for Myology, Department of Human Movement Sciences, Faculty of Behavioural and Movement Sciences, Amsterdam Movement Sciences, Vrije Universiteit Amsterdam, De Boelelaan 1108, 1081 HZ, Amsterdam, The Netherlands
- Department of Prosthodontics, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou, China
| | - Martin Schönfelder
- School of Medicine and Health, Technical University of Munich, Connollystrasse 32, 80809, Munich, Germany
| | - Lore Becker
- Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, German Mouse Clinic, Ingolstädter Landstrasse 1, 85764, Neuherberg, Germany
| | - Richard T Jaspers
- Laboratory for Myology, Department of Human Movement Sciences, Faculty of Behavioural and Movement Sciences, Amsterdam Movement Sciences, Vrije Universiteit Amsterdam, De Boelelaan 1108, 1081 HZ, Amsterdam, The Netherlands
- Department of Prosthodontics, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou, China
| | - Peter S Zammit
- Randall Centre for Cell and Molecular Biophysics, King's College London, New Hunt's House, Guy's Campus, London, SE1 1UL, UK
| | - Henning Wackerhage
- School of Medicine and Health, Technical University of Munich, Connollystrasse 32, 80809, Munich, Germany
| |
Collapse
|
7
|
Ganassi M, Zammit PS, Hughes SM. Isolation, Culture, and Analysis of Zebrafish Myofibers and Associated Muscle Stem Cells to Explore Adult Skeletal Myogenesis. Methods Mol Biol 2023; 2640:21-43. [PMID: 36995585 DOI: 10.1007/978-1-0716-3036-5_3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/31/2023]
Abstract
Adult skeletal musculature experiences continuous physical stress, and hence requires maintenance and repair to ensure its continued efficient functioning. The population of resident muscle stem cells (MuSCs), termed satellite cells, resides beneath the basal lamina of adult myofibers, contributing to both muscle hypertrophy and regeneration. Upon exposure to activating stimuli, MuSCs proliferate to generate new myoblasts that differentiate and fuse to regenerate or grow myofibers. Moreover, many teleost fish undergo continuous growth throughout life, requiring continual nuclear recruitment from MuSCs to initiate and grow new fibers, a process that contrasts with the determinate growth observed in most amniotes. In this chapter, we describe a method for the isolation, culture, and immunolabeling of adult zebrafish myofibers that permits examination of both myofiber characteristics ex vivo and the MuSC myogenic program in vitro. Morphometric analysis of isolated myofibers is suitable to assess differences among slow and fast muscles or to investigate cellular features such as sarcomeres and neuromuscular junctions. Immunostaining for Pax7, a canonical stemness marker, identifies MuSCs on isolated myofibers for study. Furthermore, the plating of viable myofibers allows MuSC activation and expansion and downstream analysis of their proliferative and differentiative dynamics, thus providing a suitable, parallel alternative to amniote models for the study of vertebrate myogenesis.
Collapse
Affiliation(s)
- Massimo Ganassi
- Randall Centre for Cell and Molecular Biophysics, King's College London, London, UK.
| | - Peter S Zammit
- Randall Centre for Cell and Molecular Biophysics, King's College London, London, UK
| | - Simon M Hughes
- Randall Centre for Cell and Molecular Biophysics, King's College London, London, UK.
| |
Collapse
|
8
|
Rodriguez-Outeiriño L, Hernandez-Torres F, Ramirez de Acuña F, Rastrojo A, Creus C, Carvajal A, Salmeron L, Montolio M, Soblechero-Martin P, Arechavala-Gomeza V, Franco D, Aranega AE. miR-106b is a novel target to promote muscle regeneration and restore satellite stem cell function in injured Duchenne dystrophic muscle. MOLECULAR THERAPY - NUCLEIC ACIDS 2022; 29:769-786. [PMID: 36159592 PMCID: PMC9463180 DOI: 10.1016/j.omtn.2022.08.025] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Accepted: 08/14/2022] [Indexed: 10/26/2022]
|
9
|
Hernández-Camacho JD, Fernández-Ayala DJM, Vicente-García C, Navas-Enamorado I, López-Lluch G, Oliva C, Artuch R, Garcia-Villoria J, Ribes A, de Cabo R, Carvajal JJ, Navas P. Calorie Restriction Rescues Mitochondrial Dysfunction in Adck2-Deficient Skeletal Muscle. Front Physiol 2022; 13:898792. [PMID: 35936917 PMCID: PMC9351392 DOI: 10.3389/fphys.2022.898792] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Accepted: 06/22/2022] [Indexed: 11/20/2022] Open
Abstract
ADCK2 haploinsufficiency-mediated mitochondrial coenzyme Q deficiency in skeletal muscle causes mitochondrial myopathy associated with defects in beta-oxidation of fatty acids, aged-matched metabolic reprogramming, and defective physical performance. Calorie restriction has proven to increase lifespan and delay the onset of chronic diseases associated to aging. To study the possible treatment by food deprivation, heterozygous Adck2 knockout mice were fed under 40% calorie restriction (CR) and the phenotype was followed for 7 months. The overall glucose and fatty acids metabolism in muscle was restored in mutant mice to WT levels after CR. CR modulated the skeletal muscle metabolic profile of mutant mice, partially rescuing the profile of WT animals. The analysis of mitochondria isolated from skeletal muscle demonstrated that CR increased both CoQ levels and oxygen consumption rate (OCR) based on both glucose and fatty acids substrates, along with mitochondrial mass. The elevated aerobic metabolism fits with an increase of type IIa fibers, and a reduction of type IIx in mutant muscles, reaching WT levels. To further explore the effect of CR over muscle stem cells, satellite cells were isolated and induced to differentiate in culture media containing serum from animals in either ad libitum or CR diets for 72 h. Mutant cells showed slower differentiation alongside with decreased oxygen consumption. In vitro differentiation of mutant cells was increased under CR serum reaching levels of WT isolated cells, recovering respiration measured by OCR and partially beta-oxidation of fatty acids. The overall increase of skeletal muscle bioenergetics following CR intervention is paralleled with a physical activity improvement, with some increases in two and four limbs strength tests, and weights strength test. Running wheel activity was also partially improved in mutant mice under CR. These results demonstrate that CR intervention, which has been shown to improve age-associated physical and metabolic decline in WT mice, also recovers the defective aerobic metabolism and differentiation of skeletal muscle in mice caused by ADCK2 haploinsufficiency.
Collapse
Affiliation(s)
- Juan Diego Hernández-Camacho
- Centro Andaluz de Biología del Desarrollo, Universidad Pablo de Olavide-CSIC-JA, Sevilla, Spain
- CIBERER, Instituto de Salud Carlos III, Madrid, Spain
| | - Daniel J. M. Fernández-Ayala
- Centro Andaluz de Biología del Desarrollo, Universidad Pablo de Olavide-CSIC-JA, Sevilla, Spain
- CIBERER, Instituto de Salud Carlos III, Madrid, Spain
| | - Cristina Vicente-García
- Centro Andaluz de Biología del Desarrollo, Universidad Pablo de Olavide-CSIC-JA, Sevilla, Spain
| | - Ignacio Navas-Enamorado
- Centro Andaluz de Biología del Desarrollo, Universidad Pablo de Olavide-CSIC-JA, Sevilla, Spain
- Atsena Therapeutics, Durham, NC, United States
| | - Guillermo López-Lluch
- Centro Andaluz de Biología del Desarrollo, Universidad Pablo de Olavide-CSIC-JA, Sevilla, Spain
- CIBERER, Instituto de Salud Carlos III, Madrid, Spain
| | - Clara Oliva
- CIBERER, Instituto de Salud Carlos III, Madrid, Spain
- Clinical Biochemistry Department, Institut de Recerca Sant Joan de Déu, Barcelona, Spain
| | - Rafael Artuch
- CIBERER, Instituto de Salud Carlos III, Madrid, Spain
- Clinical Biochemistry Department, Institut de Recerca Sant Joan de Déu, Barcelona, Spain
| | - Judith Garcia-Villoria
- CIBERER, Instituto de Salud Carlos III, Madrid, Spain
- Inborn Errors of Metabolism Section, Biochemistry and Molecular Genetics Department, Hospital Clinic, Barcelona, Spain
| | - Antonia Ribes
- CIBERER, Instituto de Salud Carlos III, Madrid, Spain
- Inborn Errors of Metabolism Section, Biochemistry and Molecular Genetics Department, Hospital Clinic, Barcelona, Spain
| | - Rafael de Cabo
- Translational Gerontology Branch, National Institute on Aging Intramural Research Program, National Institutes of Health, Baltimore, MD, United States
| | - Jaime J. Carvajal
- Centro Andaluz de Biología del Desarrollo, Universidad Pablo de Olavide-CSIC-JA, Sevilla, Spain
| | - Plácido Navas
- Centro Andaluz de Biología del Desarrollo, Universidad Pablo de Olavide-CSIC-JA, Sevilla, Spain
- CIBERER, Instituto de Salud Carlos III, Madrid, Spain
- *Correspondence: Plácido Navas,
| |
Collapse
|
10
|
Ganassi M, Zammit PS, Hughes SM. Isolation of Myofibres and Culture of Muscle Stem Cells from Adult Zebrafish. Bio Protoc 2021; 11:e4149. [PMID: 34604454 PMCID: PMC8443456 DOI: 10.21769/bioprotoc.4149] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Revised: 05/24/2021] [Accepted: 06/01/2021] [Indexed: 11/02/2022] Open
Abstract
Skeletal muscles generate force throughout life and require maintenance and repair to ensure efficiency. The population of resident muscle stem cells (MuSCs), termed satellite cells, dwells beneath the basal lamina of adult myofibres and contributes to both muscle growth and regeneration. Upon exposure to activating signals, MuSCs proliferate to generate myoblasts that differentiate and fuse to grow or regenerate myofibres. This myogenic progression resembles aspects of muscle formation and development during embryogenesis. Therefore, the study of MuSCs and their associated myofibres permits the exploration of muscle stem cell biology, including the cellular and molecular mechanisms underlying muscle formation, maintenance and repair. As most aspects of MuSC biology have been described in rodents, their relevance to other species, including humans, is unclear and would benefit from comparison to an alternative vertebrate system. Here, we describe a procedure for the isolation and immunolabelling or culture of adult zebrafish myofibres that allows examination of both myofibre characteristics and MuSC biology ex vivo. Isolated myofibres can be analysed for morphometric characteristics such as the myofibre volume and myonuclear domain to assess the dynamics of muscle growth. Immunolabelling for canonical stemness markers or reporter transgenes identifies MuSCs on isolated myofibres for cellular/molecular studies. Furthermore, viable myofibres can be plated, allowing MuSC myogenesis and analysis of proliferative and differentiative dynamics in primary progenitor cells. In conclusion, we provide a comparative system to amniote models for the study of vertebrate myogenesis, which will reveal fundamental genetic and cellular mechanisms of MuSC biology and inform aquaculture. Graphic abstract: Schematic of Myofibre Isolation and Culture of Muscle Stem Cells from Adult Zebrafish.
Collapse
Affiliation(s)
- Massimo Ganassi
- Randall Centre for Cell and Molecular Biophysics, King’s College London, SE1 1UL, UK
| | - Peter S. Zammit
- Randall Centre for Cell and Molecular Biophysics, King’s College London, SE1 1UL, UK
| | - Simon M. Hughes
- Randall Centre for Cell and Molecular Biophysics, King’s College London, SE1 1UL, UK
| |
Collapse
|
11
|
Abstract
The resident stem cell for skeletal muscle is the satellite cell. On the 50th anniversary of its discovery in 1961, we described the history of skeletal muscle research and the seminal findings made during the first 20 years in the life of the satellite cell (Scharner and Zammit 2011, doi: 10.1186/2044-5040-1-28). These studies established the satellite cell as the source of myoblasts for growth and regeneration of skeletal muscle. Now on the 60th anniversary, we highlight breakthroughs in the second phase of satellite cell research from 1980 to 2000. These include technical innovations such as isolation of primary satellite cells and viable muscle fibres complete with satellite cells in their niche, together with generation of many useful reagents including genetically modified organisms and antibodies still in use today. New methodologies were combined with description of endogenous satellite cells markers, notably Pax7. Discovery of the muscle regulatory factors Myf5, MyoD, myogenin, and MRF4 in the late 1980s revolutionized understanding of the control of both developmental and regerenative myogenesis. Emergence of genetic lineage markers facilitated identification of satellite cells in situ, and also empowered transplantation studies to examine satellite cell function. Finally, satellite cell heterogeneity and the supportive role of non-satellite cell types in muscle regeneration were described. These major advances in methodology and in understanding satellite cell biology provided further foundations for the dramatic escalation of work on muscle stem cells in the 21st century.
Collapse
Affiliation(s)
- Elise N Engquist
- Randall Centre for Cell and Molecular Biophysics, King's College London, Guy's Campus, London, SE1 1UL, UK
| | - Peter S Zammit
- Randall Centre for Cell and Molecular Biophysics, King's College London, Guy's Campus, London, SE1 1UL, UK
| |
Collapse
|
12
|
García-Prat L, Perdiguero E, Alonso-Martín S, Dell'Orso S, Ravichandran S, Brooks SR, Juan AH, Campanario S, Jiang K, Hong X, Ortet L, Ruiz-Bonilla V, Flández M, Moiseeva V, Rebollo E, Jardí M, Sun HW, Musarò A, Sandri M, Del Sol A, Sartorelli V, Muñoz-Cánoves P. FoxO maintains a genuine muscle stem-cell quiescent state until geriatric age. Nat Cell Biol 2020; 22:1307-1318. [PMID: 33106654 DOI: 10.1038/s41556-020-00593-7] [Citation(s) in RCA: 102] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Accepted: 09/15/2020] [Indexed: 02/07/2023]
Abstract
Tissue regeneration declines with ageing but little is known about whether this arises from changes in stem-cell heterogeneity. Here, in homeostatic skeletal muscle, we identify two quiescent stem-cell states distinguished by relative CD34 expression: CD34High, with stemness properties (genuine state), and CD34Low, committed to myogenic differentiation (primed state). The genuine-quiescent state is unexpectedly preserved into later life, succumbing only in extreme old age due to the acquisition of primed-state traits. Niche-derived IGF1-dependent Akt activation debilitates the genuine stem-cell state by imposing primed-state features via FoxO inhibition. Interventions to neutralize Akt and promote FoxO activity drive a primed-to-genuine state conversion, whereas FoxO inactivation deteriorates the genuine state at a young age, causing regenerative failure of muscle, as occurs in geriatric mice. These findings reveal transcriptional determinants of stem-cell heterogeneity that resist ageing more than previously anticipated and are only lost in extreme old age, with implications for the repair of geriatric muscle.
Collapse
Affiliation(s)
- Laura García-Prat
- Department of Experimental and Health Sciences, Pompeu Fabra University (UPF), CIBER on Neurodegenerative Diseases (CIBERNED), Barcelona, Spain.,Spanish National Center on Cardiovascular Research (CNIC), Madrid, Spain.,Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Eusebio Perdiguero
- Department of Experimental and Health Sciences, Pompeu Fabra University (UPF), CIBER on Neurodegenerative Diseases (CIBERNED), Barcelona, Spain
| | - Sonia Alonso-Martín
- Spanish National Center on Cardiovascular Research (CNIC), Madrid, Spain.,Neurosciences Area, Biodonostia Health Research Institute, Donostia-San Sebastián, San Sebastián, Spain
| | - Stefania Dell'Orso
- Laboratory of Muscle Stem Cells and Gene Regulation, National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS), NIH Bethesda, Bethesda, MD, USA
| | - Srikanth Ravichandran
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Stephen R Brooks
- Biodata Mining and Discovery Section, NIAMS, NIH Bethesda, Bethesda, MD, USA
| | - Aster H Juan
- Laboratory of Muscle Stem Cells and Gene Regulation, National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS), NIH Bethesda, Bethesda, MD, USA
| | - Silvia Campanario
- Department of Experimental and Health Sciences, Pompeu Fabra University (UPF), CIBER on Neurodegenerative Diseases (CIBERNED), Barcelona, Spain.,Spanish National Center on Cardiovascular Research (CNIC), Madrid, Spain
| | - Kan Jiang
- Biodata Mining and Discovery Section, NIAMS, NIH Bethesda, Bethesda, MD, USA
| | - Xiaotong Hong
- Spanish National Center on Cardiovascular Research (CNIC), Madrid, Spain
| | - Laura Ortet
- Department of Experimental and Health Sciences, Pompeu Fabra University (UPF), CIBER on Neurodegenerative Diseases (CIBERNED), Barcelona, Spain
| | - Vanessa Ruiz-Bonilla
- Department of Experimental and Health Sciences, Pompeu Fabra University (UPF), CIBER on Neurodegenerative Diseases (CIBERNED), Barcelona, Spain
| | - Marta Flández
- Spanish National Center on Cardiovascular Research (CNIC), Madrid, Spain.,Grupo de Investigación en Oncología Clínico Traslacional, Instituto de Investigación Hospital 12 de Octubre, Madrid, Spain
| | - Victoria Moiseeva
- Department of Experimental and Health Sciences, Pompeu Fabra University (UPF), CIBER on Neurodegenerative Diseases (CIBERNED), Barcelona, Spain
| | - Elena Rebollo
- Molecular Imaging Platform, Molecular Biology Institute of Barcelona (IBMB-CSIC), Barcelona, Spain
| | - Mercè Jardí
- Department of Experimental and Health Sciences, Pompeu Fabra University (UPF), CIBER on Neurodegenerative Diseases (CIBERNED), Barcelona, Spain
| | - Hong-Wei Sun
- Biodata Mining and Discovery Section, NIAMS, NIH Bethesda, Bethesda, MD, USA
| | - Antonio Musarò
- DAHFMO-Unit of Histology and Medical Embryology, Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Sapienza University of Rome, Rome, Italy
| | - Marco Sandri
- Department of Biomedical Science, University of Padova, Padova, Italy
| | - Antonio Del Sol
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg.,CIC bioGUNE, Derio, Spain.,IKERBASQUE, Basque Foundation for Science, Bilbao, Spain
| | - Vittorio Sartorelli
- Laboratory of Muscle Stem Cells and Gene Regulation, National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS), NIH Bethesda, Bethesda, MD, USA.
| | - Pura Muñoz-Cánoves
- Department of Experimental and Health Sciences, Pompeu Fabra University (UPF), CIBER on Neurodegenerative Diseases (CIBERNED), Barcelona, Spain. .,Spanish National Center on Cardiovascular Research (CNIC), Madrid, Spain. .,ICREA, Barcelona, Spain.
| |
Collapse
|
13
|
Petkova MV, Stantzou A, Morin A, Petrova O, Morales‐Gonzalez S, Seifert F, Bellec‐Dyevre J, Manoliu T, Goyenvalle A, Garcia L, Richard I, Laplace‐Builhé C, Schuelke M, Amthor H. Live‐imaging of revertant and therapeutically restored dystrophin in the
Dmd
EGFP‐mdx
mouse model for Duchenne muscular dystrophy. Neuropathol Appl Neurobiol 2020; 46:602-614. [DOI: 10.1111/nan.12639] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Revised: 06/03/2020] [Accepted: 06/08/2020] [Indexed: 01/01/2023]
Affiliation(s)
- M. V. Petkova
- Université Paris‐SaclayUVSQInsermEND‐ICAP Versailles France
- Department of Neuropediatrics Charité–Universitätsmedizin Berlincorporate member of Freie Universität BerlinHumboldt‐Universität zu BerlinBerlin Institute of Health (BIH)NeuroCure Clinical Research Center Berlin Germany
| | - A. Stantzou
- Université Paris‐SaclayUVSQInsermEND‐ICAP Versailles France
| | - A. Morin
- Université Paris‐SaclayUVSQInsermEND‐ICAP Versailles France
| | - O. Petrova
- Université Paris‐SaclayUVSQInsermEND‐ICAP Versailles France
| | - S. Morales‐Gonzalez
- Department of Neuropediatrics Charité–Universitätsmedizin Berlincorporate member of Freie Universität BerlinHumboldt‐Universität zu BerlinBerlin Institute of Health (BIH)NeuroCure Clinical Research Center Berlin Germany
| | - F. Seifert
- Department of Neuropediatrics Charité–Universitätsmedizin Berlincorporate member of Freie Universität BerlinHumboldt‐Universität zu BerlinBerlin Institute of Health (BIH)NeuroCure Clinical Research Center Berlin Germany
| | - J. Bellec‐Dyevre
- Integrare (UMR_S951)InsermGénéthonUniv EvryUniversité Paris‐Saclay Evry France
| | - T. Manoliu
- Gustave RoussyUniversité Paris‐SaclayPlate‐forme Imagerie et Cytométrie.UMS AMMCa. Villejuif France
| | - A. Goyenvalle
- Université Paris‐SaclayUVSQInsermEND‐ICAP Versailles France
- LIA BAHN Centre scientifique de Monaco Monaco
| | - L. Garcia
- Université Paris‐SaclayUVSQInsermEND‐ICAP Versailles France
- LIA BAHN Centre scientifique de Monaco Monaco
| | - I. Richard
- Integrare (UMR_S951)InsermGénéthonUniv EvryUniversité Paris‐Saclay Evry France
| | - C. Laplace‐Builhé
- Gustave RoussyUniversité Paris‐SaclayPlate‐forme Imagerie et Cytométrie.UMS AMMCa. Villejuif France
| | - M. Schuelke
- Department of Neuropediatrics Charité–Universitätsmedizin Berlincorporate member of Freie Universität BerlinHumboldt‐Universität zu BerlinBerlin Institute of Health (BIH)NeuroCure Clinical Research Center Berlin Germany
| | - H. Amthor
- Université Paris‐SaclayUVSQInsermEND‐ICAP Versailles France
- Pediatric Department University Hospital Raymond Poincaré Garches France
| |
Collapse
|
14
|
Moyle LA, Cheng RY, Liu H, Davoudi S, Ferreira SA, Nissar AA, Sun Y, Gentleman E, Simmons CA, Gilbert PM. Three-dimensional niche stiffness synergizes with Wnt7a to modulate the extent of satellite cell symmetric self-renewal divisions. Mol Biol Cell 2020; 31:1703-1713. [PMID: 32491970 PMCID: PMC7521850 DOI: 10.1091/mbc.e20-01-0078] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Satellite cells (SCs), the resident adult stem cells of skeletal muscle, are required for tissue repair throughout life. While many signaling pathways are known to control SC self-renewal, less is known about the mechanisms underlying the spatiotemporal control of self-renewal during skeletal muscle repair. Here, we measured biomechanical changes that accompany skeletal muscle regeneration and determined the implications on SC fate. Using atomic force microscopy, we quantified a 2.9-fold stiffening of the SC niche at time-points associated with planar-oriented symmetric self-renewal divisions. Immunohistochemical analysis confirms increased extracellular matrix deposition within the basal lamina. To test whether three-dimensional (3D) niche stiffness can alter SC behavior or fate, we embedded isolated SC-associated muscle fibers within biochemically inert agarose gels tuned to mimic native tissue stiffness. Time-lapse microscopy revealed that a stiff 3D niche significantly increased the proportion of planar-oriented divisions, without effecting SC viability, fibronectin deposition, or fate change. We then found that 3D niche stiffness synergizes with WNT7a, a biomolecule shown to control SC symmetric self-renewal divisions via the noncanonical WNT/planar cell polarity pathway, to modify stem cell pool expansion. Our results provide new insights into the role of 3D niche biomechanics in regulating SC fate choice.
Collapse
Affiliation(s)
- Louise A Moyle
- Institute of Biomedical Engineering, Toronto, ON M5S 3G9, Canada.,Donnelly Centre for Cellular and Biomolecular Research, Toronto, ON M5S 3E1, Canada
| | - Richard Y Cheng
- Institute of Biomedical Engineering, Toronto, ON M5S 3G9, Canada.,Donnelly Centre for Cellular and Biomolecular Research, Toronto, ON M5S 3E1, Canada
| | - Haijiao Liu
- Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, Toronto, ON M5G 1M1, Canada.,Department of Mechanical & Industrial Engineering, University of Toronto, Toronto, ON M5S 3G8, Canada
| | - Sadegh Davoudi
- Institute of Biomedical Engineering, Toronto, ON M5S 3G9, Canada.,Donnelly Centre for Cellular and Biomolecular Research, Toronto, ON M5S 3E1, Canada
| | - Silvia A Ferreira
- Centre for Craniofacial and Regenerative Biology, King's College London, London, SE1 9RT, United Kingdom
| | - Aliyah A Nissar
- Institute of Biomedical Engineering, Toronto, ON M5S 3G9, Canada.,Donnelly Centre for Cellular and Biomolecular Research, Toronto, ON M5S 3E1, Canada
| | - Yu Sun
- Department of Mechanical & Industrial Engineering, University of Toronto, Toronto, ON M5S 3G8, Canada
| | - Eileen Gentleman
- Centre for Craniofacial and Regenerative Biology, King's College London, London, SE1 9RT, United Kingdom
| | - Craig A Simmons
- Institute of Biomedical Engineering, Toronto, ON M5S 3G9, Canada.,Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, Toronto, ON M5G 1M1, Canada.,Department of Mechanical & Industrial Engineering, University of Toronto, Toronto, ON M5S 3G8, Canada
| | - Penney M Gilbert
- Institute of Biomedical Engineering, Toronto, ON M5S 3G9, Canada.,Donnelly Centre for Cellular and Biomolecular Research, Toronto, ON M5S 3E1, Canada.,Department of Cell and Systems Biology, University of Toronto, Toronto ON, M5S 1A8, Canada
| |
Collapse
|
15
|
Figeac N, Pruller J, Hofer I, Fortier M, Ortuste Quiroga HP, Banerji CRS, Zammit PS. DEPDC1B is a key regulator of myoblast proliferation in mouse and man. Cell Prolif 2020; 53:e12717. [PMID: 31825138 PMCID: PMC6985657 DOI: 10.1111/cpr.12717] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Revised: 08/19/2019] [Accepted: 09/09/2019] [Indexed: 02/07/2023] Open
Abstract
OBJECTIVES DISHEVELLED, EGL-10, PLECKSTRIN (DEP) domain-containing 1B (DEPDC1B) promotes dismantling of focal adhesions and coordinates detachment events during cell cycle progression. DEPDC1B is overexpressed in several cancers with expression inversely correlated with patient survival. Here, we analysed the role of DEPDC1B in the regulation of murine and human skeletal myogenesis. MATERIALS AND METHODS Expression dynamics of DEPDC1B were examined in murine and human myoblasts and rhabdomyosarcoma cells in vitro by RT-qPCR and/or immunolabelling. DEPDC1B function was mainly tested via siRNA-mediated gene knockdown. RESULTS DEPDC1B was expressed in proliferating murine and human myoblasts, with expression then decreasing markedly during myogenic differentiation. SiRNA-mediated knockdown of DEPDC1B reduced myoblast proliferation and induced entry into myogenic differentiation, with deregulation of key cell cycle regulators (cyclins, CDK, CDKi). DEPDC1B and β-catenin co-knockdown was unable to rescue proliferation in myoblasts, suggesting that DEPDC1B functions independently of canonical WNT signalling during myogenesis. DEPDC1B can also suppress RHOA activity in some cell types, but DEPDC1B and RHOA co-knockdown actually had an additive effect by both further reducing proliferation and enhancing myogenic differentiation. DEPDC1B was expressed in human Rh30 rhabdomyosarcoma cells, where DEPDC1B or RHOA knockdown promoted myogenic differentiation, but without influencing proliferation. CONCLUSION DEPDC1B plays a central role in myoblasts by driving proliferation and preventing precocious myogenic differentiation during skeletal myogenesis in both mouse and human.
Collapse
Affiliation(s)
- Nicolas Figeac
- King's College LondonRandall Centre for Cell and Molecular BiophysicsLondonUK
| | - Johanna Pruller
- King's College LondonRandall Centre for Cell and Molecular BiophysicsLondonUK
| | - Isabella Hofer
- King's College LondonRandall Centre for Cell and Molecular BiophysicsLondonUK
| | - Mathieu Fortier
- King's College LondonRandall Centre for Cell and Molecular BiophysicsLondonUK
| | | | | | - Peter S. Zammit
- King's College LondonRandall Centre for Cell and Molecular BiophysicsLondonUK
| |
Collapse
|
16
|
Figeac N, Mohamed AD, Sun C, Schönfelder M, Matallanas D, Garcia-Munoz A, Missiaglia E, Collie-Duguid E, De Mello V, Pobbati AV, Pruller J, Jaka O, Harridge SDR, Hong W, Shipley J, Vargesson N, Zammit PS, Wackerhage H. VGLL3 operates via TEAD1, TEAD3 and TEAD4 to influence myogenesis in skeletal muscle. J Cell Sci 2019; 132:jcs.225946. [PMID: 31138678 PMCID: PMC6633393 DOI: 10.1242/jcs.225946] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Accepted: 05/03/2019] [Indexed: 12/21/2022] Open
Abstract
VGLL proteins are transcriptional co-factors that bind TEAD family transcription factors to regulate events ranging from wing development in fly, to muscle fibre composition and immune function in mice. Here, we characterise Vgll3 in skeletal muscle. We found that mouse Vgll3 was expressed at low levels in healthy muscle but that its levels increased during hypertrophy or regeneration; in humans, VGLL3 was highly expressed in tissues from patients with various muscle diseases, such as in dystrophic muscle and alveolar rhabdomyosarcoma. Interaction proteomics revealed that VGLL3 bound TEAD1, TEAD3 and TEAD4 in myoblasts and/or myotubes. However, there was no interaction with proteins from major regulatory systems such as the Hippo kinase cascade, unlike what is found for the TEAD co-factors YAP (encoded by YAP1) and TAZ (encoded by WWTR1). Vgll3 overexpression reduced the activity of the Hippo negative-feedback loop, affecting expression of muscle-regulating genes including Myf5, Pitx2 and Pitx3, and genes encoding certain Wnts and IGFBPs. VGLL3 mainly repressed gene expression, regulating similar genes to those regulated by YAP and TAZ. siRNA-mediated Vgll3 knockdown suppressed myoblast proliferation, whereas Vgll3 overexpression strongly promoted myogenic differentiation. However, skeletal muscle was overtly normal in Vgll3-null mice, presumably due to feedback signalling and/or redundancy. This work identifies VGLL3 as a transcriptional co-factor operating with the Hippo signal transduction network to control myogenesis. Summary: VGLL3 interacts with TEAD transcription factors to direct expression of crucial muscle regulatory genes and contribute to the control of skeletal myogenesis.
Collapse
Affiliation(s)
- Nicolas Figeac
- Randall Centre for Cell and Molecular Biophysics, King's College London, London SE1 1UL, UK
| | - Abdalla D Mohamed
- School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Foresterhill, Aberdeen AB25 2ZD, UK.,Institute of Developmental Genetics, Helmholtz Zentrum München, German Research Center for Environment and Health, Ingolstaedter Landstrasse 1, D-85764 Munich/Neuherberg, Germany
| | - Congshan Sun
- Randall Centre for Cell and Molecular Biophysics, King's College London, London SE1 1UL, UK.,Department of Neurology, The Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Martin Schönfelder
- Faculty of Sport and Health Sciences, Technical University of Munich, Georg-Brauchle-Ring 60, 80992 Munich, Germany
| | - David Matallanas
- Systems Biology Ireland, Conway Institute, Belfield; Dublin 4, Ireland
| | | | - Edoardo Missiaglia
- Institute of Pathology, Lausanne University Hospital (CHUV), 1011 Lausanne, Switzerland
| | - Elaina Collie-Duguid
- University of Aberdeen, Centre for Genome Enabled Biology and Medicine, 23 St Machar Drive, Aberdeen AB24 3RY, UK
| | - Vanessa De Mello
- School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Foresterhill, Aberdeen AB25 2ZD, UK
| | - Ajaybabu V Pobbati
- Institute of Molecular and Cell Biology, A-STAR, 61 Biopolis Drive, Singapore 138673, Singapore
| | - Johanna Pruller
- Randall Centre for Cell and Molecular Biophysics, King's College London, London SE1 1UL, UK
| | - Oihane Jaka
- Centre for Human and Applied Physiological Sciences, King's College London, London SE1 1UL, UK
| | - Stephen D R Harridge
- Centre for Human and Applied Physiological Sciences, King's College London, London SE1 1UL, UK
| | - Wanjin Hong
- Institute of Molecular and Cell Biology, A-STAR, 61 Biopolis Drive, Singapore 138673, Singapore
| | - Janet Shipley
- Sarcoma Molecular Pathology Team, Divisions of Molecular Pathology and Cancer Therapeutics, Institute of Cancer Research, Surrey, SM2 5NG, UK
| | - Neil Vargesson
- School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Foresterhill, Aberdeen AB25 2ZD, UK
| | - Peter S Zammit
- Randall Centre for Cell and Molecular Biophysics, King's College London, London SE1 1UL, UK
| | - Henning Wackerhage
- School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Foresterhill, Aberdeen AB25 2ZD, UK .,Faculty of Sport and Health Sciences, Technical University of Munich, Georg-Brauchle-Ring 60, 80992 Munich, Germany
| |
Collapse
|
17
|
PAX3 Confers Functional Heterogeneity in Skeletal Muscle Stem Cell Responses to Environmental Stress. Cell Stem Cell 2019; 24:958-973.e9. [PMID: 31006622 DOI: 10.1016/j.stem.2019.03.019] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Revised: 02/04/2019] [Accepted: 03/22/2019] [Indexed: 11/22/2022]
Abstract
Muscle satellite cells (MuSCs) are the quiescent muscle stem cells required for adult skeletal muscle repair. The impact of environmental stress such as pollution on MuSC behavior remains unexplored. We evaluated the impact of 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) exposure, a ubiquitous and highly toxic pollutant, on MuSCs by combining in vivo mouse molecular genetic models with ex vivo studies. While all MuSCs express the transcription factor PAX7, we show that a subset also express PAX3 and exhibit resistance to environmental stress. Upon systemic TCDD treatment, PAX3-negative MuSCs display impaired survival, atypical activation, and sporadic differentiation through xenobiotic aryl hydrocarbon receptor signaling. We further show that PAX3-positive MuSCs become sensitized to environmental stress when PAX3 function is impaired and that PAX3-mediated induction of mTORC1 is required for protection. Our study, therefore, identifies a functional heterogeneity of MuSCs in response to environmental stress controlled by PAX3.
Collapse
|
18
|
Myogenin promotes myocyte fusion to balance fibre number and size. Nat Commun 2018; 9:4232. [PMID: 30315160 PMCID: PMC6185967 DOI: 10.1038/s41467-018-06583-6] [Citation(s) in RCA: 88] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Accepted: 08/31/2018] [Indexed: 01/01/2023] Open
Abstract
Each skeletal muscle acquires its unique size before birth, when terminally differentiating myocytes fuse to form a defined number of multinucleated myofibres. Although mice in which the transcription factor Myogenin is mutated lack most myogenesis and die perinatally, a specific cell biological role for Myogenin has remained elusive. Here we report that loss of function of zebrafish myog prevents formation of almost all multinucleated muscle fibres. A second, Myogenin-independent, fusion pathway in the deep myotome requires Hedgehog signalling. Lack of Myogenin does not prevent terminal differentiation; the smaller myotome has a normal number of myocytes forming more mononuclear, thin, albeit functional, fast muscle fibres. Mechanistically, Myogenin binds to the myomaker promoter and is required for expression of myomaker and other genes essential for myocyte fusion. Adult myog mutants display reduced muscle mass, decreased fibre size and nucleation. Adult-derived myog mutant myocytes show persistent defective fusion ex vivo. Myogenin is therefore essential for muscle homeostasis, regulating myocyte fusion to determine both muscle fibre number and size. Loss of the transcription factor Myogenin in mice reduces skeletal myogenesis and leads to perinatal death but how Myogenin regulates muscle formation is unclear. Here, the authors show that zebrafish Myogenin enhances Myomaker expression, muscle cell fusion and myotome size, yet decreases fast muscle fibre number.
Collapse
|
19
|
Mademtzoglou D, Asakura Y, Borok MJ, Alonso-Martin S, Mourikis P, Kodaka Y, Mohan A, Asakura A, Relaix F. Cellular localization of the cell cycle inhibitor Cdkn1c controls growth arrest of adult skeletal muscle stem cells. eLife 2018; 7:33337. [PMID: 30284969 PMCID: PMC6172026 DOI: 10.7554/elife.33337] [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] [Received: 11/04/2017] [Accepted: 09/02/2018] [Indexed: 01/01/2023] Open
Abstract
Adult skeletal muscle maintenance and regeneration depend on efficient muscle stem cell (MuSC) functions. The mechanisms coordinating cell cycle with activation, renewal, and differentiation of MuSCs remain poorly understood. Here, we investigated how adult MuSCs are regulated by CDKN1c (p57kip2), a cyclin-dependent kinase inhibitor, using mouse molecular genetics. In the absence of CDKN1c, skeletal muscle repair is severely impaired after injury. We show that CDKN1c is not expressed in quiescent MuSCs, while being induced in activated and proliferating myoblasts and maintained in differentiating myogenic cells. In agreement, isolated Cdkn1c-deficient primary myoblasts display differentiation defects and increased proliferation. We further show that the subcellular localization of CDKN1c is dynamic; while CDKN1c is initially localized to the cytoplasm of activated/proliferating myoblasts, progressive nuclear translocation leads to growth arrest during differentiation. We propose that CDKN1c activity is restricted to differentiating myoblasts by regulated cyto-nuclear relocalization, coordinating the balance between proliferation and growth arrest.
Collapse
Affiliation(s)
- Despoina Mademtzoglou
- Inserm, IMRB U955-E10, F-94010, Créteil, France.,Ecole Nationale Veterinaire d'Alfort, Faculté de medecine, F-94000, Université Paris-Est Creteil, Maison Alfort, France
| | - Yoko Asakura
- Stem Cell Institute, Paul and Sheila Wellstone Muscular Dystrophy Center, Department of Neurology, University of Minnesota Medical School, Minneapolis, United States
| | - Matthew J Borok
- Inserm, IMRB U955-E10, F-94010, Créteil, France.,Ecole Nationale Veterinaire d'Alfort, Faculté de medecine, F-94000, Université Paris-Est Creteil, Maison Alfort, France
| | - Sonia Alonso-Martin
- Inserm, IMRB U955-E10, F-94010, Créteil, France.,Ecole Nationale Veterinaire d'Alfort, Faculté de medecine, F-94000, Université Paris-Est Creteil, Maison Alfort, France
| | - Philippos Mourikis
- Inserm, IMRB U955-E10, F-94010, Créteil, France.,Ecole Nationale Veterinaire d'Alfort, Faculté de medecine, F-94000, Université Paris-Est Creteil, Maison Alfort, France
| | - Yusaku Kodaka
- Stem Cell Institute, Paul and Sheila Wellstone Muscular Dystrophy Center, Department of Neurology, University of Minnesota Medical School, Minneapolis, United States
| | - Amrudha Mohan
- Stem Cell Institute, Paul and Sheila Wellstone Muscular Dystrophy Center, Department of Neurology, University of Minnesota Medical School, Minneapolis, United States
| | - Atsushi Asakura
- Stem Cell Institute, Paul and Sheila Wellstone Muscular Dystrophy Center, Department of Neurology, University of Minnesota Medical School, Minneapolis, United States
| | - Frederic Relaix
- Inserm, IMRB U955-E10, F-94010, Créteil, France.,Ecole Nationale Veterinaire d'Alfort, Faculté de medecine, F-94000, Université Paris-Est Creteil, Maison Alfort, France.,Etablissement Français du Sang, Créteil, France.,APHP, Hopitaux Universitaires Henri Mondor, DHU Pepsy & Centre de Référence des Maladies Neuromusculaires GNMH, Créteil, France
| |
Collapse
|
20
|
Prüller J, Mannhardt I, Eschenhagen T, Zammit PS, Figeac N. Satellite cells delivered in their niche efficiently generate functional myotubes in three-dimensional cell culture. PLoS One 2018; 13:e0202574. [PMID: 30222770 PMCID: PMC6141091 DOI: 10.1371/journal.pone.0202574] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Accepted: 08/05/2018] [Indexed: 12/22/2022] Open
Abstract
Biophysical/biochemical cues from the environment contribute to regulation of the regenerative capacity of resident skeletal muscle stem cells called satellites cells. This can be observed in vitro, where muscle cell behaviour is influenced by the particular culture substrates and whether culture is performed in a 2D or 3D environment, with changes including morphology, nuclear shape and cytoskeletal organization. To create a 3D skeletal muscle model we compared collagen I, Fibrin or PEG-Fibrinogen with different sources of murine and human myogenic cells. To generate tension in the 3D scaffold, biomaterials were polymerised between two flexible silicone posts to mimic tendons. This 3D culture system has multiple advantages including being simple, fast to set up and inexpensive, so providing an accessible tool to investigate myogenesis in a 3D environment. Immortalised human and murine myoblast lines, and primary murine satellite cells showed varying degrees of myogenic differentiation when cultured in these biomaterials, with C2 myoblasts in particular forming large multinucleated myotubes in collagen I or Fibrin. However, murine satellite cells retained in their niche on a muscle fibre and embedded in 3D collagen I or Fibrin gels generated aligned, multinucleated and contractile myotubes.
Collapse
Affiliation(s)
- Johanna Prüller
- King's College London, Randall Centre for Cell and Molecular Biophysics, New Hunt's House, Guy's Campus, London, England
| | - Ingra Mannhardt
- Department of Experimental Pharmacology and Toxicology, University Medical Center Hamburg Eppendorf, Hamburg, Germany.,DZHK (German Centre for Cardiovascular Research), Hamburg, Germany
| | - Thomas Eschenhagen
- Department of Experimental Pharmacology and Toxicology, University Medical Center Hamburg Eppendorf, Hamburg, Germany.,DZHK (German Centre for Cardiovascular Research), Hamburg, Germany
| | - Peter S Zammit
- King's College London, Randall Centre for Cell and Molecular Biophysics, New Hunt's House, Guy's Campus, London, England
| | - Nicolas Figeac
- King's College London, Randall Centre for Cell and Molecular Biophysics, New Hunt's House, Guy's Campus, London, England
| |
Collapse
|
21
|
Alonso-Martin S, Auradé F, Mademtzoglou D, Rochat A, Zammit PS, Relaix F. SOXF factors regulate murine satellite cell self-renewal and function through inhibition of β-catenin activity. eLife 2018; 7:26039. [PMID: 29882512 PMCID: PMC6021169 DOI: 10.7554/elife.26039] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Accepted: 06/07/2018] [Indexed: 12/17/2022] Open
Abstract
Muscle satellite cells are the primary source of stem cells for postnatal skeletal muscle growth and regeneration. Understanding genetic control of satellite cell formation, maintenance, and acquisition of their stem cell properties is on-going, and we have identified SOXF (SOX7, SOX17, SOX18) transcriptional factors as being induced during satellite cell specification. We demonstrate that SOXF factors regulate satellite cell quiescence, self-renewal and differentiation. Moreover, ablation of Sox17 in the muscle lineage impairs postnatal muscle growth and regeneration. We further determine that activities of SOX7, SOX17 and SOX18 overlap during muscle regeneration, with SOXF transcriptional activity requisite. Finally, we show that SOXF factors also control satellite cell expansion and renewal by directly inhibiting the output of β-catenin activity, including inhibition of Ccnd1 and Axin2. Together, our findings identify a key regulatory function of SoxF genes in muscle stem cells via direct transcriptional control and interaction with canonical Wnt/β-catenin signaling.
Collapse
Affiliation(s)
- Sonia Alonso-Martin
- Institut Mondor de Recherche Biomédicale, INSERM U955-E10, Créteil, France.,Université Paris Est, Faculté de Medecine, Créteil, France.,Ecole Nationale Veterinaire d'Alfort, Maison Alfort, France
| | - Frédéric Auradé
- Sorbonne Université, INSERM U974, Center for Research in Myology, Paris, France
| | - Despoina Mademtzoglou
- Institut Mondor de Recherche Biomédicale, INSERM U955-E10, Créteil, France.,Université Paris Est, Faculté de Medecine, Créteil, France.,Ecole Nationale Veterinaire d'Alfort, Maison Alfort, France
| | - Anne Rochat
- Sorbonne Université, INSERM U974, Center for Research in Myology, Paris, France
| | - Peter S Zammit
- Randall Centre for Cell and Molecular Biophysics, King's College London, London, United Kingdom
| | - Frédéric Relaix
- Institut Mondor de Recherche Biomédicale, INSERM U955-E10, Créteil, France.,Université Paris Est, Faculté de Medecine, Créteil, France.,Ecole Nationale Veterinaire d'Alfort, Maison Alfort, France.,Etablissement Français du Sang, Creteil, France.,APHP, Hopitaux UniversitairesHenri Mondor, Centre de Référence des Maladies Neuromusculaires GNMH, Créteil, France
| |
Collapse
|
22
|
Morphoregulatory functions of the RNA-binding motif protein 3 in cell spreading, polarity and migration. Sci Rep 2018; 8:7367. [PMID: 29743635 PMCID: PMC5943363 DOI: 10.1038/s41598-018-25668-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Accepted: 03/27/2018] [Indexed: 02/01/2023] Open
Abstract
RNA-binding proteins are emerging as key regulators of transitions in cell morphology. The RNA-binding motif protein 3 (RBM3) is a cold-inducible RNA-binding protein with broadly relevant roles in cellular protection, and putative functions in cancer and development. Several findings suggest that RBM3 has morphoregulatory functions germane to its roles in these contexts. For example, RBM3 helps maintain the morphological integrity of cell protrusions during cell stress and disease. Moreover, it is highly expressed in migrating neurons of the developing brain and in cancer invadopodia, suggesting roles in migration. We here show that RBM3 regulates cell polarity, spreading and migration. RBM3 was present in spreading initiation centers, filopodia and blebs that formed during cell spreading in cell lines and primary myoblasts. Reducing RBM3 triggered exaggerated spreading, increased RhoA expression, and a loss of polarity that was rescued by Rho kinase inhibition and overexpression of CRMP2. High RBM3 expression enhanced the motility of cells migrating by a mesenchymal mode involving extension of long protrusions, whereas RBM3 knockdown slowed migration, greatly reducing the ability of cells to extend protrusions and impairing multiple processes that require directional migration. These data establish novel functions of RBM3 of potential significance to tissue repair, metastasis and development.
Collapse
|
23
|
Rayagiri SS, Ranaldi D, Raven A, Mohamad Azhar NIF, Lefebvre O, Zammit PS, Borycki AG. Basal lamina remodeling at the skeletal muscle stem cell niche mediates stem cell self-renewal. Nat Commun 2018. [PMID: 29540680 PMCID: PMC5852002 DOI: 10.1038/s41467-018-03425-3] [Citation(s) in RCA: 139] [Impact Index Per Article: 19.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
A central question in stem cell biology is the relationship between stem cells and their niche. Although previous reports have uncovered how signaling molecules released by niche cells support stem cell function, the role of the extra-cellular matrix (ECM) within the niche is unclear. Here, we show that upon activation, skeletal muscle stem cells (satellite cells) induce local remodeling of the ECM and the deposition of laminin-α1 and laminin-α5 into the basal lamina of the satellite cell niche. Genetic ablation of laminin-α1, disruption of integrin-α6 signaling or blocking matrix metalloproteinase activity impairs satellite cell expansion and self-renewal. Collectively, our findings establish that remodeling of the ECM is an integral process of stem cell activity to support propagation and self-renewal, and may explain the effect laminin-α1-containing supports have on embryonic and adult stem cells, as well as the regenerative activity of exogenous laminin-111 therapy. Extracellular matrix (ECM) remodelling is thought to have effects on muscle stem cells that support muscle homeostasis. Here the authors show ECM remodeling controls satellite cell self-renewal through deposition of laminin-α1 into the satellite cell niche.
Collapse
Affiliation(s)
- Shantisree Sandeepani Rayagiri
- Department of Biomedical Science, University of Sheffield, Firth Court, Western Bank, Sheffield, S10 2TN, UK.,Biotherapeutics Development Unit, Cancer Research UK, Clare Hall laboratories, Blanche Lane, South Mimms, Hertfordshire, EN6 3LD, UK
| | - Daniele Ranaldi
- Department of Biomedical Science, University of Sheffield, Firth Court, Western Bank, Sheffield, S10 2TN, UK
| | - Alexander Raven
- Department of Biomedical Science, University of Sheffield, Firth Court, Western Bank, Sheffield, S10 2TN, UK.,MRC Centre for Regenerative Medicine, SCRM Building, University of Edinburgh, 5 Little France Drive, Edinburgh, EH16 4UU, UK
| | - Nur Izzah Farhana Mohamad Azhar
- Department of Biomedical Science, University of Sheffield, Firth Court, Western Bank, Sheffield, S10 2TN, UK.,Oxford Publishing (Malaysia), Shah Alam, 40150, Selangor Darul Ehsan, Malaysia
| | - Olivier Lefebvre
- Inserm U1109 MN3T, F-67200, Strasbourg, France.,Université de Strasbourg, F-67000, Strasbourg, France.,LabEx Medalis Université de Strasbourg, F-67000, Strasbourg, France.,Fédération de Médecine Translationnelle de Strasbourg (FMTS), F-67000, Strasbourg, France
| | - Peter S Zammit
- Randall Centre for Cell and Molecular Biophysics, Faculty of Life Sciences & Medicine King's College London, New Hunt's House, Guy's Campus, London, SE1 1UL, UK
| | - Anne-Gaëlle Borycki
- Department of Biomedical Science, University of Sheffield, Firth Court, Western Bank, Sheffield, S10 2TN, UK.
| |
Collapse
|
24
|
Goel AJ, Krauss RS. Ex Vivo Visualization and Analysis of the Muscle Stem Cell Niche. Methods Mol Biol 2018; 2002:39-50. [PMID: 30178310 DOI: 10.1007/7651_2018_177] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Adult skeletal muscle stem cells, termed satellite cells, are essential for regenerating muscle after tissue damage. Satellite cells are located in a specialized microenvironment between muscle fibers and their surrounding basal lamina. This local niche serves as a compartment to preserve satellite cell function and provides signals that facilitate the rapid response to injury. Visualization of this local niche enables the elucidation of such niche-derived signals. Here, we describe techniques for isolating single myofibers with their associated satellite cells for ex vivo visualization and analysis of an intact muscle stem cell niche.
Collapse
Affiliation(s)
- Aviva J Goel
- Department of Cell, Developmental, and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Robert S Krauss
- Department of Cell, Developmental, and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
25
|
Distinguishing States of Arrest: Genome-Wide Descriptions of Cellular Quiescence Using ChIP-Seq and RNA-Seq Analysis. Methods Mol Biol 2018; 1686:215-239. [PMID: 29030824 DOI: 10.1007/978-1-4939-7371-2_16] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Regenerative potential in adult stem cells is closely associated with the establishment of-and exit from-a temporary state of quiescence. Emerging evidence not only provides a rationale for the link between lineage determination programs and cell cycle regulation but also highlights the understanding of quiescence as an actively maintained cellular program, encompassing networks and mechanisms beyond mitotic inactivity or metabolic restriction. Interrogating the quiescent genome and transcriptome using deep-sequencing technologies offers an unprecedented view of the global mechanisms governing this reversibly arrested cellular state and its importance for cell identity. While many efforts have identified and isolated pure target stem cell populations from a variety of adult tissues, there is a growing appreciation that their isolation from the stem cell niche in vivo leads to activation and loss of hallmarks of quiescence. Thus, in vitro models that recapitulate the dynamic reversibly arrested stem cell state in culture and lend themselves to comparison with the activated or differentiated state are useful templates for genome-wide analysis of the quiescence network.In this chapter, we describe the methods that can be adopted for whole genome epigenomic and transcriptomic analysis of cells derived from one such established culture model where mouse myoblasts are triggered to enter or exit quiescence as homogeneous populations. The ability to synchronize myoblasts in G0 permits insights into the genome in "deep quiescence." The culture methods for generating large populations of quiescent myoblasts in either 2D or 3D culture formats are described in detail in a previous chapter in this series (Arora et al. Methods Mol Biol 1556:283-302, 2017). Among the attractive features of this model are that genes isolated from quiescent myoblasts in culture mark satellite cells in vivo (Sachidanandan et al., J Cell Sci 115:2701-2712, 2002) providing a validation of its approximation of the molecular state of true stem cells. Here, we provide our working protocols for ChIP-seq and RNA-seq analysis, focusing on those experimental elements that require standardization for optimal analysis of chromatin and RNA from quiescent myoblasts, and permitting useful and revealing comparisons with proliferating myoblasts or differentiated myotubes.
Collapse
|
26
|
Advanced Methods to Study the Cross Talk Between Fibro-Adipogenic Progenitors and Muscle Stem Cells. Methods Mol Biol 2018; 1687:231-256. [PMID: 29067668 DOI: 10.1007/978-1-4939-7374-3_17] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Functional interactions between muscle (satellite) stem cells-MuSCs-and other cellular components of their niche (the fibro-adipogenic progenitors-FAPs) coordinate regeneration of injured as well as diseased skeletal muscles. These interactions are largely mediated by secretory networks, whose integrity is critical to determine whether repair occurs by compensatory regeneration leading to formation of new contractile fibers, or by maladaptive formation of fibrotic scars and fat infiltration. Here we provide the description of methods for isolation of FAPs and MuSCs from muscles of wild type and dystrophic mice, and protocols of cocultures as well as MuSC's exposure to FAP- derived exosomes. These methods and protocols can be exploited in murine models of acute muscle injury to investigate salient features of physiological repair, and in models of muscular diseases to identify dysregulated networks that compromise functional interactions between cellular components of the regeneration environment during disease progression. We predict that exporting these procedures to patient-derived muscle samples will contribute to advance our understanding of human skeletal myogenesis and related disorders.
Collapse
|
27
|
Banerji CRS, Panamarova M, Hebaishi H, White RB, Relaix F, Severini S, Zammit PS. PAX7 target genes are globally repressed in facioscapulohumeral muscular dystrophy skeletal muscle. Nat Commun 2017; 8:2152. [PMID: 29255294 PMCID: PMC5735185 DOI: 10.1038/s41467-017-01200-4] [Citation(s) in RCA: 73] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2016] [Accepted: 08/25/2017] [Indexed: 11/09/2022] Open
Abstract
Facioscapulohumeral muscular dystrophy (FSHD) is a prevalent, incurable myopathy, linked to hypomethylation of D4Z4 repeats on chromosome 4q causing expression of the DUX4 transcription factor. However, DUX4 is difficult to detect in FSHD muscle biopsies and it is debatable how robust changes in DUX4 target gene expression are as an FSHD biomarker. PAX7 is a master regulator of myogenesis that rescues DUX4-mediated apoptosis. Here, we show that suppression of PAX7 target genes is a hallmark of FSHD, and that it is as major a signature of FSHD muscle as DUX4 target gene expression. This is shown using meta-analysis of over six FSHD muscle biopsy gene expression studies, and validated by RNA-sequencing on FSHD patient-derived myoblasts. DUX4 also inhibits PAX7 from activating its transcriptional target genes and vice versa. Furthermore, PAX7 target gene repression can explain oxidative stress sensitivity and epigenetic changes in FSHD. Thus, PAX7 target gene repression is a hallmark of FSHD that should be considered in the investigation of FSHD pathology and therapy. Facioscapulohumeral muscular dystrophy is a myopathy linked to ectopic expression of the DUX4 transcription factor. The authors show that the suppression of targets genes of the myogenesis regulator PAX7 is a signature of FSHD, and might explain oxidative stress sensitivity and epigenetic changes.
Collapse
Affiliation(s)
- Christopher R S Banerji
- Randall Centre of Cell and Molecular Biophysics, New Hunt's House, King's College London, Guy's Campus, London, SE1 1UL, UK. .,Department of Computer Science, University College London, London, WC1E 6BT, UK. .,Centre of Mathematics and Physics in the Life Sciences and Experimental Biology, University College London, London, WC1E 6BT, UK. .,Statistical Cancer Genomics, Paul O'Gorman Building, UCL Cancer Institute, University College London, London, WC1E 6BT, UK.
| | - Maryna Panamarova
- Randall Centre of Cell and Molecular Biophysics, New Hunt's House, King's College London, Guy's Campus, London, SE1 1UL, UK
| | - Husam Hebaishi
- Randall Centre of Cell and Molecular Biophysics, New Hunt's House, King's College London, Guy's Campus, London, SE1 1UL, UK
| | - Robert B White
- Randall Centre of Cell and Molecular Biophysics, New Hunt's House, King's College London, Guy's Campus, London, SE1 1UL, UK.,School of Anatomy, Physiology & Human Biology, The University of Western Australia, Crawley, WA, 6009, Australia
| | - Frédéric Relaix
- Paris Est-Creteil University, IMRB U955, Faculté de médecine 8 rue du Général Sarrail, 94000, Créteil, France
| | - Simone Severini
- Department of Computer Science, University College London, London, WC1E 6BT, UK.,Institute of Natural Sciences, Shanghai Jiao Tong University, 800 Dongchuan Road, Minhang District, 200240, Shanghai, China
| | - Peter S Zammit
- Randall Centre of Cell and Molecular Biophysics, New Hunt's House, King's College London, Guy's Campus, London, SE1 1UL, UK.
| |
Collapse
|
28
|
Stantzou A, Schirwis E, Swist S, Alonso-Martin S, Polydorou I, Zarrouki F, Mouisel E, Beley C, Julien A, Le Grand F, Garcia L, Colnot C, Birchmeier C, Braun T, Schuelke M, Relaix F, Amthor H. BMP signaling regulates satellite cell-dependent postnatal muscle growth. Development 2017; 144:2737-2747. [PMID: 28694257 DOI: 10.1242/dev.144089] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2016] [Accepted: 06/21/2017] [Indexed: 02/03/2023]
Abstract
Postnatal growth of skeletal muscle largely depends on the expansion and differentiation of resident stem cells, the so-called satellite cells. Here, we demonstrate that postnatal satellite cells express components of the bone morphogenetic protein (BMP) signaling machinery. Overexpression of noggin in postnatal mice (to antagonize BMP ligands), satellite cell-specific knockout of Alk3 (the gene encoding the BMP transmembrane receptor) or overexpression of inhibitory SMAD6 decreased satellite cell proliferation and accretion during myofiber growth, and ultimately retarded muscle growth. Moreover, reduced BMP signaling diminished the adult satellite cell pool. Abrogation of BMP signaling in satellite cell-derived primary myoblasts strongly diminished cell proliferation and upregulated the expression of cell cycle inhibitors p21 and p57 In conclusion, these results show that BMP signaling defines postnatal muscle development by regulating satellite cell-dependent myofiber growth and the generation of the adult muscle stem cell pool.
Collapse
Affiliation(s)
- Amalia Stantzou
- Versailles Saint-Quentin-en-Yvelines University, INSERM U1179, LIA BAHN CSM, Montigny-le-Bretonneux 78180, France.,Department of Neuropediatrics and NeuroCure Clinical Research Center, Charité University-Medicine, Berlin 10117, Germany.,Pierre et Marie Curie University, Paris Sorbonne, INSERM, UMRS974, CNRS FRE3617, Center for Research in Myology, Paris 75013, France
| | - Elija Schirwis
- Versailles Saint-Quentin-en-Yvelines University, INSERM U1179, LIA BAHN CSM, Montigny-le-Bretonneux 78180, France.,Pierre et Marie Curie University, Paris Sorbonne, INSERM, UMRS974, CNRS FRE3617, Center for Research in Myology, Paris 75013, France.,Developmental Biology/Signal Transduction Group, Max Delbruck Center for Molecular Medicine, Berlin 13092, Germany
| | - Sandra Swist
- Department of Cardiac Development and Remodeling, Max-Planck-Institute for Heart and Lung Research, Bad Nauheim 61231, Germany.,Department of Cardiovascular Physiology, Ruhr University Bochum, D-44780 Bochum, Germany
| | - Sonia Alonso-Martin
- Pierre et Marie Curie University, Paris Sorbonne, INSERM, UMRS974, CNRS FRE3617, Center for Research in Myology, Paris 75013, France.,INSERM, Paris Est University, IMRB U955-E10, Créteil 94010, France
| | - Ioanna Polydorou
- Versailles Saint-Quentin-en-Yvelines University, INSERM U1179, LIA BAHN CSM, Montigny-le-Bretonneux 78180, France.,Department of Neuropediatrics and NeuroCure Clinical Research Center, Charité University-Medicine, Berlin 10117, Germany
| | - Faouzi Zarrouki
- Versailles Saint-Quentin-en-Yvelines University, INSERM U1179, LIA BAHN CSM, Montigny-le-Bretonneux 78180, France
| | - Etienne Mouisel
- Pierre et Marie Curie University, Paris Sorbonne, INSERM, UMRS974, CNRS FRE3617, Center for Research in Myology, Paris 75013, France.,Paul Sabatier University, Inserm UMR 1048, Toulouse 31432, France
| | | | - Anaïs Julien
- Paris Descartes-Sorbonne Paris Cité University, Inserm UMR1163, Imagine Institute, Paris 75015, France
| | - Fabien Le Grand
- Pierre et Marie Curie University, Paris Sorbonne, INSERM, UMRS974, CNRS FRE3617, Center for Research in Myology, Paris 75013, France
| | - Luis Garcia
- Versailles Saint-Quentin-en-Yvelines University, INSERM U1179, LIA BAHN CSM, Montigny-le-Bretonneux 78180, France
| | - Céline Colnot
- Paris Descartes-Sorbonne Paris Cité University, Inserm UMR1163, Imagine Institute, Paris 75015, France
| | - Carmen Birchmeier
- Developmental Biology/Signal Transduction Group, Max Delbruck Center for Molecular Medicine, Berlin 13092, Germany
| | - Thomas Braun
- Department of Cardiac Development and Remodeling, Max-Planck-Institute for Heart and Lung Research, Bad Nauheim 61231, Germany
| | - Markus Schuelke
- Department of Neuropediatrics and NeuroCure Clinical Research Center, Charité University-Medicine, Berlin 10117, Germany
| | - Frédéric Relaix
- Pierre et Marie Curie University, Paris Sorbonne, INSERM, UMRS974, CNRS FRE3617, Center for Research in Myology, Paris 75013, France.,INSERM, Paris Est University, IMRB U955-E10, Créteil 94010, France
| | - Helge Amthor
- Versailles Saint-Quentin-en-Yvelines University, INSERM U1179, LIA BAHN CSM, Montigny-le-Bretonneux 78180, France .,Pediatric Department, University Hospital Raymond Poincaré, Garches 92380, France
| |
Collapse
|
29
|
Sun C, De Mello V, Mohamed A, Ortuste Quiroga HP, Garcia-Munoz A, Al Bloshi A, Tremblay AM, von Kriegsheim A, Collie-Duguid E, Vargesson N, Matallanas D, Wackerhage H, Zammit PS. Common and Distinctive Functions of the Hippo Effectors Taz and Yap in Skeletal Muscle Stem Cell Function. Stem Cells 2017; 35:1958-1972. [PMID: 28589555 PMCID: PMC5575518 DOI: 10.1002/stem.2652] [Citation(s) in RCA: 90] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Accepted: 01/07/2017] [Indexed: 12/13/2022]
Abstract
Hippo pathway downstream effectors Yap and Taz play key roles in cell proliferation and regeneration, regulating gene expression especially via Tead transcription factors. To investigate their role in skeletal muscle stem cells, we analyzed Taz in vivo and ex vivo in comparison with Yap. Small interfering RNA knockdown or retroviral‐mediated expression of wild‐type human or constitutively active TAZ mutants in satellite cells showed that TAZ promoted proliferation, a function shared with YAP. However, at later stages of myogenesis, TAZ also enhanced myogenic differentiation of myoblasts, whereas YAP inhibits such differentiation. Functionally, while muscle growth was mildly affected in Taz (gene Wwtr1–/–) knockout mice, there were no overt effects on regeneration. Conversely, conditional knockout of Yap in satellite cells of Pax7Cre‐ERT2/+: Yapfl°x/fl°x:Rosa26Lacz mice produced a regeneration deficit. To identify potential mechanisms, microarray analysis showed many common TAZ/YAP target genes, but TAZ also regulates some genes independently of YAP, including myogenic genes such as Pax7, Myf5, and Myod1 (ArrayExpress–E‐MTAB‐5395). Proteomic analysis revealed many novel binding partners of TAZ/YAP in myogenic cells, but TAZ also interacts with proteins distinct from YAP that are often involved in myogenesis and aspects of cytoskeleton organization (ProteomeXchange–PXD005751). Neither TAZ nor YAP bind members of the Wnt destruction complex but both regulated expression of Wnt and Wnt‐cross talking genes with known roles in myogenesis. Finally, TAZ operates through Tead4 to enhance myogenic differentiation. In summary, Taz and Yap have overlapping functions in promoting myoblast proliferation but Taz then switches to enhance myogenic differentiation. Stem Cells2017;35:1958–1972
Collapse
Affiliation(s)
- Congshan Sun
- Randall Division of Cell and Molecular Biophysics, King's College London, London, UK
| | - Vanessa De Mello
- School of Medicine, Medical Sciences & Nutrition, University of Aberdeen, Foresterhill, Aberdeen, Scotland, UK
| | - Abdalla Mohamed
- School of Medicine, Medical Sciences & Nutrition, University of Aberdeen, Foresterhill, Aberdeen, Scotland, UK
| | | | | | - Abdullah Al Bloshi
- School of Medicine, Medical Sciences & Nutrition, University of Aberdeen, Foresterhill, Aberdeen, Scotland, UK
| | - Annie M Tremblay
- Stem Cell Program, Children's Hospital, Boston, Massachusetts, USA.,Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, Massachusetts, USA.,Harvard Stem Cell Institute, Cambridge, Massachusetts, USA
| | | | - Elaina Collie-Duguid
- Centre for Genome Enabled Biology and Medicine, School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Foresterhill, Aberdeen, Scotland, UK
| | - Neil Vargesson
- School of Medicine, Medical Sciences & Nutrition, University of Aberdeen, Foresterhill, Aberdeen, Scotland, UK
| | | | - Henning Wackerhage
- School of Medicine, Medical Sciences & Nutrition, University of Aberdeen, Foresterhill, Aberdeen, Scotland, UK.,Faculty of Sport and Health Sciences, Technical University of Munich, Munich, Germany
| | - Peter S Zammit
- Randall Division of Cell and Molecular Biophysics, King's College London, London, UK
| |
Collapse
|
30
|
Prostaglandin E2 is essential for efficacious skeletal muscle stem-cell function, augmenting regeneration and strength. Proc Natl Acad Sci U S A 2017; 114:6675-6684. [PMID: 28607093 PMCID: PMC5495271 DOI: 10.1073/pnas.1705420114] [Citation(s) in RCA: 155] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Skeletal muscles harbor quiescent muscle-specific stem cells (MuSCs) capable of tissue regeneration throughout life. Muscle injury precipitates a complex inflammatory response in which a multiplicity of cell types, cytokines, and growth factors participate. Here we show that Prostaglandin E2 (PGE2) is an inflammatory cytokine that directly targets MuSCs via the EP4 receptor, leading to MuSC expansion. An acute treatment with PGE2 suffices to robustly augment muscle regeneration by either endogenous or transplanted MuSCs. Loss of PGE2 signaling by specific genetic ablation of the EP4 receptor in MuSCs impairs regeneration, leading to decreased muscle force. Inhibition of PGE2 production through nonsteroidal anti-inflammatory drug (NSAID) administration just after injury similarly hinders regeneration and compromises muscle strength. Mechanistically, the PGE2 EP4 interaction causes MuSC expansion by triggering a cAMP/phosphoCREB pathway that activates the proliferation-inducing transcription factor, Nurr1 Our findings reveal that loss of PGE2 signaling to MuSCs during recovery from injury impedes muscle repair and strength. Through such gain- or loss-of-function experiments, we found that PGE2 signaling acts as a rheostat for muscle stem-cell function. Decreased PGE2 signaling due to NSAIDs or increased PGE2 due to exogenous delivery dictates MuSC function, which determines the outcome of regeneration. The markedly enhanced and accelerated repair of damaged muscles following intramuscular delivery of PGE2 suggests a previously unrecognized indication for this therapeutic agent.
Collapse
|
31
|
Monge C, DiStasio N, Rossi T, Sébastien M, Sakai H, Kalman B, Boudou T, Tajbakhsh S, Marty I, Bigot A, Mouly V, Picart C. Quiescence of human muscle stem cells is favored by culture on natural biopolymeric films. Stem Cell Res Ther 2017; 8:104. [PMID: 28464938 PMCID: PMC5414338 DOI: 10.1186/s13287-017-0556-8] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Revised: 03/01/2017] [Accepted: 04/06/2017] [Indexed: 12/18/2022] Open
Abstract
Background Satellite cells are quiescent resident muscle stem cells that present an important potential to regenerate damaged tissue. However, this potential is diminished once they are removed from their niche environment in vivo, prohibiting the long-term study and genetic investigation of these cells. This study therefore aimed to provide a novel biomaterial platform for the in-vitro culture of human satellite cells that maintains their stem-like quiescent state, an important step for cell therapeutic studies. Methods Human muscle satellite cells were isolated from two donors and cultured on soft biopolymeric films of controlled stiffness. Cell adhesive phenotype, maintenance of satellite cell quiescence and capacity for gene manipulation were investigated using FACS, western blotting, fluorescence microscopy and electron microscopy. Results About 85% of satellite cells cultured in vitro on soft biopolymer films for 3 days maintained expression of the quiescence marker Pax7, as compared with 60% on stiffer films and 50% on tissue culture plastic. The soft biopolymeric films allowed satellite cell culture for up to 6 days without renewing the media. These cells retained their stem-like properties, as evidenced by the expression of stem cell markers and reduced expression of differentiated markers. In addition, 95% of cells grown on these soft biopolymeric films were in the G0/G1 stage of the cell cycle, as opposed to those grown on plastic that became activated and began to proliferate and differentiate. Conclusions Our study identifies a new biomaterial made of a biopolymer thin film for the maintenance of the quiescence state of muscle satellite cells. These cells could be activated at any point simply by replating them onto a plastic culture dish. Furthermore, these cells could be genetically manipulated by viral transduction, showing that this biomaterial may be further used for therapeutic strategies. Electronic supplementary material The online version of this article (doi:10.1186/s13287-017-0556-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Claire Monge
- CNRS, UMR 5628, LMGP, 3 parvis Louis Néel, F-38016, Grenoble, France. .,Université de Grenoble Alpes, Grenoble Institute of Technology, 3 parvis Louis Néel, F-38016, Grenoble, France.
| | - Nicholas DiStasio
- CNRS, UMR 5628, LMGP, 3 parvis Louis Néel, F-38016, Grenoble, France.,Université de Grenoble Alpes, Grenoble Institute of Technology, 3 parvis Louis Néel, F-38016, Grenoble, France
| | - Thomas Rossi
- CNRS, UMR 5628, LMGP, 3 parvis Louis Néel, F-38016, Grenoble, France.,Université de Grenoble Alpes, Grenoble Institute of Technology, 3 parvis Louis Néel, F-38016, Grenoble, France
| | - Muriel Sébastien
- Université de Grenoble Alpes, Grenoble Institut des Neurosciences, GIN, Chemin Fortuné Ferrini, F-3800, Grenoble, France.,INSERM, U1216, F-38000, Grenoble, France
| | - Hiroshi Sakai
- Stem Cells & Development, Department of Developmental & Stem Cell Biology, Institut Pasteur, 25 rue du Dr Roux, Paris, 75015, France.,CNRS, UMR 3738; Institut Pasteur, 25 rue du Dr Roux, Paris, 75015, France
| | - Benoit Kalman
- CNRS, UMR 5628, LMGP, 3 parvis Louis Néel, F-38016, Grenoble, France.,Université de Grenoble Alpes, Grenoble Institute of Technology, 3 parvis Louis Néel, F-38016, Grenoble, France
| | - Thomas Boudou
- CNRS, UMR 5628, LMGP, 3 parvis Louis Néel, F-38016, Grenoble, France.,Université de Grenoble Alpes, Grenoble Institute of Technology, 3 parvis Louis Néel, F-38016, Grenoble, France
| | - Shahragim Tajbakhsh
- Stem Cells & Development, Department of Developmental & Stem Cell Biology, Institut Pasteur, 25 rue du Dr Roux, Paris, 75015, France.,CNRS, UMR 3738; Institut Pasteur, 25 rue du Dr Roux, Paris, 75015, France
| | - Isabelle Marty
- Université de Grenoble Alpes, Grenoble Institut des Neurosciences, GIN, Chemin Fortuné Ferrini, F-3800, Grenoble, France.,INSERM, U1216, F-38000, Grenoble, France
| | - Anne Bigot
- Sorbonne Universités, UPMC Université Paris 06, INSERM UMRS974, CNRS FRE3617, Center for Research in Myology, 47 Boulevard de l'hôpital, 75013, Paris, France
| | - Vincent Mouly
- Sorbonne Universités, UPMC Université Paris 06, INSERM UMRS974, CNRS FRE3617, Center for Research in Myology, 47 Boulevard de l'hôpital, 75013, Paris, France
| | - Catherine Picart
- CNRS, UMR 5628, LMGP, 3 parvis Louis Néel, F-38016, Grenoble, France. .,Université de Grenoble Alpes, Grenoble Institute of Technology, 3 parvis Louis Néel, F-38016, Grenoble, France.
| |
Collapse
|
32
|
Moyle LA, Blanc E, Jaka O, Prueller J, Banerji CR, Tedesco FS, Harridge SD, Knight RD, Zammit PS. Ret function in muscle stem cells points to tyrosine kinase inhibitor therapy for facioscapulohumeral muscular dystrophy. eLife 2016; 5. [PMID: 27841748 PMCID: PMC5108591 DOI: 10.7554/elife.11405] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2015] [Accepted: 09/01/2016] [Indexed: 12/16/2022] Open
Abstract
Facioscapulohumeral muscular dystrophy (FSHD) involves sporadic expression of DUX4, which inhibits myogenesis and is pro-apoptotic. To identify target genes, we over-expressed DUX4 in myoblasts and found that the receptor tyrosine kinase Ret was significantly up-regulated, suggesting a role in FSHD. RET is dynamically expressed during myogenic progression in mouse and human myoblasts. Constitutive expression of either RET9 or RET51 increased myoblast proliferation, whereas siRNA-mediated knockdown of Ret induced myogenic differentiation. Suppressing RET activity using Sunitinib, a clinically-approved tyrosine kinase inhibitor, rescued differentiation in both DUX4-expressing murine myoblasts and in FSHD patient-derived myoblasts. Importantly, Sunitinib also increased engraftment and differentiation of FSHD myoblasts in regenerating mouse muscle. Thus, DUX4-mediated activation of Ret prevents myogenic differentiation and could contribute to FSHD pathology by preventing satellite cell-mediated repair. Rescue of DUX4-induced pathology by Sunitinib highlights the therapeutic potential of tyrosine kinase inhibitors for treatment of FSHD. DOI:http://dx.doi.org/10.7554/eLife.11405.001
Collapse
Affiliation(s)
- Louise A Moyle
- Randall Division of Cell and Molecular Biophysics, King's College London, London, United Kingdom.,Department of Cell and Developmental Biology, University College London, London, United Kingdom
| | - Eric Blanc
- Randall Division of Cell and Molecular Biophysics, King's College London, London, United Kingdom.,Core Unit Bioinformatics, Berlin Institute of Health, Berlin, Germany.,Institute of Pathology, Charite Universitatsmedizin Berlin, Berlin, Germany
| | - Oihane Jaka
- Centre of Human and Aerospace Physiological Sciences, King's College London, London, United Kingdom
| | - Johanna Prueller
- Randall Division of Cell and Molecular Biophysics, King's College London, London, United Kingdom
| | - Christopher Rs Banerji
- Randall Division of Cell and Molecular Biophysics, King's College London, London, United Kingdom
| | | | - Stephen Dr Harridge
- Centre of Human and Aerospace Physiological Sciences, King's College London, London, United Kingdom
| | - Robert D Knight
- Craniofacial Development and Stem Cell Biology, King's College London, London, United Kingdom
| | - Peter S Zammit
- Randall Division of Cell and Molecular Biophysics, King's College London, London, United Kingdom
| |
Collapse
|
33
|
Knopp P, Krom YD, Banerji CRS, Panamarova M, Moyle LA, den Hamer B, van der Maarel SM, Zammit PS. DUX4 induces a transcriptome more characteristic of a less-differentiated cell state and inhibits myogenesis. J Cell Sci 2016; 129:3816-3831. [PMID: 27744317 PMCID: PMC5087662 DOI: 10.1242/jcs.180372] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2016] [Accepted: 08/13/2016] [Indexed: 12/14/2022] Open
Abstract
Skeletal muscle wasting in facioscapulohumeral muscular dystrophy (FSHD) results in substantial morbidity. On a disease-permissive chromosome 4qA haplotype, genomic and/or epigenetic changes at the D4Z4 macrosatellite repeat allows transcription of the DUX4 retrogene. Analysing transgenic mice carrying a human D4Z4 genomic locus from an FSHD-affected individual showed that DUX4 was transiently induced in myoblasts during skeletal muscle regeneration. Centromeric to the D4Z4 repeats is an inverted D4Z4 unit encoding DUX4c. Expression of DUX4, DUX4c and DUX4 constructs, including constitutively active, dominant-negative and truncated versions, revealed that DUX4 activates target genes to inhibit proliferation and differentiation of satellite cells, but that it also downregulates target genes to suppress myogenic differentiation. These transcriptional changes elicited by DUX4 in mouse have significant overlap with genes regulated by DUX4 in man. Comparison of DUX4 and DUX4c transcriptional perturbations revealed that DUX4 regulates genes involved in cell proliferation, whereas DUX4c regulates genes engaged in angiogenesis and muscle development, with both DUX4 and DUX4c modifing genes involved in urogenital development. Transcriptomic analysis showed that DUX4 operates through both target gene activation and repression to orchestrate a transcriptome characteristic of a less-differentiated cell state.
Collapse
Affiliation(s)
- Paul Knopp
- Randall Division of Cell and Molecular Biophysics, Faculty of Life Sciences and Medicine, New Hunt's House, King's College London, Guy's Campus, London SE1 1UL, UK
| | - Yvonne D Krom
- Department of Human Genetics, Leiden University Medical Center, Leiden, Postbus 9600, 2300 RC, The Netherlands
| | - Christopher R S Banerji
- Randall Division of Cell and Molecular Biophysics, Faculty of Life Sciences and Medicine, New Hunt's House, King's College London, Guy's Campus, London SE1 1UL, UK Centre of Mathematics and Physics in the Life Sciences and Experimental Biology, University College London, London WC1E 6BT, UK
| | - Maryna Panamarova
- Randall Division of Cell and Molecular Biophysics, Faculty of Life Sciences and Medicine, New Hunt's House, King's College London, Guy's Campus, London SE1 1UL, UK
| | - Louise A Moyle
- Randall Division of Cell and Molecular Biophysics, Faculty of Life Sciences and Medicine, New Hunt's House, King's College London, Guy's Campus, London SE1 1UL, UK
| | - Bianca den Hamer
- Department of Human Genetics, Leiden University Medical Center, Leiden, Postbus 9600, 2300 RC, The Netherlands
| | - Silvère M van der Maarel
- Department of Human Genetics, Leiden University Medical Center, Leiden, Postbus 9600, 2300 RC, The Netherlands
| | - Peter S Zammit
- Randall Division of Cell and Molecular Biophysics, Faculty of Life Sciences and Medicine, New Hunt's House, King's College London, Guy's Campus, London SE1 1UL, UK
| |
Collapse
|
34
|
Alonso-Martin S, Rochat A, Mademtzoglou D, Morais J, de Reyniès A, Auradé F, Chang THT, Zammit PS, Relaix F. Gene Expression Profiling of Muscle Stem Cells Identifies Novel Regulators of Postnatal Myogenesis. Front Cell Dev Biol 2016; 4:58. [PMID: 27446912 PMCID: PMC4914952 DOI: 10.3389/fcell.2016.00058] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2016] [Accepted: 06/02/2016] [Indexed: 01/02/2023] Open
Abstract
Skeletal muscle growth and regeneration require a population of muscle stem cells, the satellite cells, located in close contact to the myofiber. These cells are specified during fetal and early postnatal development in mice from a Pax3/7 population of embryonic progenitor cells. As little is known about the genetic control of their formation and maintenance, we performed a genome-wide chronological expression profile identifying the dynamic transcriptomic changes involved in establishment of muscle stem cells through life, and acquisition of muscle stem cell properties. We have identified multiple genes and pathways associated with satellite cell formation, including set of genes specifically induced (EphA1, EphA2, EfnA1, EphB1, Zbtb4, Zbtb20) or inhibited (EphA3, EphA4, EphA7, EfnA2, EfnA3, EfnA4, EfnA5, EphB2, EphB3, EphB4, EfnBs, Zfp354c, Zcchc5, Hmga2) in adult stem cells. Ephrin receptors and ephrins ligands have been implicated in cell migration and guidance in many tissues including skeletal muscle. Here we show that Ephrin receptors and ephrins ligands are also involved in regulating the adult myogenic program. Strikingly, impairment of EPHB1 function in satellite cells leads to increased differentiation at the expense of self-renewal in isolated myofiber cultures. In addition, we identified new transcription factors, including several zinc finger proteins. ZFP354C and ZCCHC5 decreased self-renewal capacity when overexpressed, whereas ZBTB4 increased it, and ZBTB20 induced myogenic progression. The architectural and transcriptional regulator HMGA2 was involved in satellite cell activation. Together, our study shows that transcriptome profiling coupled with myofiber culture analysis, provides an efficient system to identify and validate candidate genes implicated in establishment/maintenance of muscle stem cells. Furthermore, tour de force transcriptomic profiling provides a wealth of data to inform for future stem cell-based muscle therapies.
Collapse
Affiliation(s)
- Sonia Alonso-Martin
- Institut Mondor de Recherche Biomédicale, INSERM U955-E10Créteil, France; Université Paris Est, Faculté de MedecineCréteil, France; Ecole Nationale Veterinaire d'AlfortMaison Alfort, France
| | - Anne Rochat
- Institut Mondor de Recherche Biomédicale, INSERM U955-E10 Créteil, France
| | - Despoina Mademtzoglou
- Institut Mondor de Recherche Biomédicale, INSERM U955-E10Créteil, France; Université Paris Est, Faculté de MedecineCréteil, France; Ecole Nationale Veterinaire d'AlfortMaison Alfort, France
| | - Jessica Morais
- Institut Mondor de Recherche Biomédicale, INSERM U955-E10 Créteil, France
| | - Aurélien de Reyniès
- Programme Cartes d'Identité des Tumeurs, Ligue Nationale Contre le Cancer Paris, France
| | - Frédéric Auradé
- Sorbonne Universités, UPMC Univ Paris 06, INSERM UMRS974, Center for Research in Myology Paris, France
| | - Ted Hung-Tse Chang
- Institut Mondor de Recherche Biomédicale, INSERM U955-E10 Créteil, France
| | - Peter S Zammit
- Randall Division of Cell and Molecular Biophysics, King's College London London, UK
| | - Frédéric Relaix
- Institut Mondor de Recherche Biomédicale, INSERM U955-E10Créteil, France; Université Paris Est, Faculté de MedecineCréteil, France; Ecole Nationale Veterinaire d'AlfortMaison Alfort, France; Etablissement Français du SangCréteil, France; APHP, Hopitaux Universitaires Henri Mondor, DHU Pepsy and Centre de Référence des Maladies Neuromusculaires GNMHCréteil, France
| |
Collapse
|
35
|
Kitajima Y, Ono Y. Estrogens maintain skeletal muscle and satellite cell functions. J Endocrinol 2016; 229:267-75. [PMID: 27048232 DOI: 10.1530/joe-15-0476] [Citation(s) in RCA: 109] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Accepted: 04/05/2016] [Indexed: 12/25/2022]
Abstract
Estrogens have crucial roles in an extensive range of physiological functions regulating cellular proliferation and differentiation, development, homeostasis, and metabolism. Therefore, prolonged estrogen insufficiency influences various types of tissues expressing estrogen receptors (ERs). Although ERs are expressed in skeletal muscle and its stem cells, called satellite cells, how prolonged estrogen insufficiency affects their function remains unclear. In this study, we investigated the effect of estrogen reduction on muscle in young ovariectomized (OVX) female mice. We found that reduced estrogens resulted in muscle atrophy in a time-dependent manner. Muscle force generation was reduced in OVX mice. Interestingly, prolonged estrogen insufficiency shifted fiber types toward faster myosin heavy chain isoforms. The number of satellite cells per isolated myofiber was unchanged, while satellite cell expansion, differentiation, and self-renewal were all markedly impaired in OVX mice. Indeed, muscle regeneration was significantly compromised in OVX mice. Taken together, our results demonstrate that estrogens are essential for comprehensively maintaining muscle function with its insufficiency affecting muscle strength and regeneration in young female mice.
Collapse
Affiliation(s)
- Yuriko Kitajima
- Department of Stem Cell BiologyAtomic Bomb Disease Institute, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Yusuke Ono
- Department of Stem Cell BiologyAtomic Bomb Disease Institute, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| |
Collapse
|
36
|
Lack of phosphatidylethanolamine N -methyltransferase in mice does not promote fatty acid oxidation in skeletal muscle. Biochim Biophys Acta Mol Cell Biol Lipids 2016; 1861:119-129. [DOI: 10.1016/j.bbalip.2015.11.008] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2015] [Revised: 10/08/2015] [Accepted: 11/16/2015] [Indexed: 01/01/2023]
|
37
|
Abstract
Skeletal muscle stem cells are satellite cells that play crucial roles in tissue repair and regeneration after muscle injury. Accumulating evidence indicates that satellite cells are genetically and functionally heterogeneous, even within the same muscle. A small population of satellite cells possesses "stemness" and exhibits the remarkable ability to regenerate through robust self-renewal when transplanted into a regenerating muscle niche. In contrast, not all satellite cells self-renew. For example, some cells are committed myogenic progenitors that immediately undergo myogenic differentiation with minimal cell division after activation. Recent studies illuminate the cellular and molecular characteristics of the functional heterogeneity among satellite cells. To evaluate heterogeneity and stem cell dynamics, here we describe methods to conduct a clonal analysis of satellite cells and to visualize a slowly dividing cell population.
Collapse
Affiliation(s)
- Yasuo Kitajima
- Department of Stem Cell Biology, Atomic Bomb Disease Institute, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, 852-8523, Japan
| | - Shizuka Ogawa
- Department of Stem Cell Biology, Atomic Bomb Disease Institute, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, 852-8523, Japan
| | - Yusuke Ono
- Department of Stem Cell Biology, Atomic Bomb Disease Institute, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, 852-8523, Japan.
| |
Collapse
|
38
|
Ortuste Quiroga HP, Goto K, Zammit PS. Isolation, Cryosection and Immunostaining of Skeletal Muscle. Methods Mol Biol 2016; 1460:85-100. [PMID: 27492168 DOI: 10.1007/978-1-4939-3810-0_8] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Adult skeletal muscle is maintained and repaired by resident stem cells called satellite cells, located between the plasmalemma of a muscle fiber, and the surrounding basal lamina. When needed, satellite cells are activated to form proliferative myoblasts, that then differentiate and fuse to existing muscle fibers, or fuse together to form replacement myofibers. In parallel, a proportion of satellite cells self-renew, to maintain the stem cell pool. To date, Pax7 is the marker of choice for identifying quiescent satellite cells. Co-immunostaining of skeletal muscle with Pax7 and laminin allows both identification of satellite cells, and the myofiber that they are associated with. Furthermore, satellite cells can be followed through the early stages of the myogenic program by co-immunostaining with myogenic regulatory factors such as MyoD. To test genetically modified mice for satellite cell expression, co-immunostaining can be performed for Pax7 and reporter genes such as eGFP. Here, we describe a method for identification of satellite cells in skeletal muscle sections, including muscle isolation, cryosectioning and co-immunostaining for Pax7 and laminin.
Collapse
Affiliation(s)
- Huascar P Ortuste Quiroga
- Randall Division of Cell and Molecular Biophysics, King's College London, New Hunt's House, Guy's Campus, London, SE1 1UL, UK
| | - Katsumasa Goto
- Department of Physiology, Graduate School of Health Sciences, Toyohashi SOZO University, 20-1 Matsushita, Ushikawa, Toyohashi, Aichi, 440-8511, Japan
| | - Peter S Zammit
- Randall Division of Cell and Molecular Biophysics, King's College London, New Hunt's House, Guy's Campus, London, SE1 1UL, UK.
| |
Collapse
|
39
|
Abstract
Polycomb group (PcG) proteins are key epigenetic factors responsible for the proper spatiotemporal repression of defined transcriptional programs along the process of cell differentiation, including myogenesis. The discovery of the pivotal role played by PcG factors during myogenic differentiation relied on the possibility to culture myogenic cells in vitro. We describe here the methods currently used to isolate muscle stem cells (MuSCs) both from single myofibers and from bulk muscles by fluorescence-activated cell sorting (FACS), highlighting experimental details and critical steps. Through these techniques MuSCs can be efficiently isolated and cultured in vitro to recapitulate the different phases of myogenesis: activation, expansion, differentiation, and self-renewal.
Collapse
Affiliation(s)
- Chiara Mozzetta
- CNR Institute of Cellular Biology and Neurobiology, IRCCS Santa Lucia Foundation, Via Del Fosso di Fiorano 64, Rome, 00143, Italy.
- Department of Biology and Biotechnology, University of Sapienza, P. le Aldo Moro 5, Rome, 00185, Italy.
| |
Collapse
|
40
|
Banerji CRS, Knopp P, Moyle LA, Severini S, Orrell RW, Teschendorff AE, Zammit PS. β-Catenin is central to DUX4-driven network rewiring in facioscapulohumeral muscular dystrophy. J R Soc Interface 2015; 12:20140797. [PMID: 25551153 DOI: 10.1098/rsif.2014.0797] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Facioscapulohumeral muscular dystrophy (FSHD) is an incurable disease, characterized by skeletal muscle weakness and wasting. Genetically, FSHD is characterized by contraction or hypomethylation of repeat D4Z4 units on chromosome 4, which causes aberrant expression of the transcription factor DUX4 from the last repeat. Many genes have been implicated in FSHD pathophysiology, but an integrated molecular model is currently lacking. We developed a novel differential network methodology, Interactome Sparsification and Rewiring (InSpiRe), which detects network rewiring between phenotypes by integrating gene expression data with known protein interactions. Using InSpiRe, we performed a meta-analysis of multiple microarray datasets from FSHD muscle biopsies, then removed secondary rewiring using non-FSHD datasets, to construct a unified network of rewired interactions. Our analysis identified β-catenin as the main coordinator of FSHD-associated protein interaction signalling, with pathways including canonical Wnt, HIF1-α and TNF-α clearly perturbed. To detect transcriptional changes directly elicited by DUX4, gene expression profiling was performed using microarrays on murine myoblasts. This revealed that DUX4 significantly modified expression of the genes in our FSHD network. Furthermore, we experimentally confirmed that Wnt/β-catenin signalling is affected by DUX4 in murine myoblasts. Thus, we provide the first unified molecular map of FSHD signalling, capable of uncovering pathomechanisms and guiding therapeutic development.
Collapse
|
41
|
Figeac N, Zammit PS. Coordinated action of Axin1 and Axin2 suppresses β-catenin to regulate muscle stem cell function. Cell Signal 2015; 27:1652-65. [PMID: 25866367 DOI: 10.1016/j.cellsig.2015.03.025] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2015] [Accepted: 03/23/2015] [Indexed: 01/01/2023]
Abstract
The resident stem cells of skeletal muscle are satellite cells, which are regulated by both canonical and non-canonical Wnt pathways. Canonical Wnt signalling promotes differentiation, and is controlled at many levels, including via Axin1 and Axin2-mediated β-catenin degradation. Axin1 and Axin2 are thought equivalent suppressors of canonical Wnt signalling, although Axin2 is also a Wnt target gene. We show that Axin1 expression was higher in proliferating satellite cells, while Axin2 was up-regulated during differentiation. siRNA-mediated Axin1 knockdown changed cell morphology, suppressed proliferation and promoted myogenic differentiation. Simultaneous knockdown of both Axin1 and β-catenin rescued proliferation and partially, premature differentiation. Surprisingly, retroviral-mediated overexpression of Axin2 was unable to compensate for knockdown of Axin1 in satellite cells, indicating that Axin1 and Axin2 are not fully redundant. Isolated satellite cells from Axin2-null mice also had no major phenotype. However, siRNA-mediated knockdown of Axin1 in Axin2-null cells strongly inhibited proliferation, while inducing differentiation, clear nuclear localisation of β-catenin, up-regulation of canonical Wnt target genes (Axin2, Lef1, Tcf4, Pitx2c and Lgr5) and activation of a TCF reporter construct. Again, concomitant knockdown of Axin1 and β-catenin in Axin2-null satellite cells rescued morphology and proliferation, but only partially prevented precocious differentiation. Thus, Axin1 and Axin2 do not have equivalent functions in satellite cells, but are both involved in repression of Wnt/β-catenin signalling to maintain proliferation and contribute to controlling timely myogenic differentiation.
Collapse
Affiliation(s)
- Nicolas Figeac
- King's College London, Randall Division of Cell & Molecular Biophysics, New Hunt's House, Guy's Campus, United Kingdom.
| | - Peter S Zammit
- King's College London, Randall Division of Cell & Molecular Biophysics, New Hunt's House, Guy's Campus, United Kingdom.
| |
Collapse
|
42
|
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: 6.2] [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
|