1
|
Marano N, Holaska JM. The role of inner nuclear membrane protein emerin in myogenesis. FASEB J 2025; 39:e70514. [PMID: 40178931 PMCID: PMC11967984 DOI: 10.1096/fj.202500323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2025] [Accepted: 03/25/2025] [Indexed: 04/05/2025]
Abstract
Emerin, a ubiquitously expressed inner nuclear membrane protein, plays a central role in maintaining nuclear structure and genomic organization, and in regulating gene expression and cellular signaling pathways. These functions are critical for proper myogenic differentiation and are closely linked to the pathology of Emery-Dreifuss muscular dystrophy 1 (EDMD1), a laminopathy caused by mutations in the EMD gene. Emerin, along with other nuclear lamina proteins, modulates chromatin organization, cell signaling, gene expression, and cellular mechanotransduction, processes essential for muscle development and homeostasis. Loss of emerin function disrupts chromatin localization, causes dysregulated gene expression, and alters nucleoskeletal organization, resulting in impaired myogenic differentiation. Recent findings suggest that emerin tethers repressive chromatin at the nuclear envelope, a process essential for robust myogenesis. This review provides an in-depth discussion of emerin's multifaceted roles in nuclear organization, gene regulation, and cellular signaling, highlighting its importance in myogenic differentiation and disease progression.
Collapse
Affiliation(s)
- Nicholas Marano
- Department of Biomedical SciencesCooper Medical School of Rowan UniversityCamdenNew JerseyUSA
- Rowan‐Virtua School of Translational Biomedical Engineering and SciencesStratfordNew JerseyUSA
| | - James M. Holaska
- Department of Biomedical SciencesCooper Medical School of Rowan UniversityCamdenNew JerseyUSA
- Rowan‐Virtua School of Translational Biomedical Engineering and SciencesStratfordNew JerseyUSA
| |
Collapse
|
2
|
Sutcu HH, Montagne B, Ricchetti M. DNA-PKcs regulates myogenesis in an Akt-dependent manner independent of induced DNA damage. Cell Death Differ 2023; 30:1900-1915. [PMID: 37400716 PMCID: PMC10406879 DOI: 10.1038/s41418-023-01177-2] [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] [Received: 06/23/2022] [Revised: 04/26/2023] [Accepted: 05/03/2023] [Indexed: 07/05/2023] Open
Abstract
Skeletal muscle regeneration relies on muscle stem (satellite) cells. We previously demonstrated that satellite cells efficiently and accurately repair radiation-induced DNA double-strand breaks (DSBs) via the DNA-dependent kinase DNA-PKcs. We show here that DNA-PKcs affects myogenesis independently of its role in DSB repair. Consequently, this process does not require the accumulation of DSBs and it is also independent of caspase-induced DNA damage. We report that in myogenic cells DNA-PKcs is essential for the expression of the differentiation factor Myogenin in an Akt2-dependent manner. DNA-PKcs interacts with the p300-containing complex that activates Myogenin transcription. We show also that SCID mice that are deficient in DNA-PKcs, and are used for transplantation and muscle regeneration studies, display altered myofiber composition and delayed myogenesis upon injury. These defects are exacerbated after repeated injury/regeneration events resulting in reduced muscle size. We thus identify a novel, caspase-independent, regulation of myogenic differentiation, and define a differentiation phase that does not involve the DNA damage/repair process.
Collapse
Affiliation(s)
- Haser Hasan Sutcu
- Institut Pasteur, Team Stability of Nuclear & Mitochondrial DNA, Department of Developmental and Stem Cell Biology, CNRS UMR3738, 75015, Paris, France
- Université Pierre et Marie Curie (Sorbonne Universities, ED515), Paris, France
- Institut de Radioprotection et de Sûrété Nucléaire (IRSN), Radiobiology of Accidental Exposure Laboratory (PSE-SANTE/SERAMED/LRAcc), B.P. 17, 92262 Fontenay-aux-Roses, Cedex, France
| | - Benjamin Montagne
- Institut Pasteur, Team Stability of Nuclear & Mitochondrial DNA, Department of Developmental and Stem Cell Biology, CNRS UMR3738, 75015, Paris, France
- Institut Pasteur, Molecular Mechanisms of Pathological and Physiological Ageing, Department of Developmental and Stem Cell Biology, Paris, France
| | - Miria Ricchetti
- Institut Pasteur, Team Stability of Nuclear & Mitochondrial DNA, Department of Developmental and Stem Cell Biology, CNRS UMR3738, 75015, Paris, France.
- Institut Pasteur, Molecular Mechanisms of Pathological and Physiological Ageing, Department of Developmental and Stem Cell Biology, Paris, France.
| |
Collapse
|
3
|
Den Hartog L, Asakura A. Implications of notch signaling in duchenne muscular dystrophy. Front Physiol 2022; 13:984373. [PMID: 36237531 PMCID: PMC9553129 DOI: 10.3389/fphys.2022.984373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Accepted: 09/05/2022] [Indexed: 11/13/2022] Open
Abstract
This review focuses upon the implications of the Notch signaling pathway in muscular dystrophies, particularly Duchenne muscular dystrophy (DMD): a pervasive and catastrophic condition concerned with skeletal muscle degeneration. Prior work has defined the pathogenesis of DMD, and several therapeutic approaches have been undertaken in order to regenerate skeletal muscle tissue and ameliorate the phenotype. There is presently no cure for DMD, but a promising avenue for novel therapies is inducing muscle regeneration via satellite cells (muscle stem cells). One specific target using this approach is the Notch signaling pathway. The canonical Notch signaling pathway has been well-characterized and it ultimately governs cell fate decision, cell proliferation, and induction of differentiation. Additionally, inhibition of the Notch signaling pathway has been directly implicated in the deficits seen with muscular dystrophies. Here, we explore the connection between the Notch signaling pathway and DMD, as well as how Notch signaling may be targeted to improve the muscle degeneration seen in muscular dystrophies.
Collapse
|
4
|
Rodriguez-Outeiriño L, Hernandez-Torres F, Ramírez-de Acuña F, Matías-Valiente L, Sanchez-Fernandez C, Franco D, Aranega AE. Muscle Satellite Cell Heterogeneity: Does Embryonic Origin Matter? Front Cell Dev Biol 2021; 9:750534. [PMID: 34722534 PMCID: PMC8554119 DOI: 10.3389/fcell.2021.750534] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Accepted: 09/27/2021] [Indexed: 12/25/2022] Open
Abstract
Muscle regeneration is an important homeostatic process of adult skeletal muscle that recapitulates many aspects of embryonic myogenesis. Satellite cells (SCs) are the main muscle stem cells responsible for skeletal muscle regeneration. SCs reside between the myofiber basal lamina and the sarcolemma of the muscle fiber in a quiescent state. However, in response to physiological stimuli or muscle trauma, activated SCs transiently re-enter the cell cycle to proliferate and subsequently exit the cell cycle to differentiate or self-renew. Recent evidence has stated that SCs display functional heterogeneity linked to regenerative capability with an undifferentiated subgroup that is more prone to self-renewal, as well as committed progenitor cells ready for myogenic differentiation. Several lineage tracing studies suggest that such SC heterogeneity could be associated with different embryonic origins. Although it has been established that SCs are derived from the central dermomyotome, how a small subpopulation of the SCs progeny maintain their stem cell identity while most progress through the myogenic program to construct myofibers is not well understood. In this review, we synthesize the works supporting the different developmental origins of SCs as the genesis of their functional heterogeneity.
Collapse
Affiliation(s)
- Lara Rodriguez-Outeiriño
- Department of Experimental Biology, Faculty of Experimental Sciences, University of Jaén, Jaén, Spain
- Medina Foundation, Technology Park of Health Sciences, Granada, Spain
| | - Francisco Hernandez-Torres
- Medina Foundation, Technology Park of Health Sciences, Granada, Spain
- Department of Biochemistry and Molecular Biology III and Immunology, Faculty of Medicine, University of Granada, Granada, Spain
| | - F. Ramírez-de Acuña
- Department of Experimental Biology, Faculty of Experimental Sciences, University of Jaén, Jaén, Spain
- Medina Foundation, Technology Park of Health Sciences, Granada, Spain
| | - Lidia Matías-Valiente
- Department of Experimental Biology, Faculty of Experimental Sciences, University of Jaén, Jaén, Spain
- Medina Foundation, Technology Park of Health Sciences, Granada, Spain
| | - Cristina Sanchez-Fernandez
- Department of Experimental Biology, Faculty of Experimental Sciences, University of Jaén, Jaén, Spain
- Medina Foundation, Technology Park of Health Sciences, Granada, Spain
| | - Diego Franco
- Department of Experimental Biology, Faculty of Experimental Sciences, University of Jaén, Jaén, Spain
- Medina Foundation, Technology Park of Health Sciences, Granada, Spain
| | - Amelia Eva Aranega
- Department of Experimental Biology, Faculty of Experimental Sciences, University of Jaén, Jaén, Spain
- Medina Foundation, Technology Park of Health Sciences, Granada, Spain
| |
Collapse
|
5
|
Sun KX, Jiang XY, Li X, Su YJ, Wang JL, Zhang L, Yang YM, Zhu XJ. Deletion of phosphatidylserine flippase β-subunit Tmem30a in satellite cells leads to delayed skeletal muscle regeneration. Zool Res 2021; 42:650-659. [PMID: 34472226 PMCID: PMC8455468 DOI: 10.24272/j.issn.2095-8137.2021.195] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Abstract
Phosphatidylserine (PS) is distributed asymmetrically in the plasma membrane of eukaryotic cells. Phosphatidylserine flippase (P4-ATPase) transports PS from the outer leaflet of the lipid bilayer to the inner leaflet of the membrane to maintain PS asymmetry. The β subunit TMEM30A is indispensable for transport and proper function of P4-ATPase. Previous studies have shown that the ATP11A and TMEM30A complex is the molecular switch for myotube formation. However, the role of Tmem30a in skeletal muscle regeneration remains elusive. In the current study, Tmem30a was highly expressed in the tibialis anterior (TA) muscles of dystrophin-null (mdx) mice and BaCl2-induced muscle injury model mice. We generated a satellite cell (SC)-specific Tmem30a conditional knockout (cKO) mouse model to investigate the role of Tmem30a in skeletal muscle regeneration. The regenerative ability of cKO mice was evaluated by analyzing the number and diameter of regenerated SCs after the TA muscles were injured by BaCl2-injection. Compared to the control mice, the cKO mice showed decreased Pax7+ and MYH3+ SCs, indicating diminished SC proliferation, and decreased expression of muscular regulatory factors (MYOD and MYOG), suggesting impaired myoblast proliferation in skeletal muscle regeneration. Taken together, these results demonstrate the essential role of Tmem30a in skeletal muscle regeneration.
Collapse
Affiliation(s)
- Kuan-Xiang Sun
- Health Management Center, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan 610072, China.,Sichuan Provincial Key Laboratory for Human Disease Gene Study, Center for Medical Genetics, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan 610072, China.,Research Unit for Blindness Prevention of Chinese Academy of Medical Sciences (2019RU026), Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu, Sichuan 610072 China
| | - Xiao-Yan Jiang
- Health Management Center, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan 610072, China.,Sichuan Provincial Key Laboratory for Human Disease Gene Study, Center for Medical Genetics, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan 610072, China
| | - Xiao Li
- Health Management Center, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan 610072, China.,Sichuan Provincial Key Laboratory for Human Disease Gene Study, Center for Medical Genetics, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan 610072, China
| | - Yu-Jing Su
- Sichuan Provincial Key Laboratory for Human Disease Gene Study, Center for Medical Genetics, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan 610072, China
| | - Ju-Lin Wang
- Sichuan Provincial Key Laboratory for Human Disease Gene Study, Center for Medical Genetics, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan 610072, China
| | - Lin Zhang
- Sichuan Provincial Key Laboratory for Human Disease Gene Study, Center for Medical Genetics, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan 610072, China
| | - Ye-Ming Yang
- Sichuan Provincial Key Laboratory for Human Disease Gene Study, Center for Medical Genetics, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan 610072, China
| | - Xian-Jun Zhu
- Health Management Center, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan 610072, China.,Sichuan Provincial Key Laboratory for Human Disease Gene Study, Center for Medical Genetics, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan 610072, China.,Research Unit for Blindness Prevention of Chinese Academy of Medical Sciences (2019RU026), Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu, Sichuan 610072 China.,Department of Ophthalmology, First People's Hospital of Shangqiu, Shangqiu, Henan 476000, China.,Key Laboratory of Tibetan Medicine Research, Chinese Academy of Sciences and Qinghai Provincial Key Laboratory of Tibetan Medicine Research, Northwest Institute of Plateau Biology, Xining, Qinghai 810008, China. E-mail:
| |
Collapse
|
6
|
Yuan S, Zheng S, Zheng K, Gao Y, Chen M, Li Y, Bai X. Sympathetic activity is correlated with satellite cell aging and myogenesis via β2-adrenoceptor. Stem Cell Res Ther 2021; 12:505. [PMID: 34530910 PMCID: PMC8447727 DOI: 10.1186/s13287-021-02571-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Accepted: 08/26/2021] [Indexed: 02/09/2023] Open
Abstract
Background and objective Sympathetic activity plays an important role in the proliferation and differentiation of stem cells, and it changes over time, thereby exerting differential effects on various stem cell types. Aging causes sympathetic hyperactivity in aged tissues and blunts sympathetic nerves regulation, and sympathetic abnormalities play a role in aging-related diseases. Currently, the effect of sympathetic activity on skeletal muscle stem cells, namely satellite cells (SCs), is unclear. This study aimed to investigate the effects of skeletal muscle sympathetic activity on SC aging and skeletal muscle repair. Materials and methods To evaluate skeletal muscle and fibrotic areas, numbers of SCs and myonuclei per muscle fiber, β2-adrenoceptor (β2-ADR) expression, muscle repair, and sympathetic innervation in skeletal muscle, aged mice, young mice that underwent chemical sympathectomy (CS) were utilized. Mice with a tibialis anterior muscle injury were treated by barium chloride (BaCl2) and clenbuterol (CLB) in vivo. SCs or C2C12 cells were differentiated into myotubes and treated with or without CLB. Immunofluorescence, western blot, sirius red, and hematoxylin–eosin were used to evaluate SCs, myogenic repair and differentiation. Results The number of SCs, sympathetic activity, and reparability of muscle injury in aged mice were significantly decreased, compared with those in young mice. The above characteristics of young mice that underwent CS were similar to those of aged mice. While CLB promoted the repair of muscle injury in aged and CS mice and ameliorated the reduction in the SC number and sympathetic activity, the effects of CLB on the SCs and sympathetic nerves in young mice were not significant. CLB inhibited the myogenic differentiation of C2C12 cells in vitro. We further found that NF-κB and ERK1/2 signaling pathways were activated during myogenic differentiation, and this process could be inhibited by CLB. Conclusion Normal sympathetic activity promoted the stemness of SCs to thereby maintain a steady state. It also could maintain total and self-renewing number of SCs and maintain a quiescent state, which was correlated with skeletal SCs via β2-ADR. Normal sympathetic activity was also beneficial for the myogenic repair of injured skeletal muscle. Supplementary Information The online version contains supplementary material available at 10.1186/s13287-021-02571-8.
Collapse
Affiliation(s)
- Shiguo Yuan
- Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Center for Orthopaedic Surgery, The Third Affiliated Hospital, Southern Medical University, Guangzhou, 510630, China.,Department of Orthopaedic Surgery, Hainan Province Hospital of Traditional Chinese Medicine, Haikou, 570203, China
| | - Sheng Zheng
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Kai Zheng
- School of Basic Medical Sciences, Youjiang Medical University for Nationalities, Baise, 533000, China
| | - Yanping Gao
- Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Center for Orthopaedic Surgery, The Third Affiliated Hospital, Southern Medical University, Guangzhou, 510630, China
| | - Meixiong Chen
- Department of Orthopaedic Surgery, Hainan Province Hospital of Traditional Chinese Medicine, Haikou, 570203, China
| | - Yikai Li
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, China.
| | - Xiaochun Bai
- Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Center for Orthopaedic Surgery, The Third Affiliated Hospital, Southern Medical University, Guangzhou, 510630, China.
| |
Collapse
|
7
|
Westman AM, Peirce SM, Christ GJ, Blemker SS. Agent-based model provides insight into the mechanisms behind failed regeneration following volumetric muscle loss injury. PLoS Comput Biol 2021; 17:e1008937. [PMID: 33970905 PMCID: PMC8110270 DOI: 10.1371/journal.pcbi.1008937] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Accepted: 04/01/2021] [Indexed: 12/22/2022] Open
Abstract
Skeletal muscle possesses a remarkable capacity for repair and regeneration following a variety of injuries. When successful, this highly orchestrated regenerative process requires the contribution of several muscle resident cell populations including satellite stem cells (SSCs), fibroblasts, macrophages and vascular cells. However, volumetric muscle loss injuries (VML) involve simultaneous destruction of multiple tissue components (e.g., as a result of battlefield injuries or vehicular accidents) and are so extensive that they exceed the intrinsic capability for scarless wound healing and result in permanent cosmetic and functional deficits. In this scenario, the regenerative process fails and is dominated by an unproductive inflammatory response and accompanying fibrosis. The failure of current regenerative therapeutics to completely restore functional muscle tissue is not surprising considering the incomplete understanding of the cellular mechanisms that drive the regeneration response in the setting of VML injury. To begin to address this profound knowledge gap, we developed an agent-based model to predict the tissue remodeling response following surgical creation of a VML injury. Once the model was able to recapitulate key aspects of the tissue remodeling response in the absence of repair, we validated the model by simulating the tissue remodeling response to VML injury following implantation of either a decellularized extracellular matrix scaffold or a minced muscle graft. The model suggested that the SSC microenvironment and absence of pro-differentiation SSC signals were the most important aspects of failed muscle regeneration in VML injuries. The major implication of this work is that agent-based models may provide a much-needed predictive tool to optimize the design of new therapies, and thereby, accelerate the clinical translation of regenerative therapeutics for VML injuries.
Collapse
Affiliation(s)
- Amanda M. Westman
- Biomedical Engineering, University of Virginia, Charlottesville, Virginia, United States of America
| | - Shayn M. Peirce
- Biomedical Engineering, University of Virginia, Charlottesville, Virginia, United States of America
- Ophthalmology, University of Virginia, Charlottesville, Virginia, United States of America
| | - George J. Christ
- Biomedical Engineering, University of Virginia, Charlottesville, Virginia, United States of America
- Orthopaedic Surgery, University of Virginia, Charlottesville, Virginia, United States of America
- * E-mail: (GJC); (SSB)
| | - Silvia S. Blemker
- Biomedical Engineering, University of Virginia, Charlottesville, Virginia, United States of America
- Ophthalmology, University of Virginia, Charlottesville, Virginia, United States of America
- Orthopaedic Surgery, University of Virginia, Charlottesville, Virginia, United States of America
- Mechanical and Aerospace Engineering, University of Virginia, Charlottesville, Virginia, United States of America
- * E-mail: (GJC); (SSB)
| |
Collapse
|
8
|
Coppens S, Barnard AM, Puusepp S, Pajusalu S, Õunap K, Vargas-Franco D, Bruels CC, Donkervoort S, Pais L, Chao KR, Goodrich JK, England EM, Weisburd B, Ganesh VS, Gudmundsson S, O'Donnell-Luria A, Nigul M, Ilves P, Mohassel P, Siddique T, Milone M, Nicolau S, Maroofian R, Houlden H, Hanna MG, Quinlivan R, Beiraghi Toosi M, Ghayoor Karimiani E, Costagliola S, Deconinck N, Kadhim H, Macke E, Lanpher BC, Klee EW, Łusakowska A, Kostera-Pruszczyk A, Hahn A, Schrank B, Nishino I, Ogasawara M, El Sherif R, Stojkovic T, Nelson I, Bonne G, Cohen E, Boland-Augé A, Deleuze JF, Meng Y, Töpf A, Vilain C, Pacak CA, Rivera-Zengotita ML, Bönnemann CG, Straub V, Handford PA, Draper I, Walter GA, Kang PB. A form of muscular dystrophy associated with pathogenic variants in JAG2. Am J Hum Genet 2021; 108:840-856. [PMID: 33861953 PMCID: PMC8206160 DOI: 10.1016/j.ajhg.2021.03.020] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 03/26/2021] [Indexed: 02/09/2023] Open
Abstract
JAG2 encodes the Notch ligand Jagged2. The conserved Notch signaling pathway contributes to the development and homeostasis of multiple tissues, including skeletal muscle. We studied an international cohort of 23 individuals with genetically unsolved muscular dystrophy from 13 unrelated families. Whole-exome sequencing identified rare homozygous or compound heterozygous JAG2 variants in all 13 families. The identified bi-allelic variants include 10 missense variants that disrupt highly conserved amino acids, a nonsense variant, two frameshift variants, an in-frame deletion, and a microdeletion encompassing JAG2. Onset of muscle weakness occurred from infancy to young adulthood. Serum creatine kinase (CK) levels were normal or mildly elevated. Muscle histology was primarily dystrophic. MRI of the lower extremities revealed a distinct, slightly asymmetric pattern of muscle involvement with cores of preserved and affected muscles in quadriceps and tibialis anterior, in some cases resembling patterns seen in POGLUT1-associated muscular dystrophy. Transcriptome analysis of muscle tissue from two participants suggested misregulation of genes involved in myogenesis, including PAX7. In complementary studies, Jag2 downregulation in murine myoblasts led to downregulation of multiple components of the Notch pathway, including Megf10. Investigations in Drosophila suggested an interaction between Serrate and Drpr, the fly orthologs of JAG1/JAG2 and MEGF10, respectively. In silico analysis predicted that many Jagged2 missense variants are associated with structural changes and protein misfolding. In summary, we describe a muscular dystrophy associated with pathogenic variants in JAG2 and evidence suggests a disease mechanism related to Notch pathway dysfunction.
Collapse
Affiliation(s)
- Sandra Coppens
- Center of Human Genetics, Université Libre de Bruxelles, 1070 Brussels, Belgium
| | - Alison M Barnard
- Department of Physical Therapy, University of Florida College of Public Health and Health Professions, Gainesville, FL 32610, USA
| | - Sanna Puusepp
- Department of Clinical Genetics, United Laboratories, Tartu University Hospital, Tartu 50406, Estonia; Institute of Clinical Medicine, University of Tartu, Tartu 50406, Estonia
| | - Sander Pajusalu
- Department of Clinical Genetics, United Laboratories, Tartu University Hospital, Tartu 50406, Estonia; Institute of Clinical Medicine, University of Tartu, Tartu 50406, Estonia
| | - Katrin Õunap
- Department of Clinical Genetics, United Laboratories, Tartu University Hospital, Tartu 50406, Estonia; Institute of Clinical Medicine, University of Tartu, Tartu 50406, Estonia
| | - Dorianmarie Vargas-Franco
- Division of Pediatric Neurology, Department of Pediatrics, University of Florida College of Medicine, Gainesville, FL 32610, USA
| | - Christine C Bruels
- Division of Pediatric Neurology, Department of Pediatrics, University of Florida College of Medicine, Gainesville, FL 32610, USA
| | - Sandra Donkervoort
- Neuromuscular and Neurogenetic Disorders of Childhood Section, Neurogenetics Branch, NINDS, NIH, Bethesda, MD 20892, USA
| | - Lynn Pais
- Broad Center for Mendelian Genomics, Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Analytic and Translational Genetics Unit and Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Katherine R Chao
- Broad Center for Mendelian Genomics, Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Analytic and Translational Genetics Unit and Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Julia K Goodrich
- Broad Center for Mendelian Genomics, Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Analytic and Translational Genetics Unit and Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Eleina M England
- Broad Center for Mendelian Genomics, Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Analytic and Translational Genetics Unit and Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA 02114, USA; Division of Genetics and Genomics, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Ben Weisburd
- Broad Center for Mendelian Genomics, Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Analytic and Translational Genetics Unit and Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Vijay S Ganesh
- Broad Center for Mendelian Genomics, Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Analytic and Translational Genetics Unit and Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA 02114, USA; Department of Neurology, Brigham & Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Sanna Gudmundsson
- Broad Center for Mendelian Genomics, Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Analytic and Translational Genetics Unit and Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA 02114, USA; Division of Genetics and Genomics, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Anne O'Donnell-Luria
- Broad Center for Mendelian Genomics, Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Analytic and Translational Genetics Unit and Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA 02114, USA; Division of Genetics and Genomics, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Mait Nigul
- Department of Radiology, Tartu University Hospital, Tartu 50406, Estonia
| | - Pilvi Ilves
- Institute of Clinical Medicine, University of Tartu, Tartu 50406, Estonia; Department of Radiology, Tartu University Hospital, Tartu 50406, Estonia
| | - Payam Mohassel
- Neuromuscular and Neurogenetic Disorders of Childhood Section, Neurogenetics Branch, NINDS, NIH, Bethesda, MD 20892, USA
| | - Teepu Siddique
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | | | - Stefan Nicolau
- Department of Neurology, Mayo Clinic, Rochester, MN 55905, USA
| | - Reza Maroofian
- Department of Neuromuscular Disorders, University College London Institute of Neurology, London WC1E 6BT, UK
| | - Henry Houlden
- Department of Neuromuscular Disorders, University College London Institute of Neurology, London WC1E 6BT, UK
| | - Michael G Hanna
- Department of Neuromuscular Disorders, University College London Institute of Neurology, London WC1E 6BT, UK
| | - Ros Quinlivan
- Department of Neuromuscular Disorders, University College London Institute of Neurology, London WC1E 6BT, UK
| | - Mehran Beiraghi Toosi
- Pediatric Neurology Department, Ghaem Hospital, Mashhad University of Medical Sciences, Mashhad 9176999311, Iran
| | - Ehsan Ghayoor Karimiani
- Molecular and Clinical Sciences Institute, St. George's, University of London, Cranmer Terrace, London SW17 0RE, UK; Innovative Medical Research Center, Mashhad Branch, Islamic Azad University, Mashhad 9187147578, Iran
| | - Sabine Costagliola
- Institut de Recherche Interdisciplinaire en Biologie Humaine et Moleculaire, Université Libre de Bruxelles, 1070 Brussels, Belgium
| | - Nicolas Deconinck
- Centre de Référence Neuromusculaire and Paediatric Neurology Department, Hôpital Universitaire des Enfants Reine Fabiola, Université Libre de Bruxelles, 1020 Brussels, Belgium
| | - Hazim Kadhim
- Neuropathology Unit, Department of Anatomic Pathology and Reference Center for Neuromuscular Pathology, Brugmann University Hospital-Children's Hospital, Université Libre de Bruxelles, 1020 Brussels, Belgium
| | - Erica Macke
- Center for Individualized Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | - Brendan C Lanpher
- Center for Individualized Medicine, Mayo Clinic, Rochester, MN 55905, USA; Department of Clinical Genomics, Mayo Clinic, Rochester, MN 55905, USA
| | - Eric W Klee
- Center for Individualized Medicine, Mayo Clinic, Rochester, MN 55905, USA; Department of Clinical Genomics, Mayo Clinic, Rochester, MN 55905, USA
| | - Anna Łusakowska
- Department of Neurology, Medical University of Warsaw, 02-091 Warsaw, Poland
| | | | - Andreas Hahn
- Department of Child Neurology, Justus-Liebig-University Giessen, 35390 Giessen, Germany
| | - Bertold Schrank
- Department of Neurology, DKD HELIOS Klinik Wiesbaden, 65191 Wiesbaden, Germany
| | - Ichizo Nishino
- Department of Neuromuscular Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Tokyo 187-8551, Japan
| | - Masashi Ogasawara
- Department of Neuromuscular Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Tokyo 187-8551, Japan
| | - Rasha El Sherif
- Myo-Care Neuromuscular Center, Myo-Care National Foundation, Cairo 11865, Egypt
| | - Tanya Stojkovic
- APHP, Nord-Est/Ile-de-France Neuromuscular Reference Center, Myology Institute, Pitié-Salpêtrière Hospital, 75013 Paris, France; Sorbonne Université, INSERM, Center of Research in Myology, UMRS974, 75651 Paris Cedex 13, France
| | - Isabelle Nelson
- Sorbonne Université, INSERM, Center of Research in Myology, UMRS974, 75651 Paris Cedex 13, France
| | - Gisèle Bonne
- Sorbonne Université, INSERM, Center of Research in Myology, UMRS974, 75651 Paris Cedex 13, France
| | - Enzo Cohen
- Sorbonne Université, INSERM, Center of Research in Myology, UMRS974, 75651 Paris Cedex 13, France
| | - Anne Boland-Augé
- Université Paris-Saclay, CEA, Centre National de Recherche en Génomique Humaine, 91057 Evry, France
| | - Jean-François Deleuze
- Université Paris-Saclay, CEA, Centre National de Recherche en Génomique Humaine, 91057 Evry, France
| | - Yao Meng
- Department of Biochemistry, University of Oxford, Oxford OX1 3QU, UK
| | - Ana Töpf
- John Walton Muscular Dystrophy Research Centre, Newcastle University and Newcastle Hospitals NHS Foundation Trust, Newcastle upon Tyne NE1 3BZ, UK
| | - Catheline Vilain
- Center of Human Genetics, Université Libre de Bruxelles, 1070 Brussels, Belgium
| | - Christina A Pacak
- Department of Pediatrics, University of Florida College of Medicine, Gainesville, FL 32610, USA; Paul and Sheila Wellstone Muscular Dystrophy Center, University of Minnesota Medical School, Minneapolis, MN 55455, USA; Department of Neurology, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| | | | - Carsten G Bönnemann
- Neuromuscular and Neurogenetic Disorders of Childhood Section, Neurogenetics Branch, NINDS, NIH, Bethesda, MD 20892, USA
| | - Volker Straub
- John Walton Muscular Dystrophy Research Centre, Newcastle University and Newcastle Hospitals NHS Foundation Trust, Newcastle upon Tyne NE1 3BZ, UK
| | - Penny A Handford
- Department of Biochemistry, University of Oxford, Oxford OX1 3QU, UK
| | - Isabelle Draper
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA 02111, USA
| | - Glenn A Walter
- Department of Physiology and Functional Genomics, University of Florida College of Medicine, Gainesville, FL 32610, USA
| | - Peter B Kang
- Division of Pediatric Neurology, Department of Pediatrics, University of Florida College of Medicine, Gainesville, FL 32610, USA; Paul and Sheila Wellstone Muscular Dystrophy Center, University of Minnesota Medical School, Minneapolis, MN 55455, USA; Department of Neurology, University of Minnesota Medical School, Minneapolis, MN 55455, USA; Institute for Translational Neuroscience, University of Minnesota Medical School, Minneapolis, MN 55455, USA.
| |
Collapse
|
9
|
Moriscot A, Miyabara EH, Langeani B, Belli A, Egginton S, Bowen TS. Firearms-related skeletal muscle trauma: pathophysiology and novel approaches for regeneration. NPJ Regen Med 2021; 6:17. [PMID: 33772028 PMCID: PMC7997931 DOI: 10.1038/s41536-021-00127-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Accepted: 02/24/2021] [Indexed: 02/07/2023] Open
Abstract
One major cause of traumatic injury is firearm-related wounds (i.e., ballistic trauma), common in both civilian and military populations, which is increasing in prevalence and has serious long-term health and socioeconomic consequences worldwide. Common primary injuries of ballistic trauma include soft-tissue damage and loss, haemorrhage, bone fracture, and pain. The majority of injuries are of musculoskeletal origin and located in the extremities, such that skeletal muscle offers a major therapeutic target to aid recovery and return to normal daily activities. However, the underlying pathophysiology of skeletal muscle ballistic trauma remains poorly understood, with limited evidence-based treatment options. As such, this review will address the topic of firearm-related skeletal muscle injury and regeneration. We first introduce trauma ballistics and the immediate injury of skeletal muscle, followed by detailed coverage of the underlying biological mechanisms involved in regulating skeletal muscle dysfunction following injury, with a specific focus on the processes of muscle regeneration, muscle wasting and vascular impairments. Finally, we evaluate novel approaches for minimising muscle damage and enhancing muscle regeneration after ballistic trauma, which may have important relevance for primary care in victims of violence.
Collapse
Affiliation(s)
- Anselmo Moriscot
- Department of Anatomy, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Elen H Miyabara
- Department of Anatomy, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | | | - Antonio Belli
- NIHR Surgical Reconstruction and Microbiology Research Centre, University of Birmingham, Birmingham, UK
| | - Stuart Egginton
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds, UK
| | - T Scott Bowen
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds, UK.
| |
Collapse
|
10
|
Fang Y, Liang F, Yuan R, Zhu Q, Cai S, Chen K, Zhang J, Luo X, Chen Y, Mo D. High mobility group box 2 regulates skeletal muscle development through ribosomal protein S6 kinase 1. FASEB J 2020; 34:12367-12378. [DOI: 10.1096/fj.202001183r] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 06/30/2020] [Accepted: 07/02/2020] [Indexed: 01/01/2023]
Affiliation(s)
- Ying Fang
- State Key Laboratory of Biocontrol School of Life Sciences Sun Yat‐sen University Guangzhou China
| | - Feng Liang
- State Key Laboratory of Biocontrol School of Life Sciences Sun Yat‐sen University Guangzhou China
| | - Renqiang Yuan
- State Key Laboratory of Biocontrol School of Life Sciences Sun Yat‐sen University Guangzhou China
| | - Qi Zhu
- State Key Laboratory of Biocontrol School of Life Sciences Sun Yat‐sen University Guangzhou China
| | - Shufang Cai
- State Key Laboratory of Biocontrol School of Life Sciences Sun Yat‐sen University Guangzhou China
| | - Keren Chen
- State Key Laboratory of Biocontrol School of Life Sciences Sun Yat‐sen University Guangzhou China
| | - Junyan Zhang
- State Key Laboratory of Biocontrol School of Life Sciences Sun Yat‐sen University Guangzhou China
| | - Xiaorong Luo
- State Key Laboratory of Biocontrol School of Life Sciences Sun Yat‐sen University Guangzhou China
| | - Yaosheng Chen
- State Key Laboratory of Biocontrol School of Life Sciences Sun Yat‐sen University Guangzhou China
| | - Delin Mo
- State Key Laboratory of Biocontrol School of Life Sciences Sun Yat‐sen University Guangzhou China
| |
Collapse
|
11
|
Cappellari O, Mantuano P, De Luca A. "The Social Network" and Muscular Dystrophies: The Lesson Learnt about the Niche Environment as a Target for Therapeutic Strategies. Cells 2020; 9:cells9071659. [PMID: 32660168 PMCID: PMC7407800 DOI: 10.3390/cells9071659] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Revised: 07/01/2020] [Accepted: 07/02/2020] [Indexed: 12/12/2022] Open
Abstract
The muscle stem cells niche is essential in neuromuscular disorders. Muscle injury and myofiber death are the main triggers of muscle regeneration via satellite cell activation. However, in degenerative diseases such as muscular dystrophy, regeneration still keep elusive. In these pathologies, stem cell loss occurs over time, and missing signals limiting damaged tissue from activating the regenerative process can be envisaged. It is unclear what comes first: the lack of regeneration due to satellite cell defects, their pool exhaustion for degeneration/regeneration cycles, or the inhibitory mechanisms caused by muscle damage and fibrosis mediators. Herein, Duchenne muscular dystrophy has been taken as a paradigm, as several drugs have been tested at the preclinical and clinical levels, targeting secondary events in the complex pathogenesis derived from lack of dystrophin. We focused on the crucial roles that pro-inflammatory and pro-fibrotic cytokines play in triggering muscle necrosis after damage and stimulating satellite cell activation and self-renewal, along with growth and mechanical factors. These processes contribute to regeneration and niche maintenance. We review the main effects of drugs on regeneration biomarkers to assess whether targeting pathogenic events can help to protect niche homeostasis and enhance regeneration efficiency other than protecting newly formed fibers from further damage.
Collapse
|
12
|
Fukada SI, Akimoto T, Sotiropoulos A. Role of damage and management in muscle hypertrophy: Different behaviors of muscle stem cells in regeneration and hypertrophy. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2020; 1867:118742. [PMID: 32417255 DOI: 10.1016/j.bbamcr.2020.118742] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 05/07/2020] [Accepted: 05/12/2020] [Indexed: 12/12/2022]
Abstract
Skeletal muscle is a dynamic tissue with two unique abilities; one is its excellent regenerative ability, due to the activity of skeletal muscle-resident stem cells named muscle satellite cells (MuSCs); and the other is the adaptation of myofiber size in response to external stimulation, intrinsic factors, or physical activity, which is known as plasticity. Low physical activity and some disease conditions lead to the reduction of myofiber size, called atrophy, whereas hypertrophy refers to the increase in myofiber size induced by high physical activity or anabolic hormones/drugs. MuSCs are essential for generating new myofibers during regeneration and the increase in new myonuclei during hypertrophy; however, there has been little investigation of the molecular mechanisms underlying MuSC activation, proliferation, and differentiation during hypertrophy compared to those of regeneration. One reason is that 'degenerative damage' to myofibers during muscle injury or upon hypertrophy (especially overloaded muscle) is believed to trigger similar activation/proliferation of MuSCs. However, evidence suggests that degenerative damage of myofibers is not necessary for MuSC activation/proliferation during hypertrophy. When considering MuSC-based therapy for atrophy, including sarcopenia, it will be indispensable to elucidate MuSC behaviors in muscles that exhibit non-degenerative damage, because degenerated myofibers are not present in the atrophied muscles. In this review, we summarize recent findings concerning the relationship between MuSCs and hypertrophy, and discuss what remains to be discovered to inform the development and application of relevant treatments for muscle atrophy.
Collapse
Affiliation(s)
- So-Ichiro Fukada
- Project for Muscle Stem Cell Biology, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan.
| | | | - Athanassia Sotiropoulos
- Institut National de la Santé et de la Recherche Médicale U1016, Institut Cochin, Paris, France
| |
Collapse
|
13
|
Barruet E, Garcia SM, Striedinger K, Wu J, Lee S, Byrnes L, Wong A, Xuefeng S, Tamaki S, Brack AS, Pomerantz JH. Functionally heterogeneous human satellite cells identified by single cell RNA sequencing. eLife 2020; 9:51576. [PMID: 32234209 PMCID: PMC7164960 DOI: 10.7554/elife.51576] [Citation(s) in RCA: 76] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Accepted: 03/27/2020] [Indexed: 12/19/2022] Open
Abstract
Although heterogeneity is recognized within the murine satellite cell pool, a comprehensive understanding of distinct subpopulations and their functional relevance in human satellite cells is lacking. We used a combination of single cell RNA sequencing and flow cytometry to identify, distinguish, and physically separate novel subpopulations of human PAX7+ satellite cells (Hu-MuSCs) from normal muscles. We found that, although relatively homogeneous compared to activated satellite cells and committed progenitors, the Hu-MuSC pool contains clusters of transcriptionally distinct cells with consistency across human individuals. New surface marker combinations were enriched in transcriptional subclusters, including a subpopulation of Hu-MuSCs marked by CXCR4/CD29/CD56/CAV1 (CAV1+). In vitro, CAV1+ Hu-MuSCs are morphologically distinct, and characterized by resistance to activation compared to CAV1- Hu-MuSCs. In vivo, CAV1+ Hu-MuSCs demonstrated increased engraftment after transplantation. Our findings provide a comprehensive transcriptional view of normal Hu-MuSCs and describe new heterogeneity, enabling separation of functionally distinct human satellite cell subpopulations.
Collapse
Affiliation(s)
- Emilie Barruet
- Departments of Surgery and Orofacial Sciences, Division of Plastic and Reconstructive Surgery, Program in Craniofacial Biology, Eli and Edythe Broad Center of Regeneration Medicine, University of California, San Francisco, San Francisco, United States
| | - Steven M Garcia
- Departments of Surgery and Orofacial Sciences, Division of Plastic and Reconstructive Surgery, Program in Craniofacial Biology, Eli and Edythe Broad Center of Regeneration Medicine, University of California, San Francisco, San Francisco, United States
| | - Katharine Striedinger
- Departments of Surgery and Orofacial Sciences, Division of Plastic and Reconstructive Surgery, Program in Craniofacial Biology, Eli and Edythe Broad Center of Regeneration Medicine, University of California, San Francisco, San Francisco, United States
| | - Jake Wu
- Departments of Surgery and Orofacial Sciences, Division of Plastic and Reconstructive Surgery, Program in Craniofacial Biology, Eli and Edythe Broad Center of Regeneration Medicine, University of California, San Francisco, San Francisco, United States
| | - Solomon Lee
- Departments of Surgery and Orofacial Sciences, Division of Plastic and Reconstructive Surgery, Program in Craniofacial Biology, Eli and Edythe Broad Center of Regeneration Medicine, University of California, San Francisco, San Francisco, United States
| | - Lauren Byrnes
- University of California San Francisco, San Francisco, United States
| | - Alvin Wong
- Departments of Surgery and Orofacial Sciences, Division of Plastic and Reconstructive Surgery, Program in Craniofacial Biology, Eli and Edythe Broad Center of Regeneration Medicine, University of California, San Francisco, San Francisco, United States
| | - Sun Xuefeng
- Department of Orthopedic Surgery, Eli and Edythe Broad Center of Regeneration Medicine, University of California, San Francisco, San Francisco, United States
| | - Stanley Tamaki
- Departments of Surgery and Orofacial Sciences, Division of Plastic and Reconstructive Surgery, Program in Craniofacial Biology, Eli and Edythe Broad Center of Regeneration Medicine, University of California, San Francisco, San Francisco, United States
| | - Andrew S Brack
- Department of Orthopedic Surgery, Eli and Edythe Broad Center of Regeneration Medicine, University of California, San Francisco, San Francisco, United States
| | - Jason H Pomerantz
- Departments of Surgery and Orofacial Sciences, Division of Plastic and Reconstructive Surgery, Program in Craniofacial Biology, Eli and Edythe Broad Center of Regeneration Medicine, University of California, San Francisco, San Francisco, United States
| |
Collapse
|
14
|
Rion N, Castets P, Lin S, Enderle L, Reinhard JR, Rüegg MA. mTORC2 affects the maintenance of the muscle stem cell pool. Skelet Muscle 2019; 9:30. [PMID: 31791403 PMCID: PMC6886171 DOI: 10.1186/s13395-019-0217-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Accepted: 10/23/2019] [Indexed: 01/15/2023] Open
Abstract
Background The mammalian target of rapamycin complex 2 (mTORC2), containing the essential protein rictor, regulates cellular metabolism and cytoskeletal organization by phosphorylating protein kinases, such as PKB/Akt, PKC, and SGK. Inactivation of mTORC2 signaling in adult skeletal muscle affects its metabolism, but not muscle morphology and function. However, the role of mTORC2 in adult muscle stem cells (MuSCs) has not been investigated. Method Using histological, biochemical, and molecular biological methods, we characterized the muscle phenotype of mice depleted for rictor in the Myf5-lineage (RImyfKO) and of mice depleted for rictor in skeletal muscle fibers (RImKO). The proliferative and myogenic potential of MuSCs was analyzed upon cardiotoxin-induced injury in vivo and in isolated myofibers in vitro. Results Skeletal muscle of young and 14-month-old RImyfKO mice appeared normal in composition and function. MuSCs from young RImyfKO mice exhibited a similar capacity to proliferate, differentiate, and fuse as controls. In contrast, the number of MuSCs was lower in young RImyfKO mice than in controls after two consecutive rounds of cardiotoxin-induced muscle regeneration. Similarly, the number of MuSCs in RImyfKO mice decreased with age, which correlated with a decline in the regenerative capacity of mutant muscle. Interestingly, reduction in the number of MuSCs was also observed in 14-month-old RImKO muscle. Conclusions Our study shows that mTORC2 signaling is dispensable for myofiber formation, but contributes to the homeostasis of MuSCs. Loss of mTORC2 does not affect their myogenic function, but impairs the replenishment of MuSCs after repeated injuries and their maintenance during aging. These results point to an important role of mTORC2 signaling in MuSC for muscle homeostasis.
Collapse
Affiliation(s)
- Nathalie Rion
- Biozentrum, University of Basel, CH-4056, Basel, Switzerland
| | - Perrine Castets
- Biozentrum, University of Basel, CH-4056, Basel, Switzerland.,Department PHYM, Centre Médical Universitaire de Genève, Geneva, Switzerland
| | - Shuo Lin
- Biozentrum, University of Basel, CH-4056, Basel, Switzerland
| | - Leonie Enderle
- Biozentrum, University of Basel, CH-4056, Basel, Switzerland.,Toronto Recombinant Antibody Centre/The Donnelly Centre, University of Toronto, M5G 1 L6, Toronto, ON, Canada
| | | | - Markus A Rüegg
- Biozentrum, University of Basel, CH-4056, Basel, Switzerland.
| |
Collapse
|
15
|
Zhu X, Li M, Jia X, Hou W, Yang J, Zhao H, Wang G, Wang J. The homeoprotein Msx1 cooperates with Pkn1 to prevent terminal differentiation in myogenic precursor cells. Biochimie 2019; 162:55-65. [DOI: 10.1016/j.biochi.2019.04.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Accepted: 04/03/2019] [Indexed: 12/22/2022]
|
16
|
Evano B, Tajbakhsh S. Skeletal muscle stem cells in comfort and stress. NPJ Regen Med 2018; 3:24. [PMID: 30588332 PMCID: PMC6303387 DOI: 10.1038/s41536-018-0062-3] [Citation(s) in RCA: 78] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Accepted: 11/28/2018] [Indexed: 12/21/2022] Open
Abstract
Investigations on developmental and regenerative myogenesis have led to major advances in decrypting stem cell properties and potential, as well as their interactions within the evolving niche. As a consequence, regenerative myogenesis has provided a forum to investigate intrinsic regulators of stem cell properties as well as extrinsic factors, including stromal cells, during normal growth and following injury and disease. Here we review some of the latest advances in the field that have exposed fundamental processes including regulation of stress following trauma and ageing, senescence, DNA damage control and modes of symmetric and asymmetric cell divisions. Recent studies have begun to explore the nature of the niche that is distinct in different muscle groups, and that is altered from prenatal to postnatal stages, and during ageing. We also discuss heterogeneities among muscle stem cells and how distinct properties within the quiescent and proliferating cell states might impact on homoeostasis and regeneration. Interestingly, cellular quiescence, which was thought to be a passive cell state, is regulated by multiple mechanisms, many of which are deregulated in various contexts including ageing. These and other factors including metabolic activity and genetic background can impact on the efficiency of muscle regeneration.
Collapse
Affiliation(s)
- Brendan Evano
- Stem Cells and Development, Department of Developmental & Stem Cell Biology, Institut Pasteur, 75015 Paris, France
- CNRS UMR 3738, Institut Pasteur, 75015 Paris, France
| | - Shahragim Tajbakhsh
- Stem Cells and Development, Department of Developmental & Stem Cell Biology, Institut Pasteur, 75015 Paris, France
- CNRS UMR 3738, Institut Pasteur, 75015 Paris, France
| |
Collapse
|
17
|
Gayraud-Morel B, Le Bouteiller M, Commere PH, Cohen-Tannoudji M, Tajbakhsh S. Notchless defines a stage-specific requirement for ribosome biogenesis during lineage progression in adult skeletal myogenesis. Development 2018; 145:145/23/dev162636. [PMID: 30478226 DOI: 10.1242/dev.162636] [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] [Received: 12/18/2017] [Accepted: 10/02/2018] [Indexed: 11/20/2022]
Abstract
Cell fate decisions occur through the action of multiple factors, including signalling molecules and transcription factors. Recently, the regulation of translation has emerged as an important step for modulating cellular function and fate, as exemplified by ribosomes that play distinct roles in regulating cell behaviour. Notchless (Nle) is a conserved nuclear protein that is involved in a crucial step in ribosome biogenesis, and is required for the maintenance of adult haematopoietic and intestinal stem/progenitor cells. Here, we show that activated skeletal muscle satellite cells in conditional Nle mutant mice are arrested in proliferation; however, deletion of Nle in myofibres does not impair myogenesis. Furthermore, conditional deletion of Nle in satellite cells during homeostasis did not impact on their fate for up to 3 months. In contrast, loss of Nle function in primary myogenic cells blocked proliferation because of major defects in ribosome formation. Taken together, we show that muscle stem cells undergo a stage-specific regulation of ribosome biogenesis, thereby underscoring the importance of differential modulation of mRNA translation for controlling cell fate decisions.
Collapse
Affiliation(s)
- Barbara Gayraud-Morel
- Stem Cells and Development, Department of Developmental & Stem Cell Biology, Institut Pasteur, 25 rue du Dr Roux, 75015 Paris, France.,CNRS UMR 3738, Institut Pasteur, 25 rue du Dr Roux, 75015 Paris, France
| | - Marie Le Bouteiller
- CNRS UMR 3738, Institut Pasteur, 25 rue du Dr Roux, 75015 Paris, France.,Early Mammalian Development and Stem Cell Biology, Department of Developmental & Stem Cell Biology, Institut Pasteur, 25 rue du Dr Roux, 75015 Paris, France
| | - Pierre-Henri Commere
- Plateforme de Cytometrie, Institut Pasteur, 25 rue du Dr Roux, 75015 Paris, France
| | - Michel Cohen-Tannoudji
- CNRS UMR 3738, Institut Pasteur, 25 rue du Dr Roux, 75015 Paris, France.,Early Mammalian Development and Stem Cell Biology, Department of Developmental & Stem Cell Biology, Institut Pasteur, 25 rue du Dr Roux, 75015 Paris, France
| | - Shahragim Tajbakhsh
- Stem Cells and Development, Department of Developmental & Stem Cell Biology, Institut Pasteur, 25 rue du Dr Roux, 75015 Paris, France .,CNRS UMR 3738, Institut Pasteur, 25 rue du Dr Roux, 75015 Paris, France
| |
Collapse
|
18
|
Motohashi N, Uezumi A, Asakura A, Ikemoto-Uezumi M, Mori S, Mizunoe Y, Takashima R, Miyagoe-Suzuki Y, Takeda S, Shigemoto K. Tbx1 regulates inherited metabolic and myogenic abilities of progenitor cells derived from slow- and fast-type muscle. Cell Death Differ 2018; 26:1024-1036. [PMID: 30154444 DOI: 10.1038/s41418-018-0186-4] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2018] [Revised: 07/18/2018] [Accepted: 07/27/2018] [Indexed: 11/09/2022] Open
Abstract
Skeletal muscle is divided into slow- and fast-type muscles, which possess distinct contractile and metabolic properties. Myogenic progenitors associated with each muscle fiber type are known to intrinsically commit to specific muscle fiber lineage during embryonic development. However, it is still unclear whether the functionality of postnatal adult myogenic cells is attributable to the muscle fiber in which they reside, and whether the characteristics of myogenic cells derived from slow- and fast-type fibers can be distinguished at the genetic level. In this study, we isolated adult satellite cells from slow- and fast-type muscle individually and observed that satellite cells from each type of muscle generated myotubes expressing myosin heavy chain isoforms similar to their original muscle, and showed different metabolic features. Notably, we discovered that slow muscle-derived cells had low potential to differentiate but high potential to self-renew compared with fast muscle-derived cells. Additionally, cell transplantation experiments of slow muscle-derived cells into fast-type muscle revealed that slow muscle-derived cells could better contribute to myofiber formation and satellite cell constitution than fast muscle-derived cells, suggesting that the recipient muscle fiber type may not affect the predetermined abilities of myogenic cells. Gene expression analyses identified T-box transcriptional factor Tbx1 as a highly expressed gene in fast muscle-derived myoblasts. Gain- and loss-of-function experiments revealed that Tbx1 modulated muscle fiber types and oxidative metabolism in myotubes, and that Tbx1 stimulated myoblast differentiation, but did not regulate myogenic cell self-renewal. Our data suggest that metabolic and myogenic properties of myogenic progenitor cells vary depending on the type of muscle from which they originate, and that Tbx1 expression partially explains the functional differences of myogenic cells derived from fast-type and slow-type muscles.
Collapse
Affiliation(s)
- Norio Motohashi
- Department of Geriatric Medicine, Tokyo Metropolitan Institute of Gerontology, Itabashi, Tokyo, 173-0015, Japan. .,Stem Cell Institute, Paul and Sheila Wellstone Muscular Dystrophy Center, Department of Neurology, University of Minnesota Medical School, Minneapolis, MN, 55455, USA.
| | - Akiyoshi Uezumi
- Department of Geriatric Medicine, Tokyo Metropolitan Institute of Gerontology, Itabashi, Tokyo, 173-0015, Japan
| | - Atsushi Asakura
- Stem Cell Institute, Paul and Sheila Wellstone Muscular Dystrophy Center, Department of Neurology, University of Minnesota Medical School, Minneapolis, MN, 55455, USA
| | - Madoka Ikemoto-Uezumi
- Department of Geriatric Medicine, Tokyo Metropolitan Institute of Gerontology, Itabashi, Tokyo, 173-0015, Japan
| | - Shuuichi Mori
- Department of Geriatric Medicine, Tokyo Metropolitan Institute of Gerontology, Itabashi, Tokyo, 173-0015, Japan
| | - Yuhei Mizunoe
- Department of Geriatric Medicine, Tokyo Metropolitan Institute of Gerontology, Itabashi, Tokyo, 173-0015, Japan
| | - Rumi Takashima
- Department of Geriatric Medicine, Tokyo Metropolitan Institute of Gerontology, Itabashi, Tokyo, 173-0015, Japan
| | - Yuko Miyagoe-Suzuki
- Department of Molecular Therapy, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira, Tokyo, 187-8502, Japan
| | - Shin'ichi Takeda
- Department of Molecular Therapy, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira, Tokyo, 187-8502, Japan
| | - Kazuhiro Shigemoto
- Department of Geriatric Medicine, Tokyo Metropolitan Institute of Gerontology, Itabashi, Tokyo, 173-0015, Japan
| |
Collapse
|
19
|
Virgilio KM, Martin KS, Peirce SM, Blemker SS. Agent-based model illustrates the role of the microenvironment in regeneration in healthy and mdx skeletal muscle. J Appl Physiol (1985) 2018; 125:1424-1439. [PMID: 30070607 DOI: 10.1152/japplphysiol.00379.2018] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is a progressive muscle-wasting disease with no effective treatment. Multiple mechanisms are thought to contribute to muscle wasting, including increased susceptibility to contraction-induced damage, chronic inflammation, fibrosis, altered satellite stem cell (SSC) dynamics, and impaired regenerative capacity. The goals of this project were to 1) develop an agent-based model of skeletal muscle that predicts the dynamic regenerative response of muscle cells, fibroblasts, SSCs, and inflammatory cells as a result of contraction-induced injury, 2) calibrate and validate the model parameters based on comparisons with published experimental measurements, and 3) use the model to investigate how changing isolated and combined factors known to be associated with DMD (e.g., altered fibroblast or SSC behaviors) influence muscle regeneration. Our predictions revealed that the percent of injured muscle that recovered 28 days after injury was dependent on the peak SSC counts following injury. In simulations with near-full cross-sectional area recovery (healthy, 4-wk mdx, 3-mo mdx), the SSC counts correlated with the extent of initial injury; however, in simulations with impaired regeneration (9-mo mdx), the peak SSC counts were suppressed relative to initial injury. The differences in SSC counts between these groups were emergent predictions dependent on altered microenvironment factors known to be associated with DMD. Multiple cell types influenced the peak number of SSCs, but no individual parameter predicted the differences in SSC counts. This finding suggests that interventions to target the microenvironment rather than SSCs directly could be an effective method for improving regeneration in impaired muscle. NEW & NOTEWORTHY A computational model predicted that satellite stem cell (SSC) counts are correlated with muscle cross-sectional area (CSA) recovery following injury. In simulations with impaired CSA recovery, SSC counts are suppressed relative to healthy muscle. The suppressed SSC counts were an emergent model prediction, because all simulations had equal initial SSC counts. Fibroblast and anti-inflammatory macrophage counts influenced SSC counts, but no single factor was able to predict the pathological differences in SSC counts that lead to impaired regeneration.
Collapse
Affiliation(s)
- Kelley M Virgilio
- Department of Biomedical Engineering, University of Virginia , Charlottesville, Virginia
| | - Kyle S Martin
- Department of Biomedical Engineering, University of Virginia , Charlottesville, Virginia
| | - Shayn M Peirce
- Department of Biomedical Engineering, University of Virginia , Charlottesville, Virginia
| | - Silvia S Blemker
- Department of Biomedical Engineering, University of Virginia , Charlottesville, Virginia.,Department of Orthopaedic Surgery, University of Virginia , Charlottesville, Virginia.,Department of Mechanical and Aerospace Engineering, University of Virginia , Charlottesville, Virginia
| |
Collapse
|
20
|
Pala F, Di Girolamo D, Mella S, Yennek S, Chatre L, Ricchetti M, Tajbakhsh S. Distinct metabolic states govern skeletal muscle stem cell fates during prenatal and postnatal myogenesis. J Cell Sci 2018; 131:131/14/jcs212977. [PMID: 30054310 PMCID: PMC6080609 DOI: 10.1242/jcs.212977] [Citation(s) in RCA: 103] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Accepted: 04/18/2018] [Indexed: 12/19/2022] Open
Abstract
During growth, homeostasis and regeneration, stem cells are exposed to different energy demands. Here, we characterise the metabolic pathways that mediate the commitment and differentiation of mouse skeletal muscle stem cells, and how their modulation can influence the cell state. We show that quiescent satellite stem cells have low energetic demands and perturbed oxidative phosphorylation during ageing, which is also the case for cells from post-mortem tissues. We show also that myogenic fetal cells have distinct metabolic requirements compared to those proliferating during regeneration, with the former displaying a low respiration demand relying mostly on glycolysis. Furthermore, we show distinct requirements for peroxisomal and mitochondrial fatty acid oxidation (FAO) in myogenic cells. Compromising peroxisomal but not mitochondrial FAO promotes early differentiation of myogenic cells. Acute muscle injury and pharmacological block of peroxisomal and mitochondrial FAO expose differential requirements for these organelles during muscle regeneration. Taken together, these observations indicate that changes in myogenic cell state lead to significant alterations in metabolic requirements. In addition, perturbing specific metabolic pathways impacts on myogenic cell fates and the regeneration process. Summary: Distinct energy metabolism pathways act during mouse skeletal muscle stem cell commitment and differentiation in different physiological states.
Collapse
Affiliation(s)
- Francesca Pala
- Stem Cells and Development, Department of Developmental & Stem Cell Biology, Institut Pasteur, Paris 75015, France.,CNRS UMR 3738, Institut Pasteur, Paris 75015, France
| | - Daniela Di Girolamo
- Stem Cells and Development, Department of Developmental & Stem Cell Biology, Institut Pasteur, Paris 75015, France.,CNRS UMR 3738, Institut Pasteur, Paris 75015, France.,Dipartimento di Medicina Clinica e Chirurgia, Università degli Studi di Napoli "Federico II", Via S. Pansini 5, 80131 Napoli, Italy
| | - Sébastien Mella
- Stem Cells and Development, Department of Developmental & Stem Cell Biology, Institut Pasteur, Paris 75015, France.,CNRS UMR 3738, Institut Pasteur, Paris 75015, France
| | - Siham Yennek
- Stem Cells and Development, Department of Developmental & Stem Cell Biology, Institut Pasteur, Paris 75015, France.,CNRS UMR 3738, Institut Pasteur, Paris 75015, France
| | - Laurent Chatre
- CNRS UMR 3738, Institut Pasteur, Paris 75015, France.,Stem Cells and Development, Team Stability of Nuclear and Mitochondrial DNA, Department of Developmental & Stem Cell Biology, Institut Pasteur, Paris 75015, France
| | - Miria Ricchetti
- CNRS UMR 3738, Institut Pasteur, Paris 75015, France.,Stem Cells and Development, Team Stability of Nuclear and Mitochondrial DNA, Department of Developmental & Stem Cell Biology, Institut Pasteur, Paris 75015, France
| | - Shahragim Tajbakhsh
- Stem Cells and Development, Department of Developmental & Stem Cell Biology, Institut Pasteur, Paris 75015, France .,CNRS UMR 3738, Institut Pasteur, Paris 75015, France
| |
Collapse
|
21
|
Zhang P, Li W, Wang L, Liu H, Gong J, Wang F, Chen X. Salidroside Inhibits Myogenesis by Modulating p-Smad3-Induced Myf5 Transcription. Front Pharmacol 2018; 9:209. [PMID: 29593538 PMCID: PMC5858519 DOI: 10.3389/fphar.2018.00209] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Accepted: 02/23/2018] [Indexed: 01/11/2023] Open
Abstract
Aim: Salidroside is an active compound extracted from Rhodiola rosea which is used to alleviate fatigue and enhance endurance in high altitude regions. Some studies have demonstrated that salidroside can affect precursor cell differentiation in hematopoietic stem cells, erythrocytes, and osteoblasts. The aim of this study was to investigate the effect of salidroside on myoblast differentiation and to explore the underlying molecular mechanisms of this effect. Methods: C2C12 myoblast cells were treated with different concentrations of salidroside in differentiation media. Real-time PCR, Western blotting, and immunofluorescence assay were employed to evaluate the effects of salidroside on C2C12 differentiation. RNA interference was used to reveal the important role of Myf5 in myogenesis inhibited by salidroside. Chromatin Immunoprecipitation and dual-luciferase reporter assay were utilized to explore the underlying mechanisms of salidroside-induced upregulation of Myf5. Results: We found that salidroside inhibits myogenesis by downregulating MyoD and myogenin, preserves undifferentiated reserve cell pools by upregulating Myf5. Knocking down Myf5 expression significantly rescued the myogenesis inhibited by salidroside. The effect of salidroside on myogenesis was associated with increased phosphorylated Smad3 (p-Smad3). Both SIS3 (Specific inhibitor of p-Smad3) and dominant negative Smad3 plasmid (DN-Smad3) attenuated the inhibitory effect of salidroside on C2C12 differentiation. Moreover, the induction of Myf5 transcription by salidroside was dependent on a Smad-binding site in the promoter region of Myf5 gene. Conclusion and Implications: Our findings identify a novel role and mechanism for salidroside in regulating myogenesis through p-Smad3-induced Myf5 transcription, which may have implications for its further application in combating degenerative muscular diseases caused by depletion of muscle stem cells, such as Duchenne muscular dystrophy or sarcopenia.
Collapse
Affiliation(s)
- Peng Zhang
- State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, China
| | - Wenjiong Li
- State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, China
| | - Lu Wang
- National Key Laboratory of Human Factors Engineering, China Astronaut Research and Training Center, Beijing, China
| | - Hongju Liu
- State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, China
| | - Jing Gong
- National Key Laboratory of Human Factors Engineering, China Astronaut Research and Training Center, Beijing, China
| | - Fei Wang
- National Key Laboratory of Human Factors Engineering, China Astronaut Research and Training Center, Beijing, China
| | - Xiaoping Chen
- State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, China.,National Key Laboratory of Human Factors Engineering, China Astronaut Research and Training Center, Beijing, China
| |
Collapse
|
22
|
Chal J, Al Tanoury Z, Oginuma M, Moncuquet P, Gobert B, Miyanari A, Tassy O, Guevara G, Hubaud A, Bera A, Sumara O, Garnier JM, Kennedy L, Knockaert M, Gayraud-Morel B, Tajbakhsh S, Pourquié O. Recapitulating early development of mouse musculoskeletal precursors of the paraxial mesoderm in vitro. Development 2018; 145:145/6/dev157339. [DOI: 10.1242/dev.157339] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2017] [Accepted: 02/06/2018] [Indexed: 12/13/2022]
Abstract
ABSTRACT
Body skeletal muscles derive from the paraxial mesoderm, which forms in the posterior region of the embryo. Using microarrays, we characterize novel mouse presomitic mesoderm (PSM) markers and show that, unlike the abrupt transcriptome reorganization of the PSM, neural tube differentiation is accompanied by progressive transcriptome changes. The early paraxial mesoderm differentiation stages can be efficiently recapitulated in vitro using mouse and human pluripotent stem cells. While Wnt activation alone can induce posterior PSM markers, acquisition of a committed PSM fate and efficient differentiation into anterior PSM Pax3+ identity further requires BMP inhibition to prevent progenitors from drifting to a lateral plate mesoderm fate. When transplanted into injured adult muscle, these precursors generated large numbers of immature muscle fibers. Furthermore, exposing these mouse PSM-like cells to a brief FGF inhibition step followed by culture in horse serum-containing medium allows efficient recapitulation of the myogenic program to generate myotubes and associated Pax7+ cells. This protocol results in improved in vitro differentiation and maturation of mouse muscle fibers over serum-free protocols and enables the study of myogenic cell fusion and satellite cell differentiation.
Collapse
Affiliation(s)
- Jérome Chal
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), CNRS (UMR 7104), Inserm U964, Université de Strasbourg, Illkirch Graffenstaden 67400, France
- Department of Pathology, Brigham and Women's Hospital, Boston, MA 02115, USA
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
- Harvard Stem Cell Institute, Boston, MA 02115, USA
| | - Ziad Al Tanoury
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), CNRS (UMR 7104), Inserm U964, Université de Strasbourg, Illkirch Graffenstaden 67400, France
- Department of Pathology, Brigham and Women's Hospital, Boston, MA 02115, USA
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
- Harvard Stem Cell Institute, Boston, MA 02115, USA
| | - Masayuki Oginuma
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), CNRS (UMR 7104), Inserm U964, Université de Strasbourg, Illkirch Graffenstaden 67400, France
- Department of Pathology, Brigham and Women's Hospital, Boston, MA 02115, USA
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
- Harvard Stem Cell Institute, Boston, MA 02115, USA
| | - Philippe Moncuquet
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), CNRS (UMR 7104), Inserm U964, Université de Strasbourg, Illkirch Graffenstaden 67400, France
| | - Bénédicte Gobert
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), CNRS (UMR 7104), Inserm U964, Université de Strasbourg, Illkirch Graffenstaden 67400, France
- Anagenesis Biotechnologies, Parc d'innovation, Illkirch Graffenstaden 67400, France
| | - Ayako Miyanari
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), CNRS (UMR 7104), Inserm U964, Université de Strasbourg, Illkirch Graffenstaden 67400, France
| | - Olivier Tassy
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), CNRS (UMR 7104), Inserm U964, Université de Strasbourg, Illkirch Graffenstaden 67400, France
| | - Getzabel Guevara
- Department of Pathology, Brigham and Women's Hospital, Boston, MA 02115, USA
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
- Harvard Stem Cell Institute, Boston, MA 02115, USA
| | - Alexis Hubaud
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), CNRS (UMR 7104), Inserm U964, Université de Strasbourg, Illkirch Graffenstaden 67400, France
- Department of Pathology, Brigham and Women's Hospital, Boston, MA 02115, USA
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
- Harvard Stem Cell Institute, Boston, MA 02115, USA
| | - Agata Bera
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), CNRS (UMR 7104), Inserm U964, Université de Strasbourg, Illkirch Graffenstaden 67400, France
| | - Olga Sumara
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), CNRS (UMR 7104), Inserm U964, Université de Strasbourg, Illkirch Graffenstaden 67400, France
| | - Jean-Marie Garnier
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), CNRS (UMR 7104), Inserm U964, Université de Strasbourg, Illkirch Graffenstaden 67400, France
| | - Leif Kennedy
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), CNRS (UMR 7104), Inserm U964, Université de Strasbourg, Illkirch Graffenstaden 67400, France
| | - Marie Knockaert
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), CNRS (UMR 7104), Inserm U964, Université de Strasbourg, Illkirch Graffenstaden 67400, France
- Department of Pathology, Brigham and Women's Hospital, Boston, MA 02115, USA
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
- Harvard Stem Cell Institute, Boston, MA 02115, USA
| | - Barbara Gayraud-Morel
- Department of Developmental and Stem Cell Biology, Institut Pasteur, Paris 75015, France
- CNRS UMR 3738, Institut Pasteur, Paris 75015, France
| | - Shahragim Tajbakhsh
- Department of Developmental and Stem Cell Biology, Institut Pasteur, Paris 75015, France
- CNRS UMR 3738, Institut Pasteur, Paris 75015, France
| | - Olivier Pourquié
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), CNRS (UMR 7104), Inserm U964, Université de Strasbourg, Illkirch Graffenstaden 67400, France
- Department of Pathology, Brigham and Women's Hospital, Boston, MA 02115, USA
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
- Harvard Stem Cell Institute, Boston, MA 02115, USA
| |
Collapse
|
23
|
Abstract
Skeletal muscle satellite cells are quiescent adult resident stem cells that activate, proliferate and differentiate to generate myofibres following injury. They harbour a robust proliferation potential and self-renewing capacity enabling lifelong muscle regeneration. Although several classes of microRNAs were shown to regulate adult myogenesis, systematic examination of stage-specific microRNAs during lineage progression from the quiescent state is lacking. Here we provide a genome-wide assessment of the expression of small RNAs during the quiescence/activation transition and differentiation by RNA-sequencing. We show that the majority of small RNAs present in quiescent, activated and differentiated muscle cells belong to the microRNA class. Furthermore, by comparing expression in distinct cell states, we report a massive and dynamic regulation of microRNAs, both in numbers and amplitude, highlighting their pivotal role in regulation of quiescence, activation and differentiation. We also identify a number of microRNAs with reliable and specific expression in quiescence including several maternally-expressed miRNAs generated at the imprinted Dlk1-Dio3 locus. Unexpectedly, the majority of class-switching miRNAs are associated with the quiescence/activation transition suggesting a poised program that is actively repressed. These data constitute a key resource for functional analyses of miRNAs in skeletal myogenesis, and more broadly, in the regulation of stem cell self-renewal and tissue homeostasis.
Collapse
|
24
|
Perandini LA, Chimin P, Lutkemeyer DDS, Câmara NOS. Chronic inflammation in skeletal muscle impairs satellite cells function during regeneration: can physical exercise restore the satellite cell niche? FEBS J 2018; 285:1973-1984. [PMID: 29473995 DOI: 10.1111/febs.14417] [Citation(s) in RCA: 96] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2017] [Revised: 01/27/2018] [Accepted: 02/19/2018] [Indexed: 12/23/2022]
Abstract
Chronic inflammation impairs skeletal muscle regeneration. Although many cells are involved in chronic inflammation, macrophages seem to play an important role in impaired muscle regeneration since these cells are associated with skeletal muscle stem cell (namely, satellite cells) activation and fibro-adipogenic progenitor cell (FAP) survival. Specifically, an imbalance of M1 and M2 macrophages seems to lead to impaired satellite cell activation, and these are the main cells that function during skeletal muscle regeneration, after muscle damage. Additionally, this imbalance leads to the accumulation of FAPs in skeletal muscle, with aberrant production of pro-fibrotic factors (e.g., extracellular matrix components), impairing the niche for proper satellite cell activation and differentiation. Treatments aiming to block the inflammatory pro-fibrotic response are partially effective due to their side effects. Therefore, strategies reverting chronic inflammation into a pro-regenerative pattern are required. In this review, we first describe skeletal muscle resident macrophage ontogeny and homeostasis, and explain how macrophages are replenished after muscle injury. We next discuss the potential role of chronic physical activity and exercise in restoring the M1 and M2 macrophage balance and consequently, the satellite cell niche to improve skeletal muscle regeneration after injury.
Collapse
Affiliation(s)
- Luiz Augusto Perandini
- Laboratory of Transplantation Immunobiology, Department of Immunology, Institute of Biomedical Sciences, University of Sao Paulo, Brazil
| | - Patricia Chimin
- Department of Physical Education, Physical Education and Sports Center, Londrina State University, Brazil
| | - Diego da Silva Lutkemeyer
- Laboratory of Transplantation Immunobiology, Department of Immunology, Institute of Biomedical Sciences, University of Sao Paulo, Brazil
| | - Niels Olsen Saraiva Câmara
- Laboratory of Transplantation Immunobiology, Department of Immunology, Institute of Biomedical Sciences, University of Sao Paulo, Brazil.,Laboratory of Clinical and Experimental Immunology, Division of Nephrology, Department of Medicine, Federal University of Sao Paulo, Brazil
| |
Collapse
|
25
|
Yamamoto M, Legendre NP, Biswas AA, Lawton A, Yamamoto S, Tajbakhsh S, Kardon G, Goldhamer DJ. Loss of MyoD and Myf5 in Skeletal Muscle Stem Cells Results in Altered Myogenic Programming and Failed Regeneration. Stem Cell Reports 2018; 10:956-969. [PMID: 29478898 PMCID: PMC5918368 DOI: 10.1016/j.stemcr.2018.01.027] [Citation(s) in RCA: 103] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Revised: 01/22/2018] [Accepted: 01/23/2018] [Indexed: 11/22/2022] Open
Abstract
MyoD and Myf5 are fundamental regulators of skeletal muscle lineage determination in the embryo, and their expression is induced in satellite cells following muscle injury. MyoD and Myf5 are also expressed by satellite cell precursors developmentally, although the relative contribution of historical and injury-induced expression to satellite cell function is unknown. We show that satellite cells lacking both MyoD and Myf5 (double knockout [dKO]) are maintained with aging in uninjured muscle. However, injured muscle fails to regenerate and dKO satellite cell progeny accumulate in damaged muscle but do not undergo muscle differentiation. dKO satellite cell progeny continue to express markers of myoblast identity, although their myogenic programming is labile, as demonstrated by dramatic morphological changes and increased propensity for non-myogenic differentiation. These data demonstrate an absolute requirement for either MyoD or Myf5 in muscle regeneration and indicate that their expression after injury stabilizes myogenic identity and confers the capacity for muscle differentiation. MyoD or Myf5 expression in satellite cells is essential for muscle regeneration Satellite cells lacking both regulatory genes exhibit labile myogenic programming A single functional allele of either MyoD or Myf5 can support muscle regeneration Satellite cells lacking both MyoD and Myf5 are maintained with aging
Collapse
Affiliation(s)
- Masakazu Yamamoto
- Department of Molecular & Cell Biology, University of Connecticut Stem Cell Institute, University of Connecticut, 91 N. Eagleville Road, Storrs, CT 06269, USA
| | - Nicholas P Legendre
- Department of Molecular & Cell Biology, University of Connecticut Stem Cell Institute, University of Connecticut, 91 N. Eagleville Road, Storrs, CT 06269, USA
| | - Arpita A Biswas
- Department of Molecular & Cell Biology, University of Connecticut Stem Cell Institute, University of Connecticut, 91 N. Eagleville Road, Storrs, CT 06269, USA
| | - Alexander Lawton
- Department of Molecular & Cell Biology, University of Connecticut Stem Cell Institute, University of Connecticut, 91 N. Eagleville Road, Storrs, CT 06269, USA
| | - Shoko Yamamoto
- Department of Molecular & Cell Biology, University of Connecticut Stem Cell Institute, University of Connecticut, 91 N. Eagleville Road, Storrs, CT 06269, USA
| | - Shahragim Tajbakhsh
- Institut Pasteur, Stem Cells & Development, CNRS URA 2578, 25 rue du Dr. Roux, 75724 Paris Cedex 15, France
| | - Gabrielle Kardon
- Department of Human Genetics, University of Utah, 15 North 2030 East, Salt Lake City, UT 84112, USA
| | - David J Goldhamer
- Department of Molecular & Cell Biology, University of Connecticut Stem Cell Institute, University of Connecticut, 91 N. Eagleville Road, Storrs, CT 06269, USA.
| |
Collapse
|
26
|
Asfour HA, Allouh MZ, Said RS. Myogenic regulatory factors: The orchestrators of myogenesis after 30 years of discovery. Exp Biol Med (Maywood) 2018; 243:118-128. [PMID: 29307280 DOI: 10.1177/1535370217749494] [Citation(s) in RCA: 153] [Impact Index Per Article: 21.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Prenatal and postnatal myogenesis share many cellular and molecular aspects. Myogenic regulatory factors are basic Helix-Loop-Helix transcription factors that indispensably regulate both processes. These factors (Myf5, MyoD, Myogenin, and MRF4) function as an orchestrating cascade, with some overlapped actions. Prenatally, myogenic regulatory factors are restrictedly expressed in somite-derived myogenic progenitor cells and their derived myoblasts. Postnatally, myogenic regulatory factors are important in regulating the myogenesis process via satellite cells. Many positive and negative regulatory mechanisms exist either between myogenic regulatory factors themselves or between myogenic regulatory factors and other proteins. Upstream factors and signals are also involved in the control of myogenic regulatory factors expression within different prenatal and postnatal myogenic cells. Here, the authors have conducted a thorough and an up-to-date review of the myogenic regulatory factors since their discovery 30 years ago. This review discusses the myogenic regulatory factors structure, mechanism of action, and roles and regulations during prenatal and postnatal myogenesis. Impact statement Myogenic regulatory factors (MRFs) are key players in the process of myogenesis. Despite a considerable amount of literature regarding these factors, their exact mechanisms of actions are still incompletely understood with several overlapped functions. Herein, we revised what has hitherto been reported in the literature regarding MRF structures, molecular pathways that regulate their activities, and their roles during pre- and post-natal myogenesis. The work submitted in this review article is considered of great importance for researchers in the field of skeletal muscle formation and regeneration, as it provides a comprehensive summary of all the biological aspects of MRFs and advances a better understanding of the cellular and molecular mechanisms regulating myogenesis. Indeed, attaining a better understanding of MRFs could be utilized in developing novel therapeutic protocols for multiple myopathies.
Collapse
Affiliation(s)
- Hasan A Asfour
- Department of Anatomy, Faculty of Medicine, 37251 Jordan University of Science & Technology , Irbid 22110, Jordan
| | - Mohammed Z Allouh
- Department of Anatomy, Faculty of Medicine, 37251 Jordan University of Science & Technology , Irbid 22110, Jordan
| | - Raed S Said
- Department of Anatomy, Faculty of Medicine, 37251 Jordan University of Science & Technology , Irbid 22110, Jordan
| |
Collapse
|
27
|
Abstract
Our understanding of satellite cells, now known to be the obligate stem cells of skeletal muscle, has increased dramatically in recent years due to the introduction of new molecular, genetic, and technical resources. In addition to their role in acute repair of damaged muscle, satellite cells are of interest in the fields of aging, exercise, neuromuscular disease, and stem cell therapy, and all of these applications have driven a dramatic increase in our understanding of the activity and potential of satellite cells. However, many fundamental questions of satellite cell biology remain to be answered, including their emergence as a specific lineage, the degree and significance of heterogeneity within the satellite cell population, the roles of their interactions with other resident and infiltrating cell types during homeostasis and regeneration, and the relative roles of intrinsic vs extrinsic factors that may contribute to satellite cell dysfunction in the context of aging or disease. This review will address the current state of these open questions in satellite cell biology.
Collapse
Affiliation(s)
- Ddw Cornelison
- University of Missouri, Columbia, MO, United States; Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, MO, United States.
| |
Collapse
|
28
|
Zammit PS. Function of the myogenic regulatory factors Myf5, MyoD, Myogenin and MRF4 in skeletal muscle, satellite cells and regenerative myogenesis. Semin Cell Dev Biol 2017; 72:19-32. [PMID: 29127046 DOI: 10.1016/j.semcdb.2017.11.011] [Citation(s) in RCA: 508] [Impact Index Per Article: 63.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2017] [Revised: 11/03/2017] [Accepted: 11/06/2017] [Indexed: 12/19/2022]
Abstract
Discovery of the myogenic regulatory factor family of transcription factors MYF5, MYOD, Myogenin and MRF4 was a seminal step in understanding specification of the skeletal muscle lineage and control of myogenic differentiation during development. These factors are also involved in specification of the muscle satellite cell lineage, which becomes the resident stem cell compartment inadult skeletal muscle. While MYF5, MYOD, Myogenin and MRF4 have subtle roles in mature muscle, they again play a crucial role in directing satellite cell function to regenerate skeletal muscle: linking the genetic control of developmental and regenerative myogenesis. Here, I review the role of the myogenic regulatory factors in developing and mature skeletal muscle, satellite cell specification and muscle regeneration.
Collapse
Affiliation(s)
- Peter S Zammit
- King's College London, Randall Centre for Cell and Molecular Biophysics, London, SE1 1UL, UK.
| |
Collapse
|
29
|
Agley CC, Lewis FC, Jaka O, Lazarus NR, Velloso C, Francis-West P, Ellison-Hughes GM, Harridge SDR. Active GSK3β and an intact β-catenin TCF complex are essential for the differentiation of human myogenic progenitor cells. Sci Rep 2017; 7:13189. [PMID: 29030569 PMCID: PMC5640663 DOI: 10.1038/s41598-017-10731-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Accepted: 08/14/2017] [Indexed: 01/14/2023] Open
Abstract
Wnt-β-catenin signalling is essential for skeletal muscle myogenesis during development, but its role in adult human skeletal muscle remains unknown. Here we have used human primary CD56Pos satellite cell-derived myogenic progenitors obtained from healthy individuals to study the role of Wnt-β-catenin signalling in myogenic differentiation. We show that dephosphorylated β-catenin (active-β-catenin), the central effector of the canonical Wnt cascade, is strongly upregulated at the onset of differentiation and undergoes nuclear translocation as differentiation progresses. To establish the role of Wnt signalling in regulating the differentiation process we manipulated key nodes of this pathway through a series of β-catenin gain-of-function (GSK3 inhibition and β-catenin overexpression) or loss-of-function experiments (dominant negative TCF4). Our data showed that manipulation of these critical pathway components led to varying degrees of disruption to the normal differentiation phenotype indicating the importance of Wnt signalling in regulating this process. We reveal an independent necessity for active-β-catenin in the fusion and differentiation of human myogenic progenitors and that dominant negative inhibition of TCF4 prevents differentiation completely. Together these data add new mechanistic insights into both Wnt signalling and adult human myogenic progenitor differentiation.
Collapse
Affiliation(s)
- C C Agley
- Centre of Human and Aerospace Physiological Sciences, King's College London, London, UK. .,Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK.
| | - F C Lewis
- Centre of Human and Aerospace Physiological Sciences, King's College London, London, UK.,Stem Cell Institute, King's College London, London, UK
| | - O Jaka
- Centre of Human and Aerospace Physiological Sciences, King's College London, London, UK
| | - N R Lazarus
- Centre of Human and Aerospace Physiological Sciences, King's College London, London, UK
| | - C Velloso
- Centre of Human and Aerospace Physiological Sciences, King's College London, London, UK
| | - P Francis-West
- Department of Craniofacial development and stem cell biology, King's College London, London, UK
| | - G M Ellison-Hughes
- Centre of Human and Aerospace Physiological Sciences, King's College London, London, UK.,Stem Cell Institute, King's College London, London, UK
| | - S D R Harridge
- Centre of Human and Aerospace Physiological Sciences, King's College London, London, UK.,Stem Cell Institute, King's College London, London, UK
| |
Collapse
|
30
|
A novel paradigm links mitochondrial dysfunction with muscle stem cell impairment in sepsis. Biochim Biophys Acta Mol Basis Dis 2017; 1863:2546-2553. [PMID: 28456665 DOI: 10.1016/j.bbadis.2017.04.019] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Revised: 04/13/2017] [Accepted: 04/18/2017] [Indexed: 02/06/2023]
Abstract
Sepsis is an acute systemic inflammatory response of the body to microbial infection and a life threatening condition associated with multiple organ failure. Survivors may display long-term disability with muscle weakness that remains poorly understood. Recent data suggest that long-term myopathy in sepsis survivors is due to failure of skeletal muscle stem cells (satellite cells) to regenerate the muscle. Satellite cells impairment in the acute phase of sepsis is linked to unusual mitochondrial dysfunctions, characterized by a dramatic reduction of the mitochondrial mass and hyperactivity of residual organelles. Survivors maintain the impairment of satellite cells, including alterations of the mitochondrial DNA (mtDNA), in the long-term. This condition can be rescued by treatment with mesenchymal stem cells (MSCs) that restore mtDNA alterations and mitochondrial function in satellite cells, and in fine their regenerative potential. Injection of MSCs in turn increases the force of isolated muscle fibers and of the whole animal, and improves the survival rate. These effects occur in the context of reduced inflammation markers that also raised during sepsis. Targeting muscle stem cells mitochondria, in a context of reduced inflammation, may represent a valuable strategy to reduce morbidity and long-term impairment of the muscle upon sepsis.
Collapse
|
31
|
Daughters RS, Keirstead SA, Slack JMW. Transformation of jaw muscle satellite cells to cardiomyocytes. Differentiation 2017; 93:58-65. [PMID: 27918914 PMCID: PMC5285469 DOI: 10.1016/j.diff.2016.11.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Accepted: 11/16/2016] [Indexed: 02/06/2023]
Abstract
In the embryo a population of progenitor cells known as the second heart field forms not just parts of the heart but also the jaw muscles of the head. Here we show that it is possible to take skeletal muscle satellite cells from jaw muscles of the adult mouse and to direct their differentiation to become heart muscle cells (cardiomyocytes). This is done by exposing the cells to extracellular factors similar to those which heart progenitors would experience during normal embryonic development. By contrast, cardiac differentiation does not occur at all from satellite cells isolated from trunk and limb muscles, which originate from the somites of the embryo. The cardiomyocytes arising from jaw muscle satellite cells express a range of specific marker proteins, beat spontaneously, display long action potentials with appropriate responses to nifedipine, norepinephrine and carbachol, and show synchronized calcium transients. Our results show the existence of a persistent cardiac developmental competence in satellite cells of the adult jaw muscles, associated with their origin from the second heart field of the embryo, and suggest a possible method of obtaining cardiomyocytes from individual patients without the need for a heart biopsy.
Collapse
Affiliation(s)
- Randall S Daughters
- Stem Cell Institute, University of Minnesota, MTRF, 2001 6th Street SE, Minneapolis, MN 55455, USA
| | - Susan A Keirstead
- Stem Cell Institute, University of Minnesota, MTRF, 2001 6th Street SE, Minneapolis, MN 55455, USA
| | - Jonathan M W Slack
- Stem Cell Institute, University of Minnesota, MTRF, 2001 6th Street SE, Minneapolis, MN 55455, USA.
| |
Collapse
|
32
|
Abstract
Skeletal muscle stem cells, originally termed satellite cells for their position adjacent to differentiated muscle fibers, are absolutely required for the process of skeletal muscle repair and regeneration. In the last decade, satellite cells have become one of the most studied adult stem cell systems and have emerged as a standard model not only in the field of stem cell-driven tissue regeneration but also in stem cell dysfunction and aging. Here, we provide background in the field and discuss recent advances in our understanding of muscle stem cell function and dysfunction, particularly in the case of aging, and the potential involvement of muscle stem cells in genetic diseases such as the muscular dystrophies.
Collapse
Affiliation(s)
- Ddw Cornelison
- Division of Biological Sciences and Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, MO, USA.
| | - Eusebio Perdiguero
- Cell Biology Group, Department of Experimental and Health Sciences (DCEXS), Pompeu Fabra University (UPF), CIBER on Neurodegenerative Diseases (CIBERNED), 08003, Barcelona, Spain.
| |
Collapse
|
33
|
Gayraud-Morel B, Pala F, Sakai H, Tajbakhsh S. Isolation of Muscle Stem Cells from Mouse Skeletal Muscle. Methods Mol Biol 2017; 1556:23-39. [PMID: 28247343 DOI: 10.1007/978-1-4939-6771-1_2] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Isolation of muscle stem cells from skeletal muscle is a critical step for the study of skeletal myogenesis and regeneration. Although stem cell isolation has been performed for decades, the emergence of flow cytometry with defined cell surface markers, or transgenic mouse models, has allowed the efficient isolation of highly enriched stem cell populations. Here, we describe the isolation of mouse muscle stem cells using two different combinations of enzyme treatments allowing the release of mononucleated muscle stem cells from their niche. Mouse muscle stem cells can be further isolated as a highly enriched population by flow cytometry using fluorescent reporters or cell surface markers. We will present advantages and drawbacks of these different approaches.
Collapse
Affiliation(s)
- Barbara Gayraud-Morel
- Department of Developmental and Stem Cell Biology, Stem Cells and Development, CNRS URA 2578, Institut Pasteur, 25 rue du Dr. Roux, Paris, 75015, France.
| | - Francesca Pala
- Department of Developmental and Stem Cell Biology, Stem Cells and Development, CNRS URA 2578, Institut Pasteur, 25 rue du Dr. Roux, Paris, 75015, France
| | - Hiroshi Sakai
- Department of Developmental and Stem Cell Biology, Stem Cells and Development, CNRS URA 2578, Institut Pasteur, 25 rue du Dr. Roux, Paris, 75015, France
| | - Shahragim Tajbakhsh
- Department of Developmental and Stem Cell Biology, Stem Cells and Development, CNRS URA 2578, Institut Pasteur, 25 rue du Dr. Roux, Paris, 75015, France
| |
Collapse
|
34
|
Naldaiz-Gastesi N, Goicoechea M, Alonso-Martín S, Aiastui A, López-Mayorga M, García-Belda P, Lacalle J, San José C, Araúzo-Bravo MJ, Trouilh L, Anton-Leberre V, Herrero D, Matheu A, Bernad A, García-Verdugo JM, Carvajal JJ, Relaix F, Lopez de Munain A, García-Parra P, Izeta A. Identification and Characterization of the Dermal Panniculus Carnosus Muscle Stem Cells. Stem Cell Reports 2016; 7:411-424. [PMID: 27594590 PMCID: PMC5032673 DOI: 10.1016/j.stemcr.2016.08.002] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2015] [Revised: 08/01/2016] [Accepted: 08/01/2016] [Indexed: 01/05/2023] Open
Abstract
The dermal Panniculus carnosus (PC) muscle is important for wound contraction in lower mammals and represents an interesting model of muscle regeneration due to its high cell turnover. The resident satellite cells (the bona fide muscle stem cells) remain poorly characterized. Here we analyzed PC satellite cells with regard to developmental origin and purported function. Lineage tracing shows that they originate in Myf5+, Pax3/Pax7+ cell populations. Skin and muscle wounding increased PC myofiber turnover, with the satellite cell progeny being involved in muscle regeneration but with no detectable contribution to the wound-bed myofibroblasts. Since hematopoietic stem cells fuse to PC myofibers in the absence of injury, we also studied the contribution of bone marrow-derived cells to the PC satellite cell compartment, demonstrating that cells of donor origin are capable of repopulating the PC muscle stem cell niche after irradiation and bone marrow transplantation but may not fully acquire the relevant myogenic commitment. PC satellite cells originate from Myf5+, Pax3/Pax7+ cell lineages Skin and muscle wounding increase PC myofiber turnover Donor bone marrow cells repopulate the PC satellite niche after BMT Dermis-derived myogenesis originates from the PC satellite cell population
Collapse
Affiliation(s)
- Neia Naldaiz-Gastesi
- Tissue Engineering Laboratory, Bioengineering Area, Instituto Biodonostia, San Sebastián 20014, Spain; Neuroscience Area, Instituto Biodonostia, San Sebastián 20014, Spain; CIBERNED, Instituto de Salud Carlos III, Madrid 28029, Spain
| | - María Goicoechea
- Neuroscience Area, Instituto Biodonostia, San Sebastián 20014, Spain; CIBERNED, Instituto de Salud Carlos III, Madrid 28029, Spain
| | - Sonia Alonso-Martín
- INSERM U955-E10, Université Paris Est, Faculté de Médicine, IMRB U955-E10, Creteil 94000, France
| | - Ana Aiastui
- Neuroscience Area, Instituto Biodonostia, San Sebastián 20014, Spain; CIBERNED, Instituto de Salud Carlos III, Madrid 28029, Spain
| | - Macarena López-Mayorga
- Molecular Embryology Team, Centro Andaluz de Biología del Desarrollo, Sevilla 41013, Spain
| | - Paula García-Belda
- CIBERNED, Instituto de Salud Carlos III, Madrid 28029, Spain; Laboratorio de Neurobiología Comparada, Instituto Cavanilles, Universidad de Valencia, Valencia 46980, Spain
| | - Jaione Lacalle
- Tissue Engineering Laboratory, Bioengineering Area, Instituto Biodonostia, San Sebastián 20014, Spain; Neuroscience Area, Instituto Biodonostia, San Sebastián 20014, Spain; Faculty of Medicine and Nursing, UPV-EHU, San Sebastián 20014, Spain
| | - Carlos San José
- Animal Facility and Experimental Surgery, Instituto Biodonostia, San Sebastián 20014, Spain
| | - Marcos J Araúzo-Bravo
- Computational Biology and Systems Biomedicine, Instituto Biodonostia, San Sebastián 20014, Spain; IKERBASQUE, Basque Foundation for Science, Bilbao 48013, Spain
| | - Lidwine Trouilh
- INSA, UPS, INP, LISBP, Université de Toulouse, 31077 Toulouse, France; INRA, UMR792, Ingénierie des Systèmes Biologiques et des Procédés, 31400 Toulouse, France; CNRS, UMR5504, 31400 Toulouse, France
| | - Véronique Anton-Leberre
- INSA, UPS, INP, LISBP, Université de Toulouse, 31077 Toulouse, France; INRA, UMR792, Ingénierie des Systèmes Biologiques et des Procédés, 31400 Toulouse, France; CNRS, UMR5504, 31400 Toulouse, France
| | - Diego Herrero
- Immunology and Oncology Department, Spanish National Center for Biotechnology (CNB-CSIC), Madrid 28049, Spain
| | - Ander Matheu
- IKERBASQUE, Basque Foundation for Science, Bilbao 48013, Spain; Cellular Oncology Group, Oncology Area, Instituto Biodonostia, San Sebastián 20014, Spain
| | - Antonio Bernad
- Immunology and Oncology Department, Spanish National Center for Biotechnology (CNB-CSIC), Madrid 28049, Spain
| | - José Manuel García-Verdugo
- CIBERNED, Instituto de Salud Carlos III, Madrid 28029, Spain; Laboratorio de Neurobiología Comparada, Instituto Cavanilles, Universidad de Valencia, Valencia 46980, Spain
| | - Jaime J Carvajal
- Molecular Embryology Team, Centro Andaluz de Biología del Desarrollo, Sevilla 41013, Spain
| | - Frédéric Relaix
- INSERM U955-E10, Université Paris Est, Faculté de Médicine, IMRB U955-E10, Creteil 94000, France
| | - Adolfo Lopez de Munain
- Neuroscience Area, Instituto Biodonostia, San Sebastián 20014, Spain; CIBERNED, Instituto de Salud Carlos III, Madrid 28029, Spain; Faculty of Medicine and Nursing, Department of Neurosciences, UPV-EHU, San Sebastián 20014, Spain; Department of Neurology, Hospital Universitario Donostia, San Sebastián 20014, Spain
| | - Patricia García-Parra
- Tissue Engineering Laboratory, Bioengineering Area, Instituto Biodonostia, San Sebastián 20014, Spain; Neuroscience Area, Instituto Biodonostia, San Sebastián 20014, Spain; CIBERNED, Instituto de Salud Carlos III, Madrid 28029, Spain.
| | - Ander Izeta
- Tissue Engineering Laboratory, Bioengineering Area, Instituto Biodonostia, San Sebastián 20014, Spain; Department of Biomedical Engineering, School of Engineering, Tecnun-University of Navarra, San Sebastián 20009, Spain.
| |
Collapse
|
35
|
Marino S, Di Foggia V. Invited Review: Polycomb group genes in the regeneration of the healthy and pathological skeletal muscle. Neuropathol Appl Neurobiol 2015; 42:407-22. [PMID: 26479276 DOI: 10.1111/nan.12290] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2015] [Revised: 10/14/2015] [Accepted: 10/19/2015] [Indexed: 12/21/2022]
Abstract
The polycomb group (PcG) proteins are epigenetic repressors required during key developmental processes, such as maintenance of cell identity and stem cell differentiation. To exert their repressive function, PcG proteins assemble on chromatin into multiprotein complexes, known as polycomb repressive complex 1 and 2. In this review, we will focus on the role and mode of function of PcG proteins in the development and regeneration of the skeletal muscle, both in normal and pathological conditions and we will discuss the emerging concept of modulation of their expression to enhance the muscle-specific regenerative process for patient benefit.
Collapse
Affiliation(s)
- S Marino
- Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - V Di Foggia
- Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| |
Collapse
|
36
|
Thorley M, Malatras A, Duddy W, Le Gall L, Mouly V, Butler Browne G, Duguez S. Changes in Communication between Muscle Stem Cells and their Environment with Aging. J Neuromuscul Dis 2015; 2:205-217. [PMID: 27858742 PMCID: PMC5240546 DOI: 10.3233/jnd-150097] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Aging is associated with both muscle weakness and a loss of muscle mass, contributing towards overall frailty in the elderly. Aging skeletal muscle is also characterised by a decreasing efficiency in repair and regeneration, together with a decline in the number of adult stem cells. Commensurate with this are general changes in whole body endocrine signalling, in local muscle secretory environment, as well as in intrinsic properties of the stem cells themselves. The present review discusses the various mechanisms that may be implicated in these age-associated changes, focusing on aspects of cell-cell communication and long-distance signalling factors, such as levels of circulating growth hormone, IL-6, IGF1, sex hormones, and inflammatory cytokines. Changes in the local environment are also discussed, implicating IL-6, IL-4, FGF-2, as well as other myokines, and processes that lead to thickening of the extra-cellular matrix. These factors, involved primarily in communication, can also modulate the intrinsic properties of muscle stem cells, including reduced DNA accessibility and repression of specific genes by methylation. Finally we discuss the decrease in the stem cell pool, particularly the failure of elderly myoblasts to re-quiesce after activation, and the consequences of all these changes on general muscle homeostasis.
Collapse
Affiliation(s)
- Matthew Thorley
- Sorbonne Universités, UPMC Univ Paris 06, Center of Research in Myology UMRS 974, F-75013, Paris, France.,INSERM UMRS 974, F-75013, Paris, France.,CNRS FRE 3617, F-75013, Paris, France.,Institut de Myologie, F-75013, Paris, France
| | - Apostolos Malatras
- Sorbonne Universités, UPMC Univ Paris 06, Center of Research in Myology UMRS 974, F-75013, Paris, France.,INSERM UMRS 974, F-75013, Paris, France.,CNRS FRE 3617, F-75013, Paris, France.,Institut de Myologie, F-75013, Paris, France
| | - William Duddy
- Sorbonne Universités, UPMC Univ Paris 06, Center of Research in Myology UMRS 974, F-75013, Paris, France.,INSERM UMRS 974, F-75013, Paris, France.,CNRS FRE 3617, F-75013, Paris, France.,Institut de Myologie, F-75013, Paris, France
| | - Laura Le Gall
- Sorbonne Universités, UPMC Univ Paris 06, Center of Research in Myology UMRS 974, F-75013, Paris, France.,INSERM UMRS 974, F-75013, Paris, France.,CNRS FRE 3617, F-75013, Paris, France.,Institut de Myologie, F-75013, Paris, France
| | - Vincent Mouly
- Sorbonne Universités, UPMC Univ Paris 06, Center of Research in Myology UMRS 974, F-75013, Paris, France.,INSERM UMRS 974, F-75013, Paris, France.,CNRS FRE 3617, F-75013, Paris, France.,Institut de Myologie, F-75013, Paris, France
| | - Gillian Butler Browne
- Sorbonne Universités, UPMC Univ Paris 06, Center of Research in Myology UMRS 974, F-75013, Paris, France.,CNRS FRE 3617, F-75013, Paris, France.,INSERM UMRS 974, F-75013, Paris, France.,CNRS FRE 3617, F-75013, Paris, France.,Institut de Myologie, F-75013, Paris, France
| | - Stéphanie Duguez
- Sorbonne Universités, UPMC Univ Paris 06, Center of Research in Myology UMRS 974, F-75013, Paris, France.,INSERM UMRS 974, F-75013, Paris, France.,CNRS FRE 3617, F-75013, Paris, France.,Institut de Myologie, F-75013, Paris, France
| |
Collapse
|
37
|
Lozano-Velasco E, Vallejo D, Esteban FJ, Doherty C, Hernández-Torres F, Franco D, Aránega AE. A Pitx2-MicroRNA Pathway Modulates Cell Proliferation in Myoblasts and Skeletal-Muscle Satellite Cells and Promotes Their Commitment to a Myogenic Cell Fate. Mol Cell Biol 2015; 35:2892-909. [PMID: 26055324 PMCID: PMC4525317 DOI: 10.1128/mcb.00536-15] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2015] [Revised: 05/28/2015] [Accepted: 05/29/2015] [Indexed: 01/21/2023] Open
Abstract
The acquisition of a proliferating-cell status from a quiescent state as well as the shift between proliferation and differentiation are key developmental steps in skeletal-muscle stem cells (satellite cells) to provide proper muscle regeneration. However, how satellite cell proliferation is regulated is not fully understood. Here, we report that the c-isoform of the transcription factor Pitx2 increases cell proliferation in myoblasts by downregulating microRNA 15b (miR-15b), miR-23b, miR-106b, and miR-503. This Pitx2c-microRNA (miRNA) pathway also regulates cell proliferation in early-activated satellite cells, enhancing Myf5(+) satellite cells and thereby promoting their commitment to a myogenic cell fate. This study reveals unknown functions of several miRNAs in myoblast and satellite cell behavior and thus may have future applications in regenerative medicine.
Collapse
Affiliation(s)
- Estefanía Lozano-Velasco
- Cardiac and Skeletal Myogenesis Group, Department of Experimental Biology, University of Jaén, Jaén, Spain
| | - Daniel Vallejo
- Cardiac and Skeletal Myogenesis Group, Department of Experimental Biology, University of Jaén, Jaén, Spain
| | - Francisco J Esteban
- Cardiac and Skeletal Myogenesis Group, Department of Experimental Biology, University of Jaén, Jaén, Spain
| | - Chris Doherty
- Cardiac and Skeletal Myogenesis Group, Department of Experimental Biology, University of Jaén, Jaén, Spain
| | - Francisco Hernández-Torres
- Cardiac and Skeletal Myogenesis Group, Department of Experimental Biology, University of Jaén, Jaén, Spain
| | - Diego Franco
- Cardiac and Skeletal Myogenesis Group, Department of Experimental Biology, University of Jaén, Jaén, Spain
| | - Amelia Eva Aránega
- Cardiac and Skeletal Myogenesis Group, Department of Experimental Biology, University of Jaén, Jaén, Spain
| |
Collapse
|
38
|
Dumont NA, Wang YX, Rudnicki MA. Intrinsic and extrinsic mechanisms regulating satellite cell function. Development 2015; 142:1572-81. [PMID: 25922523 PMCID: PMC4419274 DOI: 10.1242/dev.114223] [Citation(s) in RCA: 340] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Muscle stem cells, termed satellite cells, are crucial for skeletal muscle growth and regeneration. In healthy adult muscle, satellite cells are quiescent but poised for activation. During muscle regeneration, activated satellite cells transiently re-enter the cell cycle to proliferate and subsequently exit the cell cycle to differentiate or self-renew. Recent studies have demonstrated that satellite cells are heterogeneous and that subpopulations of satellite stem cells are able to perform asymmetric divisions to generate myogenic progenitors or symmetric divisions to expand the satellite cell pool. Thus, a complex balance between extrinsic cues and intrinsic regulatory mechanisms is needed to tightly control satellite cell cycle progression and cell fate determination. Defects in satellite cell regulation or in their niche, as observed in degenerative conditions such as aging, can impair muscle regeneration. Here, we review recent discoveries of the intrinsic and extrinsic factors that regulate satellite cell behaviour in regenerating and degenerating muscles. Summary: This Review discusses how satellite stem cell behaviour is regulated during regeneration and degeneration by a complex balance between extrinsic cues and intrinsic regulatory mechanisms.
Collapse
Affiliation(s)
- Nicolas A Dumont
- Sprott Centre for Stem Cell Research, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada K1H 8L6
| | - Yu Xin Wang
- Sprott Centre for Stem Cell Research, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada K1H 8L6 Faculty of Medicine, Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario, Canada K1H 8M5
| | - Michael A Rudnicki
- Sprott Centre for Stem Cell Research, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada K1H 8L6 Faculty of Medicine, Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario, Canada K1H 8M5
| |
Collapse
|
39
|
Blais A. Myogenesis in the Genomics Era. J Mol Biol 2015; 427:2023-38. [DOI: 10.1016/j.jmb.2015.02.009] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2014] [Revised: 02/04/2015] [Accepted: 02/05/2015] [Indexed: 01/06/2023]
|
40
|
Fu X, Wang H, Hu P. Stem cell activation in skeletal muscle regeneration. Cell Mol Life Sci 2015; 72:1663-77. [PMID: 25572293 PMCID: PMC4412728 DOI: 10.1007/s00018-014-1819-5] [Citation(s) in RCA: 95] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2014] [Revised: 12/21/2014] [Accepted: 12/22/2014] [Indexed: 12/31/2022]
Abstract
Muscle stem cell (satellite cell) activation post muscle injury is a transient and critical step in muscle regeneration. It is regulated by physiological cues, signaling molecules, and epigenetic regulatory factors. The mechanisms that coherently turn on the complex activation process shortly after trauma are just beginning to be illuminated. In this review, we will discuss the current knowledge of satellite cell activation regulation.
Collapse
Affiliation(s)
- Xin Fu
- State Key Laboratory of Cell Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai, 200031, China
| | | | | |
Collapse
|
41
|
Comai G, Sambasivan R, Gopalakrishnan S, Tajbakhsh S. Variations in the Efficiency of Lineage Marking and Ablation Confound Distinctions between Myogenic Cell Populations. Dev Cell 2014; 31:654-67. [DOI: 10.1016/j.devcel.2014.11.005] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2013] [Revised: 06/16/2014] [Accepted: 11/04/2014] [Indexed: 11/24/2022]
|
42
|
Vahidi Ferdousi L, Rocheteau P, Chayot R, Montagne B, Chaker Z, Flamant P, Tajbakhsh S, Ricchetti M. More efficient repair of DNA double-strand breaks in skeletal muscle stem cells compared to their committed progeny. Stem Cell Res 2014; 13:492-507. [PMID: 25262445 DOI: 10.1016/j.scr.2014.08.005] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2013] [Revised: 07/14/2014] [Accepted: 08/15/2014] [Indexed: 01/17/2023] Open
Abstract
The loss of genome integrity in adult stem cells results in accelerated tissue aging and is possibly cancerogenic. Adult stem cells in different tissues appear to react robustly to DNA damage. We report that adult skeletal stem (satellite) cells do not primarily respond to radiation-induced DNA double-strand breaks (DSBs) via differentiation and exhibit less apoptosis compared to other myogenic cells. Satellite cells repair these DNA lesions more efficiently than their committed progeny. Importantly, non-proliferating satellite cells and post-mitotic nuclei in the fiber exhibit dramatically distinct repair efficiencies. Altogether, reduction of the repair capacity appears to be more a function of differentiation than of the proliferation status of the muscle cell. Notably, satellite cells retain a high efficiency of DSB repair also when isolated from the natural niche. Finally, we show that repair of DSB substrates is not only very efficient but, surprisingly, also very accurate in satellite cells and that accurate repair depends on the key non-homologous end-joining factor DNA-PKcs.
Collapse
Affiliation(s)
- Leyla Vahidi Ferdousi
- Institut Pasteur, Yeast Molecular Genetics, Dept. of Genomes and Genetics, 25 rue du Dr. Roux, 75724 Paris Cedex 15, France; CNRS UMR 3525, Team Stability of Nuclear and Mitochondrial DNA, Paris, France; Sorbonne Universités, UPMC, University of Paris 06, IFD-ED 515, Place Jussieu, Paris, 72252, France
| | - Pierre Rocheteau
- Institut Pasteur, Stem Cells & Development, Dept. of Developmental and Stem Cell Biology, 25 rue du Dr. Roux, 75724 Paris Cedex 15, France; CNRS URA 2578, Paris, France
| | - Romain Chayot
- Institut Pasteur, Yeast Molecular Genetics, Dept. of Genomes and Genetics, 25 rue du Dr. Roux, 75724 Paris Cedex 15, France; CNRS UMR 3525, Team Stability of Nuclear and Mitochondrial DNA, Paris, France
| | - Benjamin Montagne
- Institut Pasteur, Yeast Molecular Genetics, Dept. of Genomes and Genetics, 25 rue du Dr. Roux, 75724 Paris Cedex 15, France; CNRS UMR 3525, Team Stability of Nuclear and Mitochondrial DNA, Paris, France
| | - Zayna Chaker
- Institut Pasteur, Yeast Molecular Genetics, Dept. of Genomes and Genetics, 25 rue du Dr. Roux, 75724 Paris Cedex 15, France; CNRS UMR 3525, Team Stability of Nuclear and Mitochondrial DNA, Paris, France
| | - Patricia Flamant
- Institut Pasteur, Stem Cells & Development, Dept. of Developmental and Stem Cell Biology, 25 rue du Dr. Roux, 75724 Paris Cedex 15, France; CNRS URA 2578, Paris, France
| | - Shahragim Tajbakhsh
- Institut Pasteur, Stem Cells & Development, Dept. of Developmental and Stem Cell Biology, 25 rue du Dr. Roux, 75724 Paris Cedex 15, France; CNRS URA 2578, Paris, France
| | - Miria Ricchetti
- Institut Pasteur, Yeast Molecular Genetics, Dept. of Genomes and Genetics, 25 rue du Dr. Roux, 75724 Paris Cedex 15, France; CNRS UMR 3525, Team Stability of Nuclear and Mitochondrial DNA, Paris, France.
| |
Collapse
|
43
|
Crowell EF, Gaffuri AL, Gayraud-Morel B, Tajbakhsh S, Echard A. Engulfment of the midbody remnant after cytokinesis in mammalian cells. J Cell Sci 2014; 127:3840-51. [PMID: 25002399 DOI: 10.1242/jcs.154732] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The midbody remnant (MBR) that is generated after cytokinetic abscission has recently attracted a lot of attention, because it might have crucial consequences for cell differentiation and tumorigenesis in mammalian cells. In these cells, it has been reported that the MBR is either released into the extracellular medium or retracted into one of the two daughter cells where it can be degraded by autophagy. Here, we describe a major alternative pathway in a variety of human and mouse immortalized cells, cancer cells and primary stem cells. Using correlative light and scanning electron microscopy and quantitative assays, we found that sequential abscissions on both sides of the midbody generate free MBRs, which are tightly associated with the cell surface through a Ca(2+)/Mg(2+)-dependent receptor. Surprisingly, MBRs move over the cell surface for several hours, before being eventually engulfed by an actin-dependent phagocytosis-like mechanism. Mathematical modeling combined with experimentation further demonstrates that lysosomal activities fully account for the clearance of MBRs after engulfment. This study changes our understanding of how MBRs are inherited and degraded in mammalian cells and suggests a mechanism by which MBRs might signal over long distances between cells.
Collapse
Affiliation(s)
- Elizabeth Faris Crowell
- Institut Pasteur, Membrane Traffic and Cell Division Lab, Department of Cell Biology and Infection, 25 Rue du Dr Roux, 75015 Paris, France CNRS URA 2582, F-75015 Paris, France
| | - Anne-Lise Gaffuri
- Institut Pasteur, Membrane Traffic and Cell Division Lab, Department of Cell Biology and Infection, 25 Rue du Dr Roux, 75015 Paris, France CNRS URA 2582, F-75015 Paris, France
| | - Barbara Gayraud-Morel
- Institut Pasteur, Stem Cells and Development, Department of Developmental & Stem Cell Biology, CNRS URA 2578, 25 Rue du Dr Roux, F-75015 Paris, France
| | - Shahragim Tajbakhsh
- Institut Pasteur, Stem Cells and Development, Department of Developmental & Stem Cell Biology, CNRS URA 2578, 25 Rue du Dr Roux, F-75015 Paris, France
| | - Arnaud Echard
- Institut Pasteur, Membrane Traffic and Cell Division Lab, Department of Cell Biology and Infection, 25 Rue du Dr Roux, 75015 Paris, France CNRS URA 2582, F-75015 Paris, France
| |
Collapse
|
44
|
Chakkalakal JV, Christensen J, Xiang W, Tierney MT, Boscolo FS, Sacco A, Brack AS. Early forming label-retaining muscle stem cells require p27kip1 for maintenance of the primitive state. Development 2014; 141:1649-59. [PMID: 24715455 PMCID: PMC3978835 DOI: 10.1242/dev.100842] [Citation(s) in RCA: 105] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Across different niches, subsets of highly functional stem cells are maintained in a relatively dormant rather than proliferative state. Our understanding of proliferative dynamics in tissue-specific stem cells during conditions of increased tissue turnover remains limited. Using a TetO-H2B-GFP reporter of proliferative history, we identify skeletal muscle stem cell, or satellite cells, that retain (LRC) or lose (nonLRC) the H2B-GFP label. We show in mice that LRCs and nonLRCs are formed at birth and persist during postnatal growth and adult muscle repair. Functionally, LRCs and nonLRCs are born equivalent and transition during postnatal maturation into distinct and hierarchically organized subsets. Adult LRCs give rise to LRCs and nonLRCs; the former are able to self-renew, whereas the latter are restricted to differentiation. Expression analysis revealed the CIP/KIP family members p21(cip1) (Cdkn1a) and p27(kip1) (Cdkn1b) to be expressed at higher levels in LRCs. In accordance with a crucial role in LRC fate, loss of p27(kip1) promoted proliferation and differentiation of LRCs in vitro and impaired satellite cell self-renewal after muscle injury. By contrast, loss of p21(cip1) only affected nonLRCs, in which myogenic commitment was inhibited. Our results provide evidence that restriction of self-renewal potential to LRCs is established early in life and is maintained during increased tissue turnover through the cell cycle inhibitor p27(kip1). They also reveal the differential role of CIP/KIP family members at discrete steps within the stem cell hierarchy.
Collapse
Affiliation(s)
- Joe V Chakkalakal
- Massachusetts General Hospital, Center of Regenerative Medicine, Harvard University, Boston, MA 02114, USA
| | | | | | | | | | | | | |
Collapse
|
45
|
Rossi G, Messina G. Comparative myogenesis in teleosts and mammals. Cell Mol Life Sci 2014; 71:3081-99. [PMID: 24664432 PMCID: PMC4111864 DOI: 10.1007/s00018-014-1604-5] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2013] [Revised: 02/17/2014] [Accepted: 03/06/2014] [Indexed: 01/02/2023]
Abstract
Skeletal myogenesis has been and is currently under extensive study in both mammals and teleosts, with the latter providing a good model for skeletal myogenesis because of their flexible and conserved genome. Parallel investigations of muscle studies using both these models have strongly accelerated the advances in the field. However, when transferring the knowledge from one model to the other, it is important to take into account both their similarities and differences. The main difficulties in comparing mammals and teleosts arise from their different temporal development. Conserved aspects can be seen for muscle developmental origin and segmentation, and for the presence of multiple myogenic waves. Among the divergences, many fish have an indeterminate growth capacity throughout their entire life span, which is absent in mammals, thus implying different post-natal growth mechanisms. This review covers the current state of the art on myogenesis, with a focus on the most conserved and divergent aspects between mammals and teleosts.
Collapse
Affiliation(s)
- Giuliana Rossi
- Department of Biosciences, University of Milan, 20133, Milan, Italy
| | | |
Collapse
|
46
|
Motohashi N, Asakura A. Muscle satellite cell heterogeneity and self-renewal. Front Cell Dev Biol 2014; 2:1. [PMID: 25364710 PMCID: PMC4206996 DOI: 10.3389/fcell.2014.00001] [Citation(s) in RCA: 133] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2013] [Accepted: 01/14/2014] [Indexed: 01/17/2023] Open
Abstract
Adult skeletal muscle possesses extraordinary regeneration capacities. After muscle injury or exercise, large numbers of newly formed muscle fibers are generated within a week as a result of expansion and differentiation of a self-renewing pool of muscle stem cells termed muscle satellite cells. Normally, satellite cells are mitotically quiescent and reside beneath the basal lamina of muscle fibers. Upon regeneration, satellite cells are activated, and give rise to daughter myogenic precursor cells. After several rounds of proliferation, these myogenic precursor cells contribute to the formation of new muscle fibers. During cell division, a minor population of myogenic precursor cells returns to quiescent satellite cells as a self-renewal process. Currently, accumulating evidence has revealed the essential roles of satellite cells in muscle regeneration and the regulatory mechanisms, while it still remains to be elucidated how satellite cell self-renewal is molecularly regulated and how satellite cells are important in aging and diseased muscle. The number of satellite cells is decreased due to the changing niche during ageing, resulting in attenuation of muscle regeneration capacity. Additionally, in Duchenne muscular dystrophy (DMD) patients, the loss of satellite cell regenerative capacity and decreased satellite cell number due to continuous needs for satellite cells lead to progressive muscle weakness with chronic degeneration. Thus, it is necessary to replenish muscle satellite cells continuously. This review outlines recent findings regarding satellite cell heterogeneity, asymmetric division and molecular mechanisms in satellite cell self-renewal which is crucial for maintenance of satellite cells as a muscle stem cell pool throughout life. In addition, we discuss roles in the stem cell niche for satellite cell maintenance, as well as related cell therapies for approaching treatment of DMD.
Collapse
Affiliation(s)
- Norio Motohashi
- Department of Neurology, Paul and Sheila Wellstone Muscular Dystrophy Center, Stem Cell Institute, University of Minnesota Medical School Minneapolis, MN, USA
| | - Atsushi Asakura
- Department of Neurology, Paul and Sheila Wellstone Muscular Dystrophy Center, Stem Cell Institute, University of Minnesota Medical School Minneapolis, MN, USA
| |
Collapse
|
47
|
Mourikis P, Tajbakhsh S. Distinct contextual roles for Notch signalling in skeletal muscle stem cells. BMC DEVELOPMENTAL BIOLOGY 2014; 14:2. [PMID: 24472470 PMCID: PMC3903015 DOI: 10.1186/1471-213x-14-2] [Citation(s) in RCA: 82] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/01/2013] [Accepted: 01/13/2014] [Indexed: 11/17/2022]
Abstract
Notch signalling acts in virtually every tissue during the lifetime of metazoans. Recent studies have pointed to multiple roles for Notch in stem cells during quiescence, proliferation, temporal specification, and maintenance of the niche architecture. Skeletal muscle has served as an excellent paradigm to examine these diverse roles as embryonic, foetal, and adult skeletal muscle stem cells have different molecular signatures and functional properties, reflecting their developmental specification during ontology. Notably, Notch signalling has emerged as a major regulator of all muscle stem cells. This review will provide an overview of Notch signalling during myogenic development and postnatally, and underscore the seemingly opposing contextual activities of Notch that have lead to a reassessment of its role in myogenesis.
Collapse
Affiliation(s)
| | - Shahragim Tajbakhsh
- Stem Cells and Development, CNRS URA 2578, Department of Developmental & Stem Cell Biology, Institut Pasteur, 25 rue du Dr, Roux, 75015 Paris, France.
| |
Collapse
|
48
|
Abstract
Since the seminal discovery of the cell-fate regulator Myod, studies in skeletal myogenesis have inspired the search for cell-fate regulators of similar potential in other tissues and organs. It was perplexing that a similar transcription factor for other tissues was not found; however, it was later discovered that combinations of molecular regulators can divert somatic cell fates to other cell types. With the new era of reprogramming to induce pluripotent cells, the myogenesis paradigm can now be viewed under a different light. Here, we provide a short historical perspective and focus on how the regulation of skeletal myogenesis occurs distinctly in different scenarios and anatomical locations. In addition, some interesting features of this tissue underscore the importance of reconsidering the simple-minded view that a single stem cell population emerges after gastrulation to assure tissuegenesis. Notably, a self-renewing long-term Pax7+ myogenic stem cell population emerges during development only after a first wave of terminal differentiation occurs to establish a tissue anlagen in the mouse. How the future stem cell population is selected in this unusual scenario will be discussed. Recently, a wealth of information has emerged from epigenetic and genome-wide studies in myogenic cells. Although key transcription factors such as Pax3, Pax7, and Myod regulate only a small subset of genes, in some cases their genomic distribution and binding are considerably more promiscuous. This apparent nonspecificity can be reconciled in part by the permissivity of the cell for myogenic commitment, and also by new roles for some of these regulators as pioneer transcription factors acting on chromatin state.
Collapse
Affiliation(s)
- Glenda Comai
- Stem Cells and Development, CNRS URA 2578, Department of Developmental & Stem Cell Biology, Institut Pasteur, Paris, France
| | - Shahragim Tajbakhsh
- Stem Cells and Development, CNRS URA 2578, Department of Developmental & Stem Cell Biology, Institut Pasteur, Paris, France.
| |
Collapse
|
49
|
Zanou N, Gailly P. Skeletal muscle hypertrophy and regeneration: interplay between the myogenic regulatory factors (MRFs) and insulin-like growth factors (IGFs) pathways. Cell Mol Life Sci 2013; 70:4117-30. [PMID: 23552962 PMCID: PMC11113627 DOI: 10.1007/s00018-013-1330-4] [Citation(s) in RCA: 214] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2013] [Revised: 03/19/2013] [Accepted: 03/19/2013] [Indexed: 10/27/2022]
Abstract
Adult skeletal muscle can regenerate in response to muscle damage. This ability is conferred by the presence of myogenic stem cells called satellite cells. In response to stimuli such as injury or exercise, these cells become activated and express myogenic regulatory factors (MRFs), i.e., transcription factors of the myogenic lineage including Myf5, MyoD, myogenin, and Mrf4 to proliferate and differentiate into myofibers. The MRF family of proteins controls the transcription of important muscle-specific proteins such as myosin heavy chain and muscle creatine kinase. Different growth factors are secreted during muscle repair among which insulin-like growth factors (IGFs) are the only ones that promote both muscle cell proliferation and differentiation and that play a key role in muscle regeneration and hypertrophy. Different isoforms of IGFs are expressed during muscle repair: IGF-IEa, IGF-IEb, or IGF-IEc (also known as mechano growth factor, MGF) and IGF-II. MGF is expressed first and is observed in satellite cells and in proliferating myoblasts whereas IGF-Ia and IGF-II expression occurs at the state of muscle fiber formation. Interestingly, several studies report the induction of MRFs in response to IGFs stimulation. Inversely, IGFs expression may also be regulated by MRFs. Various mechanisms are proposed to support these interactions. In this review, we describe the general process of muscle hypertrophy and regeneration and decipher the interactions between the two groups of factors involved in the process.
Collapse
Affiliation(s)
- Nadège Zanou
- Laboratory of Cell Physiology, Institute of Neuroscience, Université catholique de Louvain, 55 av. Hippocrate, B1.55.12, 1200, Brussels, Belgium,
| | | |
Collapse
|
50
|
Ancestral Myf5 gene activity in periocular connective tissue identifies a subset of fibro/adipogenic progenitors but does not connote a myogenic origin. Dev Biol 2013; 385:366-79. [PMID: 23969310 DOI: 10.1016/j.ydbio.2013.08.010] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2013] [Revised: 07/21/2013] [Accepted: 08/13/2013] [Indexed: 11/23/2022]
Abstract
Extraocular muscles (EOM) represent a unique muscle group that controls eye movements and originates from head mesoderm, while the more typically studied body and limb muscles are somite-derived. Aiming to investigate myogenic progenitors (satellite cells) in EOM versus limb and diaphragm of adult mice, we have been using flow cytometry in combination with myogenic-specific Cre-loxP lineage marking for cell isolation. While analyzing cells from the EOM of mice that harbor Myf5(Cre)-driven GFP expression, we identified in addition to the expected GFP(+) myogenic cells (presumably satellite cells), a second dominant GFP(+) population distinguished as being Sca1(+), non-myogenic, and exhibiting a fibro/adipogenic potential. This unexpected population was not only unique to EOM compared to the other muscles but also specific to the Myf5(Cre)-driven reporter when compared to the MyoD(Cre) driver. Histological studies of periocular tissue preparations demonstrated the presence of Myf5(Cre)-driven GFP(+) cells in connective tissue locations adjacent to the muscle masses, including cells in the vasculature wall. These vasculature-associated GFP(+) cells were further identified as mural cells based on the presence of the specific XLacZ4 transgene. Unlike the EOM satellite cells that originate from a Pax3-negative lineage, these non-myogenic Myf5(Cre)-driven GFP(+) cells appear to be related to cells of a Pax3-expressing origin, presumably derived from the neural crest. In all, our lineage tracing based on multiple reporter lines has demonstrated that regardless of common ancestral expression of Myf5, there is a clear distinction between periocular myogenic and non-myogenic cell lineages according to their mutually exclusive antecedence of MyoD and Pax3 gene activity.
Collapse
|