51
|
Root ZD, Allen C, Gould C, Brewer M, Jandzik D, Medeiros DM. A Comprehensive Analysis of Fibrillar Collagens in Lamprey Suggests a Conserved Role in Vertebrate Musculoskeletal Evolution. Front Cell Dev Biol 2022; 10:809979. [PMID: 35242758 PMCID: PMC8887668 DOI: 10.3389/fcell.2022.809979] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Accepted: 01/18/2022] [Indexed: 12/03/2022] Open
Abstract
Vertebrates have distinct tissues which are not present in invertebrate chordates nor other metazoans. The rise of these tissues also coincided with at least one round of whole-genome duplication as well as a suite of lineage-specific segmental duplications. Understanding whether novel genes lead to the origin and diversification of novel cell types, therefore, is of great importance in vertebrate evolution. Here we were particularly interested in the evolution of the vertebrate musculoskeletal system, the muscles and connective tissues that support a diversity of body plans. A major component of the musculoskeletal extracellular matrix (ECM) is fibrillar collagens, a gene family which has been greatly expanded upon in vertebrates. We thus asked whether the repertoire of fibrillar collagens in vertebrates reflects differences in the musculoskeletal system. To test this, we explored the diversity of fibrillar collagens in lamprey, a jawless vertebrate which diverged from jawed vertebrates (gnathostomes) more than five hundred million years ago and has undergone its own gene duplications. Some of the principal components of vertebrate hyaline cartilage are the fibrillar collagens type II and XI, but their presence in cartilage development across all vertebrate taxa has been disputed. We particularly emphasized the characterization of genes in the lamprey hyaline cartilage, testing if its collagen repertoire was similar to that in gnathostomes. Overall, we discovered thirteen fibrillar collagens from all known gene subfamilies in lamprey and were able to identify several lineage-specific duplications. We found that, while the collagen loci have undergone rearrangement, the Clade A genes have remained linked with the hox clusters, a phenomenon also seen in gnathostomes. While the lamprey muscular tissue was largely similar to that seen in gnathostomes, we saw considerable differences in the larval lamprey skeletal tissue, with distinct collagen combinations pertaining to different cartilage types. Our gene expression analyses were unable to identify type II collagen in the sea lamprey hyaline cartilage nor any other fibrillar collagen during chondrogenesis at the stages observed, meaning that sea lamprey likely no longer require these genes during early cartilage development. Our findings suggest that fibrillar collagens were multifunctional across the musculoskeletal system in the last common ancestor of vertebrates and have been largely conserved, but these genes alone cannot explain the origin of novel cell types.
Collapse
Affiliation(s)
- Zachary D Root
- Department of Ecology and Evolutionary Biology, University of Colorado, Boulder, CO, United States
| | - Cara Allen
- Department of Ecology and Evolutionary Biology, University of Colorado, Boulder, CO, United States
| | - Claire Gould
- Department of Ecology and Evolutionary Biology, University of Colorado, Boulder, CO, United States
| | - Margaux Brewer
- Department of Ecology and Evolutionary Biology, University of Colorado, Boulder, CO, United States
| | - David Jandzik
- Department of Ecology and Evolutionary Biology, University of Colorado, Boulder, CO, United States.,Department of Zoology, Comenius University in Bratislava, Bratislava, Slovakia
| | - Daniel M Medeiros
- Department of Ecology and Evolutionary Biology, University of Colorado, Boulder, CO, United States
| |
Collapse
|
52
|
Grimaldi A, Comai G, Mella S, Tajbakhsh S. Identification of bipotent progenitors that give rise to myogenic and connective tissues in mouse. eLife 2022; 11:70235. [PMID: 35225230 PMCID: PMC9020825 DOI: 10.7554/elife.70235] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Accepted: 02/25/2022] [Indexed: 11/19/2022] Open
Abstract
How distinct cell fates are manifested by direct lineage ancestry from bipotent progenitors, or by specification of individual cell types is a key question for understanding the emergence of tissues. The interplay between skeletal muscle progenitors and associated connective tissue cells provides a model for examining how muscle functional units are established. Most craniofacial structures originate from the vertebrate-specific neural crest cells except in the dorsal portion of the head, where they arise from cranial mesoderm. Here, using multiple lineage-tracing strategies combined with single cell RNAseq and in situ analyses, we identify bipotent progenitors expressing Myf5 (an upstream regulator of myogenic fate) that give rise to both muscle and juxtaposed connective tissue. Following this bifurcation, muscle and connective tissue cells retain complementary signalling features and maintain spatial proximity. Disrupting myogenic identity shifts muscle progenitors to a connective tissue fate. The emergence of Myf5-derived connective tissue is associated with the activity of several transcription factors, including Foxp2. Interestingly, this unexpected bifurcation in cell fate was not observed in craniofacial regions that are colonised by neural crest cells. Therefore, we propose that an ancestral bi-fated program gives rise to muscle and connective tissue cells in skeletal muscles that are deprived of neural crest cells.
Collapse
Affiliation(s)
| | - Glenda Comai
- UMR 3738, Department of Developmental and Stem Cell Biology, CNRS, Paris, France
| | - Sebastien Mella
- Cytometry and Biomarkers UTechS, Institut Pasteur, Paris, France
| | | |
Collapse
|
53
|
Cicatiello AG, Sagliocchi S, Nappi A, Di Cicco E, Miro C, Murolo M, Stornaiuolo M, Dentice M. Thyroid hormone regulates glutamine metabolism and anaplerotic fluxes by inducing mitochondrial glutamate aminotransferase GPT2. Cell Rep 2022; 38:110409. [PMID: 35196498 PMCID: PMC8889437 DOI: 10.1016/j.celrep.2022.110409] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 12/29/2021] [Accepted: 01/27/2022] [Indexed: 12/27/2022] Open
Abstract
Thyroid hormones (THs) are key metabolic regulators coordinating short- and long-term energy needs. In skeletal muscle, THs modulate energy metabolism in pathophysiological conditions. Indeed, hypo- and hyperthyroidism are leading causes of muscle weakness and strength; however, the metabolic pathways underlying these effects are still poorly understood. Using molecular, biochemical, and isotope-tracing approaches combined with mass spectrometry and denervation experiments, we find that THs regulate glutamine metabolism and anaplerotic fluxes by up-regulating the glutamate pyruvate transaminase 2 (GPT2) gene. In humans, GPT2 autosomal recessive mutations cause a neurological syndrome characterized by intellectual disability, microcephaly, and progressive motor symptoms. Here, we demonstrate a role of the TH/GPT2 axis in skeletal muscle in which it regulates muscle weight and fiber diameter in resting and atrophic conditions and results in protection from muscle loss during atrophy. These results describe an anabolic route by which THs rewire glutamine metabolism toward the maintenance of muscle mass. THs induce the expression of the mitochondrial GPT2 gene in skeletal muscle The GPT2 up-regulation by THs enhances anaplerotic cycles and α-KG production GPT2 is reduced during muscle atrophy and is reactivated by THs treatment GPT2 KO+/– mice undergo muscle loss that is partially attenuated by THs
Collapse
Affiliation(s)
| | - Serena Sagliocchi
- Department of Clinical Medicine and Surgery, University of Naples "Federico II", Naples, NA 80138 Italy
| | - Annarita Nappi
- Department of Clinical Medicine and Surgery, University of Naples "Federico II", Naples, NA 80138 Italy
| | - Emery Di Cicco
- Department of Clinical Medicine and Surgery, University of Naples "Federico II", Naples, NA 80138 Italy
| | - Caterina Miro
- Department of Clinical Medicine and Surgery, University of Naples "Federico II", Naples, NA 80138 Italy
| | - Melania Murolo
- Department of Clinical Medicine and Surgery, University of Naples "Federico II", Naples, NA 80138 Italy
| | - Mariano Stornaiuolo
- Department of Pharmacy, University of Naples "Federico II", Naples, NA 80138 Italy
| | - Monica Dentice
- Department of Clinical Medicine and Surgery, University of Naples "Federico II", Naples, NA 80138 Italy; CEINGE-Biotecnologie Avanzate Scarl, Naples, NA 80131, Italy.
| |
Collapse
|
54
|
Sahinyan K, Blackburn DM, Simon MM, Lazure F, Kwan T, Bourque G, Soleimani VD. Application of ATAC-Seq for genome-wide analysis of the chromatin state at single myofiber resolution. eLife 2022; 11:72792. [PMID: 35188098 PMCID: PMC8901173 DOI: 10.7554/elife.72792] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Accepted: 02/09/2022] [Indexed: 12/11/2022] Open
Abstract
Myofibers are the main components of skeletal muscle, which is the largest tissue in the body. Myofibers are highly adaptive and can be altered under different biological and disease conditions. Therefore, transcriptional and epigenetic studies on myofibers are crucial to discover how chromatin alterations occur in the skeletal muscle under different conditions. However, due to the heterogenous nature of skeletal muscle, studying myofibers in isolation proves to be a challenging task. Single-cell sequencing has permitted the study of the epigenome of isolated myonuclei. While this provides sequencing with high dimensionality, the sequencing depth is lacking, which makes comparisons between different biological conditions difficult. Here, we report the first implementation of single myofiber ATAC-Seq, which allows for the sequencing of an individual myofiber at a depth sufficient for peak calling and for comparative analysis of chromatin accessibility under various physiological and disease conditions. Application of this technique revealed significant differences in chromatin accessibility between resting and regenerating myofibers, as well as between myofibers from a mouse model of Duchenne Muscular Dystrophy (mdx) and wild-type (WT) counterparts. This technique can lead to a wide application in the identification of chromatin regulatory elements and epigenetic mechanisms in muscle fibers during development and in muscle-wasting diseases.
Collapse
Affiliation(s)
- Korin Sahinyan
- Department of Human Genetics, McGill University, Montreal, Canada.,Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, Canada
| | - Darren M Blackburn
- Department of Human Genetics, McGill University, Montreal, Canada.,Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, Canada
| | - Marie-Michelle Simon
- Department of Human Genetics, McGill University, Montreal, Canada.,McGill Genome Centre, Montreal, Canada
| | - Felicia Lazure
- Department of Human Genetics, McGill University, Montreal, Canada.,Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, Canada
| | - Tony Kwan
- Department of Human Genetics, McGill University, Montreal, Canada.,McGill Genome Centre, Montreal, Canada
| | - Guillaume Bourque
- Department of Human Genetics, McGill University, Montreal, Canada.,McGill Genome Centre, Montreal, Canada.,Canadian Centre for Computational Genomics, Montreal, Canada
| | - Vahab D Soleimani
- Department of Human Genetics, McGill University, Montreal, Canada.,Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, Canada
| |
Collapse
|
55
|
Della Gaspera B, Weill L, Chanoine C. Evolution of Somite Compartmentalization: A View From Xenopus. Front Cell Dev Biol 2022; 9:790847. [PMID: 35111756 PMCID: PMC8802780 DOI: 10.3389/fcell.2021.790847] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Accepted: 11/26/2021] [Indexed: 11/13/2022] Open
Abstract
Somites are transitory metameric structures at the basis of the axial organization of vertebrate musculoskeletal system. During evolution, somites appear in the chordate phylum and compartmentalize mainly into the dermomyotome, the myotome, and the sclerotome in vertebrates. In this review, we summarized the existing literature about somite compartmentalization in Xenopus and compared it with other anamniote and amniote vertebrates. We also present and discuss a model that describes the evolutionary history of somite compartmentalization from ancestral chordates to amniote vertebrates. We propose that the ancestral organization of chordate somite, subdivided into a lateral compartment of multipotent somitic cells (MSCs) and a medial primitive myotome, evolves through two major transitions. From ancestral chordates to vertebrates, the cell potency of MSCs may have evolved and gave rise to all new vertebrate compartments, i.e., the dermomyome, its hypaxial region, and the sclerotome. From anamniote to amniote vertebrates, the lateral MSC territory may expand to the whole somite at the expense of primitive myotome and may probably facilitate sclerotome formation. We propose that successive modifications of the cell potency of some type of embryonic progenitors could be one of major processes of the vertebrate evolution.
Collapse
|
56
|
Jacques E, Kuang Y, Kann AP, Le Grand F, Krauss RS, Gilbert PM. The mini-IDLE 3D biomimetic culture assay enables interrogation of mechanisms governing muscle stem cell quiescence and niche repopulation. eLife 2022; 11:81738. [PMID: 36537758 PMCID: PMC9904761 DOI: 10.7554/elife.81738] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2022] [Accepted: 12/19/2022] [Indexed: 12/24/2022] Open
Abstract
Adult skeletal muscle harbours a population of muscle stem cells (MuSCs) that are required for repair after tissue injury. In youth, MuSCs return to a reversible state of cell-cycle arrest termed 'quiescence' after injury resolution. Conversely, some MuSCs in aged muscle remain semi-activated, causing a premature response to injuries that results in incomplete repair and eventual stem cell depletion. Regulating this balance between MuSC quiescence and activation may hold the key to restoring tissue homeostasis with age, but is incompletely understood. To fill this gap, we developed a simple and tractable in vitro method, to rapidly inactivate MuSCs freshly isolated from young murine skeletal muscle, and return them to a quiescent-like state for at least 1-week, which we name mini-IDLE (Inactivation and Dormancy LEveraged in vitro). This was achieved by introducing MuSCs into a 3D bioartificial niche comprised of a thin sheet of mouse myotubes, which we demonstrate provides the minimal cues necessary to induce quiescence. With different starting numbers of MuSCs, the assay revealed cellular heterogeneity and population-level adaptations that converged on a common niche repopulation density; behaviours previously observed only in vivo. Quiescence-associated hallmarks included a Pax7+CalcR+DDX6+MyoD-c-FOS- signature, quiescent-like morphologies, and polarized niche markers. Leveraging high-content bioimaging pipelines, we demonstrate a relationship between morphology and cell fate signatures for possible real-time morphology-based screening. When using MuSCs from aged muscle, they displayed aberrant proliferative activities and delayed inactivation kinetics, among other quiescence-associated defects that we show are partially rescued by wortmannin treatment. Thus, the assay offers an unprecedented opportunity to systematically investigate long-standing queries in areas such as regulation of pool size and functional heterogeneity within the MuSC population, and to uncover quiescence regulators in youth and age.
Collapse
Affiliation(s)
- Erik Jacques
- Institute of Biomedical Engineering, University of TorontoTorontoCanada,Donnelly Centre, University of TorontoTorontoCanada
| | - Yinni Kuang
- Donnelly Centre, University of TorontoTorontoCanada,Department of Cell and Systems Biology, University of TorontoTorontoCanada
| | - Allison P Kann
- Department of Cell, Developmental, and Regenerative Biology, Icahn School of Medicine at Mount SinaiNew YorkUnited States,Black Family Stem Cell Institute, Icahn School of Medicine at Mount SinaiNew YorkUnited States,Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount SinaiNew YorkUnited States
| | - Fabien Le Grand
- Université Claude Bernard Lyon 1, CNRS UMR 5261, INSERM U1315, Institut NeuroMyoGène - Pathophysiology and Genetics of Neuron and MuscleLyonFrance
| | - Robert S Krauss
- Department of Cell, Developmental, and Regenerative Biology, Icahn School of Medicine at Mount SinaiNew YorkUnited States,Black Family Stem Cell Institute, Icahn School of Medicine at Mount SinaiNew YorkUnited States,Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount SinaiNew YorkUnited States
| | - Penney M Gilbert
- Institute of Biomedical Engineering, University of TorontoTorontoCanada,Donnelly Centre, University of TorontoTorontoCanada,Department of Cell and Systems Biology, University of TorontoTorontoCanada
| |
Collapse
|
57
|
RhoA within myofibers controls satellite cell microenvironment to allow hypertrophic growth. iScience 2022; 25:103616. [PMID: 35106464 PMCID: PMC8786647 DOI: 10.1016/j.isci.2021.103616] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 08/02/2021] [Accepted: 12/09/2021] [Indexed: 12/13/2022] Open
Abstract
Adult skeletal muscle is a plastic tissue that can adapt its size to workload. Here, we show that RhoA within myofibers is needed for overload-induced hypertrophy by controlling satellite cell (SC) fusion to the growing myofibers without affecting protein synthesis. At the molecular level, we demonstrate that RhoA controls in a cell autonomous manner Erk1/2 activation and the expressions of extracellular matrix (ECM) regulators such as Mmp9/Mmp13/Adam8 and macrophage chemo-attractants such as Ccl3/Cx3cl1. Their decreased expression in RhoA mutants is associated with ECM and fibrillar collagen disorganization and lower macrophage infiltration. Moreover, matrix metalloproteinases inhibition and macrophage depletion in controls phenocopied the altered growth of RhoA mutants while having no effect in mutants showing that their action is RhoA-dependent. These findings unravel the implication of RhoA within myofibers, in the building of a permissive microenvironment for muscle hypertrophic growth and for SC accretion through ECM remodeling and inflammatory cell recruitment. RhoA within myofibers controls SC fusion and muscle hypertrophic growth RhoA controls the expression of Mmps and of macrophage chemoattractants (Ccl3/Cx3cl1) RhoA controls ECM remodeling and macrophage recruitment upon hypertrophy Mmp inhibition and macrophage depletion phenocopy the blunted growth of RhoA mutant muscles
Collapse
|
58
|
Junion G, Jagla K. Diversification of muscle types in Drosophila embryos. Exp Cell Res 2022; 410:112950. [PMID: 34838813 DOI: 10.1016/j.yexcr.2021.112950] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 11/22/2021] [Accepted: 11/24/2021] [Indexed: 12/31/2022]
Abstract
Drosophila embryonic somatic muscles represent a simple and tractable model system to study the gene regulatory networks that control diversification of cell types. Somatic myogenesis in Drosophila is initiated by intrinsic action of the mesodermal master gene twist, which activates a cascade of transcriptional outputs including myogenic differentiation factor Mef2, which triggers all aspects of the myogenic differentiation program. In parallel, the expression of a combinatorial code of identity transcription factors (iTFs) defines discrete particular features of each muscle fiber, such as number of fusion events, and specific attachment to tendon cells or innervation, thus ensuring diversification of muscle types. Here, we take the example of a subset of lateral transverse (LT) muscles and discuss how the iTF code and downstream effector genes progressively define individual LT properties such as fusion program, attachment and innervation. We discuss new challenges in the field including the contribution of posttranscriptional and epitranscriptomic regulation of gene expression in the diversification of cell types.
Collapse
Affiliation(s)
- Guillaume Junion
- Genetics Reproduction and Development Institute (iGReD), CNRS UMR6293, INSERM U1103, University of Clermont Auvergne, Clermont-Ferrand, France
| | - Krzysztof Jagla
- Genetics Reproduction and Development Institute (iGReD), CNRS UMR6293, INSERM U1103, University of Clermont Auvergne, Clermont-Ferrand, France.
| |
Collapse
|
59
|
Domenig SA, Bundschuh N, Lenardič A, Ghosh A, Kim I, Qabrati X, D'Hulst G, Bar-Nur O. CRISPR/Cas9 editing of directly reprogrammed myogenic progenitors restores dystrophin expression in a mouse model of muscular dystrophy. Stem Cell Reports 2021; 17:321-336. [PMID: 34995499 PMCID: PMC8828535 DOI: 10.1016/j.stemcr.2021.12.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 12/06/2021] [Accepted: 12/07/2021] [Indexed: 01/09/2023] Open
Abstract
Genetic mutations in dystrophin manifest in Duchenne muscular dystrophy (DMD), the most commonly inherited muscle disease. Here, we report on reprogramming of fibroblasts from two DMD mouse models into induced myogenic progenitor cells (iMPCs) by MyoD overexpression in concert with small molecule treatment. DMD iMPCs proliferate extensively, while expressing myogenic stem cell markers including Pax7 and Myf5. Additionally, DMD iMPCs readily give rise to multinucleated myofibers that express mature skeletal muscle markers; however, they lack DYSTROPHIN expression. Utilizing an exon skipping-based approach with CRISPR/Cas9, we report on genetic correction of the dystrophin mutation in DMD iMPCs and restoration of protein expression in vitro. Furthermore, engraftment of corrected DMD iMPCs into the muscles of dystrophic mice restored DYSTROPHIN expression and contributed to the muscle stem cell reservoir. Collectively, our findings report on a novel in vitro cellular model for DMD and utilize it in conjunction with gene editing to restore DYSTROPHIN expression in vivo. iMPCs generated from DMD mouse models DMD iMPCs are expandable and express satellite cell and differentiation markers Correction of the dystrophin mutation in DMD iMPCs utilizing CRISPR/Cas9 Engraftment of corrected DMD iMPCs restores DYSTROPHIN expression in vivo
Collapse
Affiliation(s)
- Seraina A Domenig
- Laboratory of Regenerative and Movement Biology, Institute of Human Movement Sciences and Sport, Department of Health Sciences and Technology, Swiss Federal Institute of Technology (ETH) Zurich, Schwerzenbach, Switzerland
| | - Nicola Bundschuh
- Laboratory of Regenerative and Movement Biology, Institute of Human Movement Sciences and Sport, Department of Health Sciences and Technology, Swiss Federal Institute of Technology (ETH) Zurich, Schwerzenbach, Switzerland
| | - Ajda Lenardič
- Laboratory of Regenerative and Movement Biology, Institute of Human Movement Sciences and Sport, Department of Health Sciences and Technology, Swiss Federal Institute of Technology (ETH) Zurich, Schwerzenbach, Switzerland
| | - Adhideb Ghosh
- Laboratory of Regenerative and Movement Biology, Institute of Human Movement Sciences and Sport, Department of Health Sciences and Technology, Swiss Federal Institute of Technology (ETH) Zurich, Schwerzenbach, Switzerland; Functional Genomics Center Zurich, Swiss Federal Institute of Technology (ETH) Zurich and University of Zurich, Zurich, Switzerland
| | - Inseon Kim
- Laboratory of Regenerative and Movement Biology, Institute of Human Movement Sciences and Sport, Department of Health Sciences and Technology, Swiss Federal Institute of Technology (ETH) Zurich, Schwerzenbach, Switzerland
| | - Xhem Qabrati
- Laboratory of Regenerative and Movement Biology, Institute of Human Movement Sciences and Sport, Department of Health Sciences and Technology, Swiss Federal Institute of Technology (ETH) Zurich, Schwerzenbach, Switzerland
| | - Gommaar D'Hulst
- Laboratory of Regenerative and Movement Biology, Institute of Human Movement Sciences and Sport, Department of Health Sciences and Technology, Swiss Federal Institute of Technology (ETH) Zurich, Schwerzenbach, Switzerland
| | - Ori Bar-Nur
- Laboratory of Regenerative and Movement Biology, Institute of Human Movement Sciences and Sport, Department of Health Sciences and Technology, Swiss Federal Institute of Technology (ETH) Zurich, Schwerzenbach, Switzerland.
| |
Collapse
|
60
|
Lescroart F, Dumas CE, Adachi N, Kelly RG. Emergence of heart and branchiomeric muscles in cardiopharyngeal mesoderm. Exp Cell Res 2021; 410:112931. [PMID: 34798131 DOI: 10.1016/j.yexcr.2021.112931] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 09/27/2021] [Accepted: 11/14/2021] [Indexed: 12/17/2022]
Abstract
Branchiomeric muscles of the head and neck originate in a population of cranial mesoderm termed cardiopharyngeal mesoderm that also contains progenitor cells contributing to growth of the embryonic heart. Retrospective lineage analysis has shown that branchiomeric muscles share a clonal origin with parts of the heart, indicating the presence of common heart and head muscle progenitor cells in the early embryo. Genetic lineage tracing and functional studies in the mouse, as well as in Ciona and zebrafish, together with recent experiments using single cell transcriptomics and multipotent stem cells, have provided further support for the existence of bipotent head and heart muscle progenitor cells. Current challenges concern defining where and when such common progenitor cells exist in mammalian embryos and how alternative myogenic derivatives emerge in cardiopharyngeal mesoderm. Addressing these questions will provide insights into mechanisms of cell fate acquisition and the evolution of vertebrate musculature, as well as clinical insights into the origins of muscle restricted myopathies and congenital defects affecting craniofacial and cardiac development.
Collapse
Affiliation(s)
| | - Camille E Dumas
- Aix-Marseille Université, CNRS UMR 7288, IBDM, 13009, Marseille, France
| | - Noritaka Adachi
- Aix-Marseille Université, CNRS UMR 7288, IBDM, 13009, Marseille, France
| | - Robert G Kelly
- Aix-Marseille Université, CNRS UMR 7288, IBDM, 13009, Marseille, France.
| |
Collapse
|
61
|
Collins BC, Kardon G. It takes all kinds: heterogeneity among satellite cells and fibro-adipogenic progenitors during skeletal muscle regeneration. Development 2021; 148:dev199861. [PMID: 34739030 PMCID: PMC8602941 DOI: 10.1242/dev.199861] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Vertebrate skeletal muscle is composed of multinucleate myofibers that are surrounded by muscle connective tissue. Following injury, muscle is able to robustly regenerate because of tissue-resident muscle stem cells, called satellite cells. In addition, efficient and complete regeneration depends on other cells resident in muscle - including fibro-adipogenic progenitors (FAPs). Increasing evidence from single-cell analyses and genetic and transplantation experiments suggests that satellite cells and FAPs are heterogeneous cell populations. Here, we review our current understanding of the heterogeneity of satellite cells, their myogenic derivatives and FAPs in terms of gene expression, anatomical location, age and timing during the regenerative process - each of which have potentially important functional consequences.
Collapse
Affiliation(s)
| | - Gabrielle Kardon
- Department of Human Genetics, University of Utah, Salt Lake City, UT 84112, USA
| |
Collapse
|
62
|
Control of satellite cell function in muscle regeneration and its disruption in ageing. Nat Rev Mol Cell Biol 2021; 23:204-226. [PMID: 34663964 DOI: 10.1038/s41580-021-00421-2] [Citation(s) in RCA: 230] [Impact Index Per Article: 57.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/01/2021] [Indexed: 12/19/2022]
Abstract
Skeletal muscle contains a designated population of adult stem cells, called satellite cells, which are generally quiescent. In homeostasis, satellite cells proliferate only sporadically and usually by asymmetric cell division to replace myofibres damaged by daily activity and maintain the stem cell pool. However, satellite cells can also be robustly activated upon tissue injury, after which they undergo symmetric divisions to generate new stem cells and numerous proliferating myoblasts that later differentiate to muscle cells (myocytes) to rebuild the muscle fibre, thereby supporting skeletal muscle regeneration. Recent discoveries show that satellite cells have a great degree of population heterogeneity, and that their cell fate choices during the regeneration process are dictated by both intrinsic and extrinsic mechanisms. Extrinsic cues come largely from communication with the numerous distinct stromal cell types in their niche, creating a dynamically interactive microenvironment. This Review discusses the role and regulation of satellite cells in skeletal muscle homeostasis and regeneration. In particular, we highlight the cell-intrinsic control of quiescence versus activation, the importance of satellite cell-niche communication, and deregulation of these mechanisms associated with ageing. The increasing understanding of how satellite cells are regulated will help to advance muscle regeneration and rejuvenation therapies.
Collapse
|
63
|
Grimaldi A, Tajbakhsh S. Diversity in cranial muscles: Origins and developmental programs. Curr Opin Cell Biol 2021; 73:110-116. [PMID: 34500235 DOI: 10.1016/j.ceb.2021.06.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Accepted: 06/24/2021] [Indexed: 01/14/2023]
Abstract
Cranial muscles have been the focus of many studies over the years because of their unique developmental programs and relative resistance to illnesses. In addition, head muscles possess clonal relationships with heart muscles and have been highly remodeled during vertebrate evolution. Here, we provide an overview of recent findings that have helped to redefine the boundaries and lineages of cranial mesoderm. These studies have important implications regarding the emergence of muscle connective tissues, which can share a common origin with skeletal muscle. We also highlight new regulatory networks of various muscle subgroups, particularly those derived from the most caudal arches, which remain poorly defined. Finally, we suggest future research avenues to characterize the nature of their intrinsic specificities and their emergence during evolution.
Collapse
Affiliation(s)
- Alexandre Grimaldi
- Stem Cells & Development Unit, 25 rue du Dr. Roux, Institut Pasteur, 75015 Paris, France; UMR CNRS 3738, Institut Pasteur, Paris, France
| | - Shahragim Tajbakhsh
- Stem Cells & Development Unit, 25 rue du Dr. Roux, Institut Pasteur, 75015 Paris, France; UMR CNRS 3738, Institut Pasteur, Paris, France.
| |
Collapse
|
64
|
Yagi M, Ji F, Charlton J, Cristea S, Messemer K, Horwitz N, Di Stefano B, Tsopoulidis N, Hoetker MS, Huebner AJ, Bar-Nur O, Almada AE, Yamamoto M, Patelunas A, Goldhamer DJ, Wagers AJ, Michor F, Meissner A, Sadreyev RI, Hochedlinger K. Dissecting dual roles of MyoD during lineage conversion to mature myocytes and myogenic stem cells. Genes Dev 2021; 35:1209-1228. [PMID: 34413137 PMCID: PMC8415322 DOI: 10.1101/gad.348678.121] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Accepted: 08/02/2021] [Indexed: 11/24/2022]
Abstract
The generation of myotubes from fibroblasts upon forced MyoD expression is a classic example of transcription factor-induced reprogramming. We recently discovered that additional modulation of signaling pathways with small molecules facilitates reprogramming to more primitive induced myogenic progenitor cells (iMPCs). Here, we dissected the transcriptional and epigenetic dynamics of mouse fibroblasts undergoing reprogramming to either myotubes or iMPCs using a MyoD-inducible transgenic model. Induction of MyoD in fibroblasts combined with small molecules generated Pax7+ iMPCs with high similarity to primary muscle stem cells. Analysis of intermediate stages of iMPC induction revealed that extinction of the fibroblast program preceded induction of the stem cell program. Moreover, key stem cell genes gained chromatin accessibility prior to their transcriptional activation, and these regions exhibited a marked loss of DNA methylation dependent on the Tet enzymes. In contrast, myotube generation was associated with few methylation changes, incomplete and unstable reprogramming, and an insensitivity to Tet depletion. Finally, we showed that MyoD's ability to bind to unique bHLH targets was crucial for generating iMPCs but dispensable for generating myotubes. Collectively, our analyses elucidate the role of MyoD in myogenic reprogramming and derive general principles by which transcription factors and signaling pathways cooperate to rewire cell identity.
Collapse
Affiliation(s)
- Masaki Yagi
- Department of Molecular Biology, Massachusetts General Hospital, Boston, Massachusetts 02114, USA.,Cancer Center and Center for Regenerative Medicine, Massachusetts General Hospital, Boston, Massachusetts 02114, USA.,Department of Genetics, Harvard Medical School, Boston, Massachusetts 02115, USA.,Harvard Stem Cell Institute, Cambridge, Massachusetts 02138, USA.,Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, Massachusetts 02142, USA
| | - Fei Ji
- Department of Molecular Biology, Massachusetts General Hospital, Boston, Massachusetts 02114, USA.,Department of Genetics, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Jocelyn Charlton
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, Massachusetts 02138, USA.,Department of Genome Regulation, Max-Planck-Institute for Molecular Genetics, 14195 Berlin, Germany
| | - Simona Cristea
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, Massachusetts 02138, USA.,Department of Data Science, Dana-Farber Cancer Institute, Boston, Massachusetts 02115, USA.,Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Kathleen Messemer
- Harvard Stem Cell Institute, Cambridge, Massachusetts 02138, USA.,Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, Massachusetts 02138, USA
| | - Naftali Horwitz
- Harvard Stem Cell Institute, Cambridge, Massachusetts 02138, USA.,Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, Massachusetts 02138, USA.,Joslin Diabetes Center, Boston, Massachusetts 02215, USA
| | - Bruno Di Stefano
- Department of Molecular Biology, Massachusetts General Hospital, Boston, Massachusetts 02114, USA.,Cancer Center and Center for Regenerative Medicine, Massachusetts General Hospital, Boston, Massachusetts 02114, USA.,Department of Genetics, Harvard Medical School, Boston, Massachusetts 02115, USA.,Harvard Stem Cell Institute, Cambridge, Massachusetts 02138, USA.,Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, Massachusetts 02142, USA
| | - Nikolaos Tsopoulidis
- Department of Molecular Biology, Massachusetts General Hospital, Boston, Massachusetts 02114, USA.,Cancer Center and Center for Regenerative Medicine, Massachusetts General Hospital, Boston, Massachusetts 02114, USA.,Department of Genetics, Harvard Medical School, Boston, Massachusetts 02115, USA.,Harvard Stem Cell Institute, Cambridge, Massachusetts 02138, USA.,Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, Massachusetts 02142, USA
| | - Michael S Hoetker
- Department of Molecular Biology, Massachusetts General Hospital, Boston, Massachusetts 02114, USA.,Cancer Center and Center for Regenerative Medicine, Massachusetts General Hospital, Boston, Massachusetts 02114, USA.,Department of Genetics, Harvard Medical School, Boston, Massachusetts 02115, USA.,Harvard Stem Cell Institute, Cambridge, Massachusetts 02138, USA.,Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, Massachusetts 02142, USA
| | - Aaron J Huebner
- Department of Molecular Biology, Massachusetts General Hospital, Boston, Massachusetts 02114, USA.,Cancer Center and Center for Regenerative Medicine, Massachusetts General Hospital, Boston, Massachusetts 02114, USA.,Department of Genetics, Harvard Medical School, Boston, Massachusetts 02115, USA.,Harvard Stem Cell Institute, Cambridge, Massachusetts 02138, USA.,Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, Massachusetts 02142, USA
| | - Ori Bar-Nur
- Department of Molecular Biology, Massachusetts General Hospital, Boston, Massachusetts 02114, USA.,Cancer Center and Center for Regenerative Medicine, Massachusetts General Hospital, Boston, Massachusetts 02114, USA.,Department of Genetics, Harvard Medical School, Boston, Massachusetts 02115, USA.,Harvard Stem Cell Institute, Cambridge, Massachusetts 02138, USA.,Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, Massachusetts 02142, USA
| | - Albert E Almada
- Harvard Stem Cell Institute, Cambridge, Massachusetts 02138, USA.,Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, Massachusetts 02138, USA.,Joslin Diabetes Center, Boston, Massachusetts 02215, USA
| | - Masakazu Yamamoto
- Department of Molecular and Cell Biology, University of Connecticut, Storrs, Connecticut 06269, USA
| | - Anthony Patelunas
- Department of Molecular and Cell Biology, University of Connecticut, Storrs, Connecticut 06269, USA
| | - David J Goldhamer
- Department of Molecular and Cell Biology, University of Connecticut, Storrs, Connecticut 06269, USA
| | - Amy J Wagers
- Harvard Stem Cell Institute, Cambridge, Massachusetts 02138, USA.,Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, Massachusetts 02138, USA.,Joslin Diabetes Center, Boston, Massachusetts 02215, USA
| | - Franziska Michor
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, Massachusetts 02142, USA.,Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, Massachusetts 02138, USA.,Department of Data Science, Dana-Farber Cancer Institute, Boston, Massachusetts 02115, USA.,The Center for Cancer Evolution, Dana-Farber Cancer Institute, Boston, Massachusetts 02115, USA.,The Ludwig Center at Harvard, Boston, Massachusetts 02115, USA.,Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, Massachusetts 02215, USA
| | - Alexander Meissner
- Harvard Stem Cell Institute, Cambridge, Massachusetts 02138, USA.,Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, Massachusetts 02142, USA.,Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, Massachusetts 02138, USA.,Department of Genome Regulation, Max-Planck-Institute for Molecular Genetics, 14195 Berlin, Germany
| | - Ruslan I Sadreyev
- Department of Molecular Biology, Massachusetts General Hospital, Boston, Massachusetts 02114, USA.,Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts 02114, USA
| | - Konrad Hochedlinger
- Department of Molecular Biology, Massachusetts General Hospital, Boston, Massachusetts 02114, USA.,Cancer Center and Center for Regenerative Medicine, Massachusetts General Hospital, Boston, Massachusetts 02114, USA.,Department of Genetics, Harvard Medical School, Boston, Massachusetts 02115, USA.,Harvard Stem Cell Institute, Cambridge, Massachusetts 02138, USA.,Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, Massachusetts 02142, USA
| |
Collapse
|
65
|
Translational control by DHX36 binding to 5'UTR G-quadruplex is essential for muscle stem-cell regenerative functions. Nat Commun 2021; 12:5043. [PMID: 34413292 PMCID: PMC8377060 DOI: 10.1038/s41467-021-25170-w] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Accepted: 06/06/2021] [Indexed: 12/30/2022] Open
Abstract
Skeletal muscle has a remarkable ability to regenerate owing to its resident stem cells (also called satellite cells, SCs). SCs are normally quiescent; when stimulated by damage, they activate and expand to form new fibers. The mechanisms underlying SC proliferative progression remain poorly understood. Here we show that DHX36, a helicase that unwinds RNA G-quadruplex (rG4) structures, is essential for muscle regeneration by regulating SC expansion. DHX36 (initially named RHAU) is barely expressed at quiescence but is highly induced during SC activation and proliferation. Inducible deletion of Dhx36 in adult SCs causes defective proliferation and muscle regeneration after damage. System-wide mapping in proliferating SCs reveals DHX36 binding predominantly to rG4 structures at various regions of mRNAs, while integrated polysome profiling shows that DHX36 promotes mRNA translation via 5′-untranslated region (UTR) rG4 binding. Furthermore, we demonstrate that DHX36 specifically regulates the translation of Gnai2 mRNA by unwinding its 5′ UTR rG4 structures and identify GNAI2 as a downstream effector of DHX36 for SC expansion. Altogether, our findings uncover DHX36 as an indispensable post-transcriptional regulator of SC function and muscle regeneration acting through binding and unwinding rG4 structures at 5′ UTR of target mRNAs. Skeletal muscle stem cells (or satellite cells, SCs) are normally quiescent but activate and expand in response to injury. Here the authors show that induction of DHX36 helicase during SC activation promotes mRNA translation by binding to 5′UTR mRNA G-quadruplexes (rG4) in targets including Gnai2 and unwinding them.
Collapse
|
66
|
Lahmann I, Griger J, Chen JS, Zhang Y, Schuelke M, Birchmeier C. Met and Cxcr4 cooperate to protect skeletal muscle stem cells against inflammation-induced damage during regeneration. eLife 2021; 10:57356. [PMID: 34350830 PMCID: PMC8370772 DOI: 10.7554/elife.57356] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2020] [Accepted: 08/04/2021] [Indexed: 12/15/2022] Open
Abstract
Acute skeletal muscle injury is followed by an inflammatory response, removal of damaged tissue, and the generation of new muscle fibers by resident muscle stem cells, a process well characterized in murine injury models. Inflammatory cells are needed to remove the debris at the site of injury and provide signals that are beneficial for repair. However, they also release chemokines, reactive oxygen species, as well as enzymes for clearance of damaged cells and fibers, which muscle stem cells have to withstand in order to regenerate the muscle. We show here that MET and CXCR4 cooperate to protect muscle stem cells against the adverse environment encountered during muscle repair. This powerful cyto-protective role was revealed by the genetic ablation of Met and Cxcr4 in muscle stem cells of mice, which resulted in severe apoptosis during early stages of regeneration. TNFα neutralizing antibodies rescued the apoptosis, indicating that TNFα provides crucial cell-death signals during muscle repair that are counteracted by MET and CXCR4. We conclude that muscle stem cells require MET and CXCR4 to protect them against the harsh inflammatory environment encountered in an acute muscle injury.
Collapse
Affiliation(s)
- Ines Lahmann
- Neurowissenschaftliches Forschungzentrum, NeuroCure Cluster of Excellence, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany.,Developmental Biology/Signal Transduction Group, Max Delbrueck Center for Molecular Medicine (MDC) in the Helmholtz Society, Berlin, Germany
| | - Joscha Griger
- Developmental Biology/Signal Transduction Group, Max Delbrueck Center for Molecular Medicine (MDC) in the Helmholtz Society, Berlin, Germany
| | - Jie-Shin Chen
- Developmental Biology/Signal Transduction Group, Max Delbrueck Center for Molecular Medicine (MDC) in the Helmholtz Society, Berlin, Germany
| | - Yao Zhang
- Developmental Biology/Signal Transduction Group, Max Delbrueck Center for Molecular Medicine (MDC) in the Helmholtz Society, Berlin, Germany
| | - Markus Schuelke
- Department of Neuropediatrics, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Carmen Birchmeier
- Neurowissenschaftliches Forschungzentrum, NeuroCure Cluster of Excellence, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany.,Developmental Biology/Signal Transduction Group, Max Delbrueck Center for Molecular Medicine (MDC) in the Helmholtz Society, Berlin, Germany
| |
Collapse
|
67
|
Roy N, Sundar S, Pillai M, Patell-Socha F, Ganesh S, Aloysius A, Rumman M, Gala H, Hughes SM, Zammit PS, Dhawan J. mRNP granule proteins Fmrp and Dcp1a differentially regulate mRNP complexes to contribute to control of muscle stem cell quiescence and activation. Skelet Muscle 2021; 11:18. [PMID: 34238354 PMCID: PMC8265057 DOI: 10.1186/s13395-021-00270-9] [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: 11/04/2020] [Accepted: 04/28/2021] [Indexed: 01/28/2023] Open
Abstract
Background During skeletal muscle regeneration, satellite stem cells use distinct pathways to repair damaged myofibers or to self-renew by returning to quiescence. Cellular/mitotic quiescence employs mechanisms that promote a poised or primed state, including altered RNA turnover and translational repression. Here, we investigate the role of mRNP granule proteins Fragile X Mental Retardation Protein (Fmrp) and Decapping protein 1a (Dcp1a) in muscle stem cell quiescence and differentiation. Methods Using isolated single muscle fibers from adult mice, we established differential enrichment of mRNP granule proteins including Fmrp and Dcp1a in muscle stem cells vs. myofibers. We investigated muscle tissue homeostasis in adult Fmr1-/- mice, analyzing myofiber cross-sectional area in vivo and satellite cell proliferation ex vivo. We explored the molecular mechanisms of Dcp1a and Fmrp function in quiescence, proliferation and differentiation in a C2C12 culture model. Here, we used polysome profiling, imaging and RNA/protein expression analysis to establish the abundance and assembly status of mRNP granule proteins in different cellular states, and the phenotype of knockdown cells. Results Quiescent muscle satellite cells are enriched for puncta containing the translational repressor Fmrp, but not the mRNA decay factor Dcp1a. MuSC isolated from Fmr1-/- mice exhibit defective proliferation, and mature myofibers show reduced cross-sectional area, suggesting a role for Fmrp in muscle homeostasis. Expression and organization of Fmrp and Dcp1a varies during primary MuSC activation on myofibers, with Fmrp puncta prominent in quiescence, but Dcp1a puncta appearing during activation/proliferation. This reciprocal expression of Fmrp and Dcp1a puncta is recapitulated in a C2C12 culture model of quiescence and activation: consistent with its role as a translational repressor, Fmrp is enriched in non-translating mRNP complexes abundant in quiescent myoblasts; Dcp1a puncta are lost in quiescence, suggesting stabilized and repressed transcripts. The function of each protein differs during proliferation; whereas Fmrp knockdown led to decreased proliferation and lower cyclin expression, Dcp1a knockdown led to increased cell proliferation and higher cyclin expression. However, knockdown of either Fmrp or Dcp1a led to compromised differentiation. We also observed cross-regulation of decay versus storage mRNP granules; knockdown of Fmrp enhances accumulation of Dcp1a puncta, whereas knockdown of Dcp1a leads to increased Fmrp in puncta. Conclusions Taken together, our results provide evidence that the balance of mRNA turnover versus utilization is specific for distinct cellular states. Supplementary Information The online version contains supplementary material available at 10.1186/s13395-021-00270-9.
Collapse
Affiliation(s)
- Nainita Roy
- Institute for Stem Cell Science and Regenerative Medicine, Bangalore, India
| | - Swetha Sundar
- Centre for Cellular and Molecular Biology, Hyderabad, India
| | - Malini Pillai
- Institute for Stem Cell Science and Regenerative Medicine, Bangalore, India
| | - Farah Patell-Socha
- Institute for Stem Cell Science and Regenerative Medicine, Bangalore, India
| | - Sravya Ganesh
- Institute for Stem Cell Science and Regenerative Medicine, Bangalore, India
| | - Ajoy Aloysius
- National Center for Biological Sciences, Bangalore, India
| | - Mohammed Rumman
- Institute for Stem Cell Science and Regenerative Medicine, Bangalore, India.,Manipal Academy of Higher Education, Manipal, India
| | - Hardik Gala
- Institute for Stem Cell Science and Regenerative Medicine, Bangalore, India.,Centre for Cellular and Molecular Biology, Hyderabad, India
| | - Simon M Hughes
- King's College London, Randall Centre for Cell & Molecular Biophysics, New Hunt's House, Guy's Campus, London, UK
| | - Peter S Zammit
- King's College London, Randall Centre for Cell & Molecular Biophysics, New Hunt's House, Guy's Campus, London, UK
| | - Jyotsna Dhawan
- Institute for Stem Cell Science and Regenerative Medicine, Bangalore, India. .,Centre for Cellular and Molecular Biology, Hyderabad, India.
| |
Collapse
|
68
|
Cheng X, Shi B, Li J. Distinct Embryonic Origin and Injury Response of Resident Stem Cells in Craniofacial Muscles. Front Physiol 2021; 12:690248. [PMID: 34276411 PMCID: PMC8281086 DOI: 10.3389/fphys.2021.690248] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Accepted: 05/06/2021] [Indexed: 02/05/2023] Open
Abstract
Craniofacial muscles emerge as a developmental novelty during the evolution from invertebrates to vertebrates, facilitating diversified modes of predation, feeding and communication. In contrast to the well-studied limb muscles, knowledge about craniofacial muscle stem cell biology has only recently starts to be gathered. Craniofacial muscles are distinct from their counterparts in other regions in terms of both their embryonic origin and their injury response. Compared with somite-derived limb muscles, pharyngeal arch-derived craniofacial muscles demonstrate delayed myofiber reconstitution and prolonged fibrosis during repair. The regeneration of muscle is orchestrated by a blended source of stem/progenitor cells, including myogenic muscle satellite cells (MuSCs), mesenchymal fibro-adipogenic progenitors (FAPs) and other interstitial progenitors. Limb muscles host MuSCs of the Pax3 lineage, and FAPs from the mesoderm, while craniofacial muscles have MuSCs of the Mesp1 lineage and FAPs from the ectoderm-derived neural crest. Both in vivo and in vitro data revealed distinct patterns of proliferation and differentiation in these craniofacial muscle stem/progenitor cells. Additionally, the proportion of cells of different embryonic origins changes throughout postnatal development in the craniofacial muscles, creating a more dynamic niche environment than in other muscles. In-depth comparative studies of the stem cell biology of craniofacial and limb muscles might inspire the development of novel therapeutics to improve the management of myopathic diseases. Based on the most up-to-date literature, we delineated the pivotal cell populations regulating craniofacial muscle repair and identified clues that might elucidate the distinct embryonic origin and injury response in craniofacial muscle cells.
Collapse
Affiliation(s)
- Xu Cheng
- State Key Laboratory of Oral Diseases, Department of Oral and Maxillofacial Surgery, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Bing Shi
- State Key Laboratory of Oral Diseases, Department of Oral and Maxillofacial Surgery, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Jingtao Li
- State Key Laboratory of Oral Diseases, Department of Oral and Maxillofacial Surgery, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
69
|
Mass Spectrometric Profiling of Extraocular Muscle and Proteomic Adaptations in the mdx-4cv Model of Duchenne Muscular Dystrophy. Life (Basel) 2021; 11:life11070595. [PMID: 34206383 PMCID: PMC8304255 DOI: 10.3390/life11070595] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 06/15/2021] [Accepted: 06/17/2021] [Indexed: 12/16/2022] Open
Abstract
Extraocular muscles (EOMs) represent a specialized type of contractile tissue with unique cellular, physiological, and biochemical properties. In Duchenne muscular dystrophy, EOMs stay functionally unaffected in the course of disease progression. Therefore, it was of interest to determine their proteomic profile in dystrophinopathy. The proteomic survey of wild type mice and the dystrophic mdx-4cv model revealed a broad spectrum of sarcomere-associated proteoforms, including components of the thick filament, thin filament, M-band and Z-disk, as well as a variety of muscle-specific markers. Interestingly, the mass spectrometric analysis revealed unusual expression levels of contractile proteins, especially isoforms of myosin heavy chain. As compared to diaphragm muscle, both proteomics and immunoblotting established isoform MyHC14 as a new potential marker in wild type EOMs, in addition to the previously identified isoforms MyHC13 and MyHC15. Comparative proteomics was employed to establish alterations in the protein expression profile between normal EOMs and dystrophin-lacking EOMs. The analysis of mdx-4cv EOMs identified elevated levels of glycolytic enzymes and molecular chaperones, as well as decreases in mitochondrial enzymes. These findings suggest a process of adaptation in dystrophin-deficient EOMs via a bioenergetic shift to more glycolytic metabolism, as well as an efficient cellular stress response in EOMs in dystrophinopathy.
Collapse
|
70
|
Abstract
Abnormalities in cranial motor nerve development cause paralytic strabismus syndromes, collectively referred to as congenital cranial dysinnervation disorders, in which patients cannot fully move their eyes. These disorders can arise through one of two mechanisms: (a) defective motor neuron specification, usually by loss of a transcription factor necessary for brainstem patterning, or (b) axon growth and guidance abnormalities of the oculomotor, trochlear, and abducens nerves. This review focuses on our current understanding of axon guidance mechanisms in the cranial motor nerves and how disease-causing mutations disrupt axon targeting. Abnormalities of axon growth and guidance are often limited to a single nerve or subdivision, even when the causative gene is ubiquitously expressed. Additionally, when one nerve is absent, its normal target muscles attract other motor neurons. Study of these disorders highlights the complexities of axon guidance and how each population of neurons uses a unique but overlapping set of axon guidance pathways. Expected final online publication date for the Annual Review of Vision Science, Volume 7 is September 2021. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
- Mary C Whitman
- Department of Ophthalmology, Boston Children's Hospital and Harvard Medical School, Boston, Massachusetts 02115, USA;
| |
Collapse
|
71
|
Yoshioka K, Nagahisa H, Miura F, Araki H, Kamei Y, Kitajima Y, Seko D, Nogami J, Tsuchiya Y, Okazaki N, Yonekura A, Ohba S, Sumita Y, Chiba K, Ito K, Asahina I, Ogawa Y, Ito T, Ohkawa Y, Ono Y. Hoxa10 mediates positional memory to govern stem cell function in adult skeletal muscle. SCIENCE ADVANCES 2021; 7:7/24/eabd7924. [PMID: 34108202 PMCID: PMC8189581 DOI: 10.1126/sciadv.abd7924] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Accepted: 04/21/2021] [Indexed: 05/04/2023]
Abstract
Muscle stem cells (satellite cells) are distributed throughout the body and have heterogeneous properties among muscles. However, functional topographical genes in satellite cells of adult muscle remain unidentified. Here, we show that expression of Homeobox-A (Hox-A) cluster genes accompanied with DNA hypermethylation of the Hox-A locus was robustly maintained in both somite-derived muscles and their associated satellite cells in adult mice, which recapitulates their embryonic origin. Somite-derived satellite cells were clearly separated from cells derived from cranial mesoderm in Hoxa10 expression. Hoxa10 inactivation led to genomic instability and mitotic catastrophe in somite-derived satellite cells in mice and human. Satellite cell-specific Hoxa10 ablation in mice resulted in a decline in the regenerative ability of somite-derived muscles, which were unobserved in cranial mesoderm-derived muscles. Thus, our results show that Hox gene expression profiles instill the embryonic history in satellite cells as positional memory, potentially modulating region-specific pathophysiology in adult muscles.
Collapse
Affiliation(s)
- Kiyoshi Yoshioka
- Department of Muscle Development and Regeneration, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto 860-0811, Japan
- Musculoskeletal Molecular Biology Research Group, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8588, Japan
- Department of Molecular Bone Biology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8588, Japan
| | - Hiroshi Nagahisa
- Department of Muscle Development and Regeneration, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto 860-0811, Japan
| | - Fumihito Miura
- Department of Biochemistry, Kyushu University Graduate School of Medical Sciences, Fukuoka 812-8582, Japan
| | - Hiromitsu Araki
- Department of Biochemistry, Kyushu University Graduate School of Medical Sciences, Fukuoka 812-8582, Japan
| | - Yasutomi Kamei
- Graduate School of Life and Environmental Sciences, Kyoto Prefectural University, Kyoto 606-8522, Japan
| | - Yasuo Kitajima
- Department of Muscle Development and Regeneration, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto 860-0811, Japan
- Musculoskeletal Molecular Biology Research Group, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8588, Japan
| | - Daiki Seko
- Department of Muscle Development and Regeneration, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto 860-0811, Japan
- Musculoskeletal Molecular Biology Research Group, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8588, Japan
- Department of Molecular Bone Biology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8588, Japan
| | - Jumpei Nogami
- Division of Transcriptomics, Medical Institute of Bioregulation, Kyushu University, Fukuoka 812-8582, Japan
| | - Yoshifumi Tsuchiya
- Department of Muscle Development and Regeneration, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto 860-0811, Japan
- Musculoskeletal Molecular Biology Research Group, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8588, Japan
| | - Narihiro Okazaki
- Department of Orthopaedic Surgery, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8501, Japan
| | - Akihiko Yonekura
- Department of Orthopaedic Surgery, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8501, Japan
| | - Seigo Ohba
- Department of Regenerative Oral Surgery, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8501, Japan
| | - Yoshinori Sumita
- Department of Regenerative Oral Surgery, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8501, Japan
| | - Ko Chiba
- Department of Orthopaedic Surgery, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8501, Japan
| | - Kosei Ito
- Department of Molecular Bone Biology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8588, Japan
| | - Izumi Asahina
- Department of Regenerative Oral Surgery, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8501, Japan
| | - Yoshihiro Ogawa
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan
| | - Takashi Ito
- Department of Biochemistry, Kyushu University Graduate School of Medical Sciences, Fukuoka 812-8582, Japan
| | - Yasuyuki Ohkawa
- Division of Transcriptomics, Medical Institute of Bioregulation, Kyushu University, Fukuoka 812-8582, Japan
| | - Yusuke Ono
- Department of Muscle Development and Regeneration, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto 860-0811, Japan.
- Musculoskeletal Molecular Biology Research Group, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8588, Japan
- Center for Metabolic Regulation of Healthy Aging, Kumamoto University, Kumamoto 860-0811, Japan
| |
Collapse
|
72
|
Blackburn DM, Lazure F, Soleimani VD. SMART approaches for genome-wide analyses of skeletal muscle stem and niche cells. Crit Rev Biochem Mol Biol 2021; 56:284-300. [PMID: 33823731 DOI: 10.1080/10409238.2021.1908950] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Muscle stem cells (MuSCs) also called satellite cells are the building blocks of skeletal muscle, the largest tissue in the human body which is formed primarily of myofibers. While MuSCs are the principal cells that directly contribute to the formation of the muscle fibers, their ability to do so depends on critical interactions with a vast array of nonmyogenic cells within their niche environment. Therefore, understanding the nature of communication between MuSCs and their niche is of key importance to understand how the skeletal muscle is maintained and regenerated after injury. MuSCs are rare and therefore difficult to study in vivo within the context of their niche environment. The advent of single-cell technologies, such as switching mechanism at 5' end of the RNA template (SMART) and tagmentation based technologies using hyperactive transposase, afford the unprecedented opportunity to perform whole transcriptome and epigenome studies on rare cells within their niche environment. In this review, we will delve into how single-cell technologies can be applied to the study of MuSCs and muscle-resident niche cells and the impact this can have on our understanding of MuSC biology and skeletal muscle regeneration.
Collapse
Affiliation(s)
- Darren M Blackburn
- Department of Human Genetics, McGill University, Montreal, Canada.,Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, Canada
| | - Felicia Lazure
- Department of Human Genetics, McGill University, Montreal, Canada.,Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, Canada
| | - Vahab D Soleimani
- Department of Human Genetics, McGill University, Montreal, Canada.,Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, Canada
| |
Collapse
|
73
|
Progressive and Coordinated Mobilization of the Skeletal Muscle Niche throughout Tissue Repair Revealed by Single-Cell Proteomic Analysis. Cells 2021; 10:cells10040744. [PMID: 33800595 PMCID: PMC8066646 DOI: 10.3390/cells10040744] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 03/02/2021] [Accepted: 03/22/2021] [Indexed: 12/12/2022] Open
Abstract
Background: Skeletal muscle is one of the only mammalian tissues capable of rapid and efficient regeneration after trauma or in pathological conditions. Skeletal muscle regeneration is driven by the muscle satellite cells, the stem cell population in interaction with their niche. Upon injury, muscle fibers undergo necrosis and muscle stem cells activate, proliferate and fuse to form new myofibers. In addition to myogenic cell populations, interaction with other cell types such as inflammatory cells, mesenchymal (fibroadipogenic progenitors—FAPs, pericytes) and vascular (endothelial) lineages are important for efficient muscle repair. While the role of the distinct populations involved in skeletal muscle regeneration is well characterized, the quantitative changes in the muscle stem cell and niche during the regeneration process remain poorly characterized. Methods: We have used mass cytometry to follow the main muscle cell types (muscle stem cells, vascular, mesenchymal and immune cell lineages) during early activation and over the course of muscle regeneration at D0, D2, D5 and D7 compared with uninjured muscles. Results: Early activation induces a number of rapid changes in the proteome of multiple cell types. Following the induction of damage, we observe a drastic loss of myogenic, vascular and mesenchymal cell lineages while immune cells invade the damaged tissue to clear debris and promote muscle repair. Immune cells constitute up to 80% of the mononuclear cells 5 days post-injury. We show that muscle stem cells are quickly activated in order to form new myofibers and reconstitute the quiescent muscle stem cell pool. In addition, our study provides a quantitative analysis of the various myogenic populations during muscle repair. Conclusions: We have developed a mass cytometry panel to investigate the dynamic nature of muscle regeneration at a single-cell level. Using our panel, we have identified early changes in the proteome of stressed satellite and niche cells. We have also quantified changes in the major cell types of skeletal muscle during regeneration and analyzed myogenic transcription factor expression in satellite cells throughout this process. Our results highlight the progressive dynamic shifts in cell populations and the distinct states of muscle stem cells adopted during skeletal muscle regeneration. Our findings give a deeper understanding of the cellular and molecular aspects of muscle regeneration.
Collapse
|
74
|
Muscle Diversity, Heterogeneity, and Gradients: Learning from Sarcoglycanopathies. Int J Mol Sci 2021; 22:ijms22052502. [PMID: 33801487 PMCID: PMC7958856 DOI: 10.3390/ijms22052502] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 02/25/2021] [Accepted: 02/26/2021] [Indexed: 12/25/2022] Open
Abstract
Skeletal muscle, the most abundant tissue in the body, is heterogeneous. This heterogeneity forms the basis of muscle diversity, which is reflected in the specialized functions of muscles in different parts of the body. However, these different parts are not always clearly delimitated, and this often gives rise to gradients within the same muscle and even across the body. During the last decade, several studies on muscular disorders both in mice and in humans have observed particular distribution patterns of muscle weakness during disease, indicating that the same mutation can affect muscles differently. Moreover, these phenotypical differences reveal gradients of severity, existing alongside other architectural gradients. These two factors are especially prominent in sarcoglycanopathies. Nevertheless, very little is known about the mechanism(s) driving the phenotypic diversity of the muscles affected by these diseases. Here, we will review the available literature on sarcoglycanopathies, focusing on phenotypic differences among affected muscles and gradients, characterization techniques, molecular signatures, and cell population heterogeneity, highlighting the possibilities opened up by new technologies. This review aims to revive research interest in the diverse disease phenotype affecting different muscles, in order to pave the way for new therapeutic interventions.
Collapse
|
75
|
Zhang Y, Lahmann I, Baum K, Shimojo H, Mourikis P, Wolf J, Kageyama R, Birchmeier C. Oscillations of Delta-like1 regulate the balance between differentiation and maintenance of muscle stem cells. Nat Commun 2021; 12:1318. [PMID: 33637744 PMCID: PMC7910593 DOI: 10.1038/s41467-021-21631-4] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Accepted: 02/04/2021] [Indexed: 02/07/2023] Open
Abstract
Cell-cell interactions mediated by Notch are critical for the maintenance of skeletal muscle stem cells. However, dynamics, cellular source and identity of functional Notch ligands during expansion of the stem cell pool in muscle growth and regeneration remain poorly characterized. Here we demonstrate that oscillating Delta-like 1 (Dll1) produced by myogenic cells is an indispensable Notch ligand for self-renewal of muscle stem cells in mice. Dll1 expression is controlled by the Notch target Hes1 and the muscle regulatory factor MyoD. Consistent with our mathematical model, our experimental analyses show that Hes1 acts as the oscillatory pacemaker, whereas MyoD regulates robust Dll1 expression. Interfering with Dll1 oscillations without changing its overall expression level impairs self-renewal, resulting in premature differentiation of muscle stem cells during muscle growth and regeneration. We conclude that the oscillatory Dll1 input into Notch signaling ensures the equilibrium between self-renewal and differentiation in myogenic cell communities.
Collapse
Affiliation(s)
- Yao Zhang
- Developmental Biology/Signal Transduction, Max-Delbrück-Center for Molecular Medicine, Berlin, Germany.
| | - Ines Lahmann
- Developmental Biology/Signal Transduction, Max-Delbrück-Center for Molecular Medicine, Berlin, Germany
| | - Katharina Baum
- Mathematical Modelling of Cellular Processes, Max-Delbrück-Center for Molecular Medicine, Berlin, Germany
- Hasso Plattner Institute, Digital Engineering Faculty, University of Potsdam, Potsdam, Germany
| | - Hiromi Shimojo
- Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto, Japan
- Graduate School of Frontier Biosciences, Osaka University, Osaka, Japan
| | | | - Jana Wolf
- Mathematical Modelling of Cellular Processes, Max-Delbrück-Center for Molecular Medicine, Berlin, Germany
- Department of Mathematics and Computer Science, Free University Berlin, Berlin, Germany
| | - Ryoichiro Kageyama
- Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto, Japan
| | - Carmen Birchmeier
- Developmental Biology/Signal Transduction, Max-Delbrück-Center for Molecular Medicine, Berlin, Germany.
- Neurowissenschaftliches Forschungzentrum, NeuroCure Cluster of Excellence, Charité-Universitätsmedizin Berlin, Berlin, Germany.
| |
Collapse
|
76
|
Machado L, Geara P, Camps J, Dos Santos M, Teixeira-Clerc F, Van Herck J, Varet H, Legendre R, Pawlotsky JM, Sampaolesi M, Voet T, Maire P, Relaix F, Mourikis P. Tissue damage induces a conserved stress response that initiates quiescent muscle stem cell activation. Cell Stem Cell 2021; 28:1125-1135.e7. [PMID: 33609440 DOI: 10.1016/j.stem.2021.01.017] [Citation(s) in RCA: 77] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Revised: 10/30/2020] [Accepted: 01/22/2021] [Indexed: 12/27/2022]
Abstract
Tissue damage dramatically alters how cells interact with their microenvironment. These changes in turn dictate cellular responses, such as stem cell activation, yet early cellular responses in vivo remain ill defined. We generated single-cell and nucleus atlases from intact, dissociated, and injured muscle and liver and identified a common stress response signature shared by multiple cell types across these organs. This prevalent stress response was detected in published datasets across a range of tissues, demonstrating high conservation but also a significant degree of data distortion in single-cell reference atlases. Using quiescent muscle stem cells as a paradigm of cell activation following injury, we captured early cell activation following muscle injury and found that an essential ERK1/2 primary proliferation signal precedes initiation of the Notch-regulated myogenic program. This study defines initial events in response to tissue perturbation and identifies a broadly conserved transcriptional stress response that acts in parallel with cell-specific adaptive alterations.
Collapse
Affiliation(s)
- Léo Machado
- Univ Paris Est Creteil, INSERM, IMRB, F-94010 Creteil, France
| | - Perla Geara
- Univ Paris Est Creteil, INSERM, IMRB, F-94010 Creteil, France
| | - Jordi Camps
- Laboratory of Translational Cardiomyology, Department of Development and Regeneration, Stem Cell Research Institute, KU Leuven, 3000 Leuven, Belgium; Bayer AG, 13353 Berlin, Germany
| | | | | | - Jens Van Herck
- Laboratory of Reproductive Genomics, Department of Human Genetics, KU Leuven, 3000 Leuven, Belgium
| | - Hugo Varet
- Hub de Bioinformatique et Biostatistique - Département Biologie Computationnelle, Institut Pasteur, USR 3756 CNRS, 75015 Paris, France; Plate-forme Biomics - Centre de Ressources et Recherches Technologiques (C2RT), Institut Pasteur, 75015 Paris, France
| | - Rachel Legendre
- Hub de Bioinformatique et Biostatistique - Département Biologie Computationnelle, Institut Pasteur, USR 3756 CNRS, 75015 Paris, France; Plate-forme Biomics - Centre de Ressources et Recherches Technologiques (C2RT), Institut Pasteur, 75015 Paris, France
| | - Jean-Michel Pawlotsky
- Univ Paris Est Creteil, INSERM, IMRB, F-94010 Creteil, France; Département de Virologie, Hôpital Henri Mondor, F-94010 Créteil, France
| | - Maurilio Sampaolesi
- Laboratory of Translational Cardiomyology, Department of Development and Regeneration, Stem Cell Research Institute, KU Leuven, 3000 Leuven, Belgium; Human Anatomy Unit, Department of Public Health, Experimental and Forensic Medicine, University of Pavia, 27100 Pavia, Italy
| | - Thierry Voet
- Laboratory of Reproductive Genomics, Department of Human Genetics, KU Leuven, 3000 Leuven, Belgium; Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge CB10 1SA, UK
| | - Pascal Maire
- Université de Paris, Institut Cochin, INSERM, CNRS, 75014 Paris, France
| | - Frederic Relaix
- Univ Paris Est Creteil, INSERM, IMRB, F-94010 Creteil, France; EnvA, IMRB, 94700 Maisons-Alfort, France; EFS, IMRB, 94010 Creteil, France; AP-HP, Hopital Mondor, Service d'histologie, F-94010 Creteil, France.
| | | |
Collapse
|
77
|
Benavente-Diaz M, Comai G, Di Girolamo D, Langa F, Tajbakhsh S. Dynamics of myogenic differentiation using a novel Myogenin knock-in reporter mouse. Skelet Muscle 2021; 11:5. [PMID: 33602287 PMCID: PMC7890983 DOI: 10.1186/s13395-021-00260-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Accepted: 01/06/2021] [Indexed: 12/14/2022] Open
Abstract
Background Myogenin is a transcription factor that is expressed during terminal myoblast differentiation in embryonic development and adult muscle regeneration. Investigation of this cell state transition has been hampered by the lack of a sensitive reporter to dynamically track cells during differentiation. Results Here, we report a knock-in mouse line expressing the tdTOMATO fluorescent protein from the endogenous Myogenin locus. Expression of tdTOMATO in MyogntdTom mice recapitulated endogenous Myogenin expression during embryonic muscle formation and adult regeneration and enabled the isolation of the MYOGENIN+ cell population. We also show that tdTOMATO fluorescence allows tracking of differentiating myoblasts in vitro and by intravital imaging in vivo. Lastly, we monitored by live imaging the cell division dynamics of differentiating myoblasts in vitro and showed that a fraction of the MYOGENIN+ population can undergo one round of cell division, albeit at a much lower frequency than MYOGENIN− myoblasts. Conclusions We expect that this reporter mouse will be a valuable resource for researchers investigating skeletal muscle biology in developmental and adult contexts. Supplementary Information The online version contains supplementary material available at 10.1186/s13395-021-00260-x.
Collapse
Affiliation(s)
- Maria Benavente-Diaz
- Stem Cells & Development Unit, Institut Pasteur, 25 rue du Dr. Roux, 75015, Paris, France.,UMR CNRS 3738, Institut Pasteur, Paris, France.,Sorbonne Universités, Complexité du Vivant, F-75005, Paris, France
| | - Glenda Comai
- Stem Cells & Development Unit, Institut Pasteur, 25 rue du Dr. Roux, 75015, Paris, France.,UMR CNRS 3738, Institut Pasteur, Paris, France
| | - Daniela Di Girolamo
- Stem Cells & Development Unit, Institut Pasteur, 25 rue du Dr. Roux, 75015, Paris, France.,UMR CNRS 3738, Institut Pasteur, Paris, France
| | - Francina Langa
- Mouse Genetics Engineering Center, Institut Pasteur, Paris, France
| | - Shahragim Tajbakhsh
- Stem Cells & Development Unit, Institut Pasteur, 25 rue du Dr. Roux, 75015, Paris, France. .,UMR CNRS 3738, Institut Pasteur, Paris, France.
| |
Collapse
|
78
|
Hoh JFY. Myosin heavy chains in extraocular muscle fibres: Distribution, regulation and function. Acta Physiol (Oxf) 2021; 231:e13535. [PMID: 32640094 DOI: 10.1111/apha.13535] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2020] [Accepted: 07/02/2020] [Indexed: 12/13/2022]
Abstract
This review examines kinetic properties and distribution of the 11 isoforms of myosin heavy chain (MyHC) expressed in extraocular muscle (EOM) fibre types and the regulation and function of these MyHCs. Although recruitment and discharge characteristics of ocular motoneurons during fixation and eye movements are well documented, work directly linking these properties with motor unit contractile speed and MyHC composition is lacking. Recruitment of motor units according to Henneman's size principle has some support in EOMs but needs consolidation. Both neurogenic and myogenic mechanisms regulate MyHC expression as in other muscle allotypes. Developmentally, multiply-innervated (MIFs) and singly-innervated fibres (SIFs) are derived presumably from distinct myoblast lineages, ending up expressing MyHCs in the slow and fast ends of the kinetic spectrum respectively. They modulate the synaptic inputs of their motoneurons through different retrogradely transported neurotrophins, thereby specifying their tonic and phasic impulse patterns. Immunohistochemical analyses of EOMs regenerating in situ and in limb muscle beds suggest that the very impulse patterns driving various ocular movements equip effectors with appropriate MyHC compositions and speeds to accomplish their tasks. These experiments also suggest that satellite cells of SIFs and MIFs are distinct lineages expressing different MyHCs during regeneration. MyHC compositions and functional characteristics of orbital fibres show longitudinal variations that facilitate linear ocular rotation during saccades. Palisade endings on global MIFs are postulated to respond to active and passive tensions by triggering axon reflexes that play important roles during fixation, saccades and vergence. How EOMs implement Listings law during ocular rotation is discussed.
Collapse
Affiliation(s)
- Joseph F. Y. Hoh
- Discipline of Physiology and the Bosch Institute School of Medical Sciences Faculty of Medicine and Health The University of Sydney Sydney NSW Australia
| |
Collapse
|
79
|
Yoshioka K, Kitajima Y, Seko D, Tsuchiya Y, Ono Y. The body region specificity in murine models of muscle regeneration and atrophy. Acta Physiol (Oxf) 2021; 231:e13553. [PMID: 32875719 PMCID: PMC7757168 DOI: 10.1111/apha.13553] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Revised: 08/15/2020] [Accepted: 08/26/2020] [Indexed: 01/06/2023]
Abstract
AIM Skeletal muscles are distributed throughout the body, presenting a variety of sizes, shapes and functions. Here, we examined whether muscle regeneration and atrophy occurred homogeneously throughout the body in mouse models. METHODS Acute muscle regeneration was induced by a single intramuscular injection of cardiotoxin in adult mice. Chronic muscle regeneration was assessed in mdx mice. Muscle atrophy in different muscles was evaluated by cancer cachexia, ageing and castration mouse models. RESULTS We found that, in the cardiotoxin-injected acute muscle injury model, head muscles slowly regenerated, while limb muscles exhibited a rapid regeneration and even overgrowth. This overgrowth was also observed in limb muscles alone (but not in head muscles) in mdx mice as chronic injury models. We described the body region-specific decline in the muscle mass in muscle atrophy models: cancer cachexia-induced, aged and castrated mice. The positional identities, including gene expression profiles and hormone sensitivity, were robustly preserved in the ectopically engrafted satellite cell-derived muscles in the castrated model. CONCLUSION Our results indicate that positional identities in muscles should be considered for the development of efficient regenerative therapies for muscle weakness, such as muscular dystrophy and age-related sarcopenia.
Collapse
Affiliation(s)
- Kiyoshi Yoshioka
- Department of Muscle Development and Regeneration Institute of Molecular Embryology and Genetics Kumamoto University Kumamoto Japan
| | - Yasuo Kitajima
- Department of Muscle Development and Regeneration Institute of Molecular Embryology and Genetics Kumamoto University Kumamoto Japan
| | - Daiki Seko
- Department of Muscle Development and Regeneration Institute of Molecular Embryology and Genetics Kumamoto University Kumamoto Japan
| | - Yoshifumi Tsuchiya
- Department of Muscle Development and Regeneration Institute of Molecular Embryology and Genetics Kumamoto University Kumamoto Japan
| | - Yusuke Ono
- Department of Muscle Development and Regeneration Institute of Molecular Embryology and Genetics Kumamoto University Kumamoto Japan
| |
Collapse
|
80
|
Yahya I, Morosan-Puopolo G, Brand-Saberi B. The CXCR4/SDF-1 Axis in the Development of Facial Expression and Non-somitic Neck Muscles. Front Cell Dev Biol 2020; 8:615264. [PMID: 33415110 PMCID: PMC7783292 DOI: 10.3389/fcell.2020.615264] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Accepted: 12/04/2020] [Indexed: 12/26/2022] Open
Abstract
Trunk and head muscles originate from distinct embryonic regions: while the trunk muscles derive from the paraxial mesoderm that becomes segmented into somites, the majority of head muscles develops from the unsegmented cranial paraxial mesoderm. Differences in the molecular control of trunk versus head and neck muscles have been discovered about 25 years ago; interestingly, differences in satellite cell subpopulations were also described more recently. Specifically, the satellite cells of the facial expression muscles share properties with heart muscle. In adult vertebrates, neck muscles span the transition zone between head and trunk. Mastication and facial expression muscles derive from the mesodermal progenitor cells that are located in the first and second branchial arches, respectively. The cucullaris muscle (non-somitic neck muscle) originates from the posterior-most branchial arches. Like other subclasses within the chemokines and chemokine receptors, CXCR4 and SDF-1 play essential roles in the migration of cells within a number of various tissues during development. CXCR4 as receptor together with its ligand SDF-1 have mainly been described to regulate the migration of the trunk muscle progenitor cells. This review first underlines our recent understanding of the development of the facial expression (second arch-derived) muscles, focusing on new insights into the migration event and how this embryonic process is different from the development of mastication (first arch-derived) muscles. Other muscles associated with the head, such as non-somitic neck muscles derived from muscle progenitor cells located in the posterior branchial arches, are also in the focus of this review. Implications on human muscle dystrophies affecting the muscles of face and neck are also discussed.
Collapse
Affiliation(s)
- Imadeldin Yahya
- Department of Anatomy and Molecular Embryology, Ruhr University Bochum, Bochum, Germany.,Department of Anatomy, Faculty of Veterinary Medicine, University of Khartoum, Khartoum, Sudan
| | | | - Beate Brand-Saberi
- Department of Anatomy and Molecular Embryology, Ruhr University Bochum, Bochum, Germany
| |
Collapse
|
81
|
Comai GE, Tesařová M, Dupé V, Rhinn M, Vallecillo-García P, da Silva F, Feret B, Exelby K, Dollé P, Carlsson L, Pryce B, Spitz F, Stricker S, Zikmund T, Kaiser J, Briscoe J, Schedl A, Ghyselinck NB, Schweitzer R, Tajbakhsh S. Local retinoic acid signaling directs emergence of the extraocular muscle functional unit. PLoS Biol 2020; 18:e3000902. [PMID: 33201874 PMCID: PMC7707851 DOI: 10.1371/journal.pbio.3000902] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Revised: 12/01/2020] [Accepted: 10/01/2020] [Indexed: 12/20/2022] Open
Abstract
Coordinated development of muscles, tendons, and their attachment sites ensures emergence of functional musculoskeletal units that are adapted to diverse anatomical demands among different species. How these different tissues are patterned and functionally assembled during embryogenesis is poorly understood. Here, we investigated the morphogenesis of extraocular muscles (EOMs), an evolutionary conserved cranial muscle group that is crucial for the coordinated movement of the eyeballs and for visual acuity. By means of lineage analysis, we redefined the cellular origins of periocular connective tissues interacting with the EOMs, which do not arise exclusively from neural crest mesenchyme as previously thought. Using 3D imaging approaches, we established an integrative blueprint for the EOM functional unit. By doing so, we identified a developmental time window in which individual EOMs emerge from a unique muscle anlage and establish insertions in the sclera, which sets these muscles apart from classical muscle-to-bone type of insertions. Further, we demonstrate that the eyeballs are a source of diffusible all-trans retinoic acid (ATRA) that allow their targeting by the EOMs in a temporal and dose-dependent manner. Using genetically modified mice and inhibitor treatments, we find that endogenous local variations in the concentration of retinoids contribute to the establishment of tendon condensations and attachment sites that precede the initiation of muscle patterning. Collectively, our results highlight how global and site-specific programs are deployed for the assembly of muscle functional units with precise definition of muscle shapes and topographical wiring of their tendon attachments.
Collapse
Affiliation(s)
- Glenda Evangelina Comai
- Stem Cells & Development Unit, Institut Pasteur, Paris, France
- CNRS UMR 3738, Institut Pasteur, Paris, France
- * E-mail: (GEC); (ST)
| | - Markéta Tesařová
- Central European Institute of Technology, Brno University of Technology, Brno, Czech Republic
| | - Valérie Dupé
- Université de Rennes, CNRS, IGDR, Rennes, France
| | - Muriel Rhinn
- IGBMC-Institut de Génétique et de Biologie Moleculaire et Cellulaire, Illkirch, France
| | | | - Fabio da Silva
- Université Côte d'Azur, INSERM, CNRS, iBV, Nice, France
- Division of Molecular Embryology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Betty Feret
- IGBMC-Institut de Génétique et de Biologie Moleculaire et Cellulaire, Illkirch, France
| | | | - Pascal Dollé
- IGBMC-Institut de Génétique et de Biologie Moleculaire et Cellulaire, Illkirch, France
| | - Leif Carlsson
- Umeå Center for Molecular Medicine, Umeå University, Umeå, Sweden
| | - Brian Pryce
- Research Division, Shriners Hospital for Children, Portland, United States of America
| | - François Spitz
- Genomics of Animal Development Unit, Institut Pasteur, Paris, France
| | - Sigmar Stricker
- Institute for Chemistry and Biochemistry, Freie Universität Berlin, Berlin, Germany
| | - Tomáš Zikmund
- Central European Institute of Technology, Brno University of Technology, Brno, Czech Republic
| | - Jozef Kaiser
- Central European Institute of Technology, Brno University of Technology, Brno, Czech Republic
| | | | | | - Norbert B. Ghyselinck
- IGBMC-Institut de Génétique et de Biologie Moleculaire et Cellulaire, Illkirch, France
| | - Ronen Schweitzer
- Research Division, Shriners Hospital for Children, Portland, United States of America
| | - Shahragim Tajbakhsh
- Stem Cells & Development Unit, Institut Pasteur, Paris, France
- CNRS UMR 3738, Institut Pasteur, Paris, France
- * E-mail: (GEC); (ST)
| |
Collapse
|
82
|
García-Prat L, Perdiguero E, Alonso-Martín S, Dell'Orso S, Ravichandran S, Brooks SR, Juan AH, Campanario S, Jiang K, Hong X, Ortet L, Ruiz-Bonilla V, Flández M, Moiseeva V, Rebollo E, Jardí M, Sun HW, Musarò A, Sandri M, Del Sol A, Sartorelli V, Muñoz-Cánoves P. FoxO maintains a genuine muscle stem-cell quiescent state until geriatric age. Nat Cell Biol 2020; 22:1307-1318. [PMID: 33106654 DOI: 10.1038/s41556-020-00593-7] [Citation(s) in RCA: 91] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Accepted: 09/15/2020] [Indexed: 02/07/2023]
Abstract
Tissue regeneration declines with ageing but little is known about whether this arises from changes in stem-cell heterogeneity. Here, in homeostatic skeletal muscle, we identify two quiescent stem-cell states distinguished by relative CD34 expression: CD34High, with stemness properties (genuine state), and CD34Low, committed to myogenic differentiation (primed state). The genuine-quiescent state is unexpectedly preserved into later life, succumbing only in extreme old age due to the acquisition of primed-state traits. Niche-derived IGF1-dependent Akt activation debilitates the genuine stem-cell state by imposing primed-state features via FoxO inhibition. Interventions to neutralize Akt and promote FoxO activity drive a primed-to-genuine state conversion, whereas FoxO inactivation deteriorates the genuine state at a young age, causing regenerative failure of muscle, as occurs in geriatric mice. These findings reveal transcriptional determinants of stem-cell heterogeneity that resist ageing more than previously anticipated and are only lost in extreme old age, with implications for the repair of geriatric muscle.
Collapse
Affiliation(s)
- Laura García-Prat
- Department of Experimental and Health Sciences, Pompeu Fabra University (UPF), CIBER on Neurodegenerative Diseases (CIBERNED), Barcelona, Spain.,Spanish National Center on Cardiovascular Research (CNIC), Madrid, Spain.,Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Eusebio Perdiguero
- Department of Experimental and Health Sciences, Pompeu Fabra University (UPF), CIBER on Neurodegenerative Diseases (CIBERNED), Barcelona, Spain
| | - Sonia Alonso-Martín
- Spanish National Center on Cardiovascular Research (CNIC), Madrid, Spain.,Neurosciences Area, Biodonostia Health Research Institute, Donostia-San Sebastián, San Sebastián, Spain
| | - Stefania Dell'Orso
- Laboratory of Muscle Stem Cells and Gene Regulation, National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS), NIH Bethesda, Bethesda, MD, USA
| | - Srikanth Ravichandran
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Stephen R Brooks
- Biodata Mining and Discovery Section, NIAMS, NIH Bethesda, Bethesda, MD, USA
| | - Aster H Juan
- Laboratory of Muscle Stem Cells and Gene Regulation, National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS), NIH Bethesda, Bethesda, MD, USA
| | - Silvia Campanario
- Department of Experimental and Health Sciences, Pompeu Fabra University (UPF), CIBER on Neurodegenerative Diseases (CIBERNED), Barcelona, Spain.,Spanish National Center on Cardiovascular Research (CNIC), Madrid, Spain
| | - Kan Jiang
- Biodata Mining and Discovery Section, NIAMS, NIH Bethesda, Bethesda, MD, USA
| | - Xiaotong Hong
- Spanish National Center on Cardiovascular Research (CNIC), Madrid, Spain
| | - Laura Ortet
- Department of Experimental and Health Sciences, Pompeu Fabra University (UPF), CIBER on Neurodegenerative Diseases (CIBERNED), Barcelona, Spain
| | - Vanessa Ruiz-Bonilla
- Department of Experimental and Health Sciences, Pompeu Fabra University (UPF), CIBER on Neurodegenerative Diseases (CIBERNED), Barcelona, Spain
| | - Marta Flández
- Spanish National Center on Cardiovascular Research (CNIC), Madrid, Spain.,Grupo de Investigación en Oncología Clínico Traslacional, Instituto de Investigación Hospital 12 de Octubre, Madrid, Spain
| | - Victoria Moiseeva
- Department of Experimental and Health Sciences, Pompeu Fabra University (UPF), CIBER on Neurodegenerative Diseases (CIBERNED), Barcelona, Spain
| | - Elena Rebollo
- Molecular Imaging Platform, Molecular Biology Institute of Barcelona (IBMB-CSIC), Barcelona, Spain
| | - Mercè Jardí
- Department of Experimental and Health Sciences, Pompeu Fabra University (UPF), CIBER on Neurodegenerative Diseases (CIBERNED), Barcelona, Spain
| | - Hong-Wei Sun
- Biodata Mining and Discovery Section, NIAMS, NIH Bethesda, Bethesda, MD, USA
| | - Antonio Musarò
- DAHFMO-Unit of Histology and Medical Embryology, Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Sapienza University of Rome, Rome, Italy
| | - Marco Sandri
- Department of Biomedical Science, University of Padova, Padova, Italy
| | - Antonio Del Sol
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg.,CIC bioGUNE, Derio, Spain.,IKERBASQUE, Basque Foundation for Science, Bilbao, Spain
| | - Vittorio Sartorelli
- Laboratory of Muscle Stem Cells and Gene Regulation, National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS), NIH Bethesda, Bethesda, MD, USA.
| | - Pura Muñoz-Cánoves
- Department of Experimental and Health Sciences, Pompeu Fabra University (UPF), CIBER on Neurodegenerative Diseases (CIBERNED), Barcelona, Spain. .,Spanish National Center on Cardiovascular Research (CNIC), Madrid, Spain. .,ICREA, Barcelona, Spain.
| |
Collapse
|
83
|
Yue L, Cheung TH. Protocol for Isolation and Characterization of In Situ Fixed Quiescent Muscle Stem Cells. STAR Protoc 2020; 1:100128. [PMID: 33377022 PMCID: PMC7757111 DOI: 10.1016/j.xpro.2020.100128] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Quiescent muscle stem cells, also called satellite cells (SCs), are essential for muscle regeneration. However, quiescent SCs are quickly activated during fluorescence-activated cell sorting (FACS) isolation. Here, we present an optimized protocol to isolate quiescent muscle stem cells from fixative-perfused mice and generate high-quality cDNA libraries for RNA-sequencing analysis. Fixation preserves the signatures of quiescent muscle stem cells in vivo. Isolated SCs can be used for downstream analysis such as immunofluorescence, RNA sequencing, and mass spectrometry. For complete information on the use and execution of this protocol, please refer to Yue et al. (2020). An optimized protocol for quiescent muscle stem cell isolation Use of fixative perfusion to preserve the gene expression signature in vivo Adaptable for various downstream analyses (e.g., transcriptome, proteome) Fixative perfusion extendable to the isolation of other cell types from tissues in vivo
Collapse
Affiliation(s)
- Lu Yue
- Division of Life Science, Center for Stem Cell Research, Center of Systems Biology and Human Health, State Key Laboratory of Molecular Neuroscience, and Molecular Neuroscience Center, Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China
| | - Tom H Cheung
- Division of Life Science, Center for Stem Cell Research, Center of Systems Biology and Human Health, State Key Laboratory of Molecular Neuroscience, and Molecular Neuroscience Center, Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China.,Hong Kong Center for Neurodegenerative Diseases, Hong Kong Science Park, Hong Kong, China.,Guangdong Provincial Key Laboratory of Brain Science, Disease and Drug Development, HKUST Shenzhen Research Institute, Shenzhen-Hong Kong Institute of Brain Science, Shenzhen, Guangdong 518057, China
| |
Collapse
|
84
|
Wurmser M, Chaverot N, Madani R, Sakai H, Negroni E, Demignon J, Saint-Pierre B, Mouly V, Amthor H, Tapscott S, Birchmeier C, Tajbakhsh S, Le Grand F, Sotiropoulos A, Maire P. SIX1 and SIX4 homeoproteins regulate PAX7+ progenitor cell properties during fetal epaxial myogenesis. Development 2020; 147:dev.185975. [PMID: 32591430 DOI: 10.1242/dev.185975] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Accepted: 06/18/2020] [Indexed: 01/09/2023]
Abstract
Pax7 expression marks stem cells in developing skeletal muscles and adult satellite cells during homeostasis and muscle regeneration. The genetic determinants that control the entrance into the myogenic program and the appearance of PAX7+ cells during embryogenesis are poorly understood. SIX homeoproteins are encoded by the sine oculis-related homeobox Six1-Six6 genes in vertebrates. Six1, Six2, Six4 and Six5 are expressed in the muscle lineage. Here, we tested the hypothesis that Six1 and Six4 could participate in the genesis of myogenic stem cells. We show that fewer PAX7+ cells occupy a satellite cell position between the myofiber and its associated basal lamina in Six1 and Six4 knockout mice (s1s4KO) at E18. However, PAX7+ cells are detected in remaining muscle masses present in the epaxial region of the double mutant embryos and are able to divide and contribute to muscle growth. To further characterize the properties of s1s4KO PAX7+ cells, we analyzed their transcriptome and tested their properties after transplantation in adult regenerating tibialis anterior muscle. Mutant stem cells contribute to hypotrophic myofibers that are not innervated but retain the ability to self-renew.
Collapse
Affiliation(s)
- Maud Wurmser
- Université de Paris, Institut Cochin, INSERM, CNRS, 24 rue du Fg St Jacques, F-75014 Paris, France
| | - Nathalie Chaverot
- Université de Paris, Institut Cochin, INSERM, CNRS, 24 rue du Fg St Jacques, F-75014 Paris, France
| | - Rouba Madani
- Université de Paris, Institut Cochin, INSERM, CNRS, 24 rue du Fg St Jacques, F-75014 Paris, France
| | - Hiroshi Sakai
- Division of Integrative Pathophysiology, Proteo-Science Center, Ehime University, Toon, Ehime, 791-0295, Japan.,Stem Cells and Development, Department of Developmental and Stem Cell Biology, Institut Pasteur, 25 rue du Dr. Roux, 75015, Paris, France.,CNRS UMR 3738, Institut Pasteur, 75015 Paris, France
| | - Elisa Negroni
- Sorbonne Université, Institut de Myologie, INSERM, 75013 Paris, France
| | - Josiane Demignon
- Université de Paris, Institut Cochin, INSERM, CNRS, 24 rue du Fg St Jacques, F-75014 Paris, France
| | - Benjamin Saint-Pierre
- Université de Paris, Institut Cochin, INSERM, CNRS, 24 rue du Fg St Jacques, F-75014 Paris, France
| | - Vincent Mouly
- Sorbonne Université, Institut de Myologie, INSERM, 75013 Paris, France
| | - Helge Amthor
- INSERM U1179, LIA BAHN CSM, Université de Versailles Saint-Quentin-en-Yvelines, Montigny-le-Bretonneux, France
| | | | | | - Shahragim Tajbakhsh
- Stem Cells and Development, Department of Developmental and Stem Cell Biology, Institut Pasteur, 25 rue du Dr. Roux, 75015, Paris, France.,CNRS UMR 3738, Institut Pasteur, 75015 Paris, France
| | - Fabien Le Grand
- Université de Paris, Institut Cochin, INSERM, CNRS, 24 rue du Fg St Jacques, F-75014 Paris, France.,Institut NeuroMyoGène, Université Claude Bernard Lyon 1, CNRS, INSERM, 69008 Lyon, France
| | - Athanassia Sotiropoulos
- Université de Paris, Institut Cochin, INSERM, CNRS, 24 rue du Fg St Jacques, F-75014 Paris, France
| | - Pascal Maire
- Université de Paris, Institut Cochin, INSERM, CNRS, 24 rue du Fg St Jacques, F-75014 Paris, France
| |
Collapse
|
85
|
Buchanan SM, Price FD, Castiglioni A, Gee AW, Schneider J, Matyas MN, Hayhurst M, Tabebordbar M, Wagers AJ, Rubin LL. Pro-myogenic small molecules revealed by a chemical screen on primary muscle stem cells. Skelet Muscle 2020; 10:28. [PMID: 33036659 PMCID: PMC7547525 DOI: 10.1186/s13395-020-00248-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Accepted: 09/14/2020] [Indexed: 11/10/2022] Open
Abstract
Satellite cells are the canonical muscle stem cells that regenerate damaged skeletal muscle. Loss of function of these cells has been linked to reduced muscle repair capacity and compromised muscle health in acute muscle injury and congenital neuromuscular diseases. To identify new pathways that can prevent loss of skeletal muscle function or enhance regenerative potential, we established an imaging-based screen capable of identifying small molecules that promote the expansion of freshly isolated satellite cells. We found several classes of receptor tyrosine kinase (RTK) inhibitors that increased freshly isolated satellite cell numbers in vitro. Further exploration of one of these compounds, the RTK inhibitor CEP-701 (also known as lestaurtinib), revealed potent activity on mouse satellite cells both in vitro and in vivo. This expansion potential was not seen upon exposure of proliferating committed myoblasts or non-myogenic fibroblasts to CEP-701. When delivered subcutaneously to acutely injured animals, CEP-701 increased both the total number of satellite cells and the rate of muscle repair, as revealed by an increased cross-sectional area of regenerating fibers. Moreover, freshly isolated satellite cells expanded ex vivo in the presence of CEP-701 displayed enhanced muscle engraftment potential upon in vivo transplantation. We provide compelling evidence that certain RTKs, and in particular RET, regulate satellite cell expansion during muscle regeneration. This study demonstrates the power of small molecule screens of even rare adult stem cell populations for identifying stem cell-targeting compounds with therapeutic potential.
Collapse
Affiliation(s)
- Sean M Buchanan
- Harvard University Department of Stem Cell and Regenerative Biology, 7 Divinity Ave, Cambridge, MA, 02138, USA
| | - Feodor D Price
- Harvard University Department of Stem Cell and Regenerative Biology, 7 Divinity Ave, Cambridge, MA, 02138, USA
| | - Alessandra Castiglioni
- Harvard University Department of Stem Cell and Regenerative Biology, 7 Divinity Ave, Cambridge, MA, 02138, USA.,Cancer Immunology Department, Genentech, 1 DNA Way, South San Francisco, CA, 94080, USA
| | - Amanda Wagner Gee
- Harvard University Department of Stem Cell and Regenerative Biology, 7 Divinity Ave, Cambridge, MA, 02138, USA
| | - Joel Schneider
- Harvard University Department of Stem Cell and Regenerative Biology, 7 Divinity Ave, Cambridge, MA, 02138, USA
| | - Mark N Matyas
- Harvard University Department of Stem Cell and Regenerative Biology, 7 Divinity Ave, Cambridge, MA, 02138, USA
| | - Monica Hayhurst
- Harvard University Department of Stem Cell and Regenerative Biology, 7 Divinity Ave, Cambridge, MA, 02138, USA
| | - Mohammadsharif Tabebordbar
- Harvard University Department of Stem Cell and Regenerative Biology, 7 Divinity Ave, Cambridge, MA, 02138, USA
| | - Amy J Wagers
- Harvard University Department of Stem Cell and Regenerative Biology, 7 Divinity Ave, Cambridge, MA, 02138, USA
| | - Lee L Rubin
- Harvard University Department of Stem Cell and Regenerative Biology, 7 Divinity Ave, Cambridge, MA, 02138, USA.
| |
Collapse
|
86
|
Evano B, Gill D, Hernando-Herraez I, Comai G, Stubbs TM, Commere PH, Reik W, Tajbakhsh S. Transcriptome and epigenome diversity and plasticity of muscle stem cells following transplantation. PLoS Genet 2020; 16:e1009022. [PMID: 33125370 PMCID: PMC7657492 DOI: 10.1371/journal.pgen.1009022] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2020] [Revised: 11/11/2020] [Accepted: 08/02/2020] [Indexed: 12/14/2022] Open
Abstract
Adult skeletal muscles are maintained during homeostasis and regenerated upon injury by muscle stem cells (MuSCs). A heterogeneity in self-renewal, differentiation and regeneration properties has been reported for MuSCs based on their anatomical location. Although MuSCs derived from extraocular muscles (EOM) have a higher regenerative capacity than those derived from limb muscles, the molecular determinants that govern these differences remain undefined. Here we show that EOM and limb MuSCs have distinct DNA methylation signatures associated with enhancers of location-specific genes, and that the EOM transcriptome is reprogrammed following transplantation into a limb muscle environment. Notably, EOM MuSCs expressed host-site specific positional Hox codes after engraftment and self-renewal within the host muscle. However, about 10% of EOM-specific genes showed engraftment-resistant expression, pointing to cell-intrinsic molecular determinants of the higher engraftment potential of EOM MuSCs. Our results underscore the molecular diversity of distinct MuSC populations and molecularly define their plasticity in response to microenvironmental cues. These findings provide insights into strategies designed to improve the functional capacity of MuSCs in the context of regenerative medicine.
Collapse
Affiliation(s)
- Brendan Evano
- Stem Cells & Development, Department of Developmental & Stem Cell Biology, Institut Pasteur, 25 rue du Dr. Roux, Paris, France
- CNRS UMR 3738, Institut Pasteur, Paris, France
| | - Diljeet Gill
- Epigenetics Programme, Babraham Institute, Cambridge, United Kingdom
| | | | - Glenda Comai
- Stem Cells & Development, Department of Developmental & Stem Cell Biology, Institut Pasteur, 25 rue du Dr. Roux, Paris, France
- CNRS UMR 3738, Institut Pasteur, Paris, France
| | - Thomas M. Stubbs
- Epigenetics Programme, Babraham Institute, Cambridge, United Kingdom
| | - Pierre-Henri Commere
- Cytometry and Biomarkers, Center for Technological Resources and Research, Institut Pasteur, 28 rue du Dr. Roux, Paris, France
| | - Wolf Reik
- Epigenetics Programme, Babraham Institute, Cambridge, United Kingdom
| | - Shahragim Tajbakhsh
- Stem Cells & Development, Department of Developmental & Stem Cell Biology, Institut Pasteur, 25 rue du Dr. Roux, Paris, France
- CNRS UMR 3738, Institut Pasteur, Paris, France
| |
Collapse
|
87
|
Stange K, Ahrens HE, von Maltzahn J, Röntgen M. Isolation and ex vivo cultivation of single myofibers from porcine muscle. In Vitro Cell Dev Biol Anim 2020; 56:585-592. [PMID: 32964376 PMCID: PMC7532130 DOI: 10.1007/s11626-020-00492-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Accepted: 08/06/2020] [Indexed: 11/27/2022]
Abstract
The isolation and cultivation of intact, single myofibers presents a superior approach for studying myogenic cells in their native position. The cells’ characteristics remain more similar to muscle tissue than in cell culture. Nevertheless, no routinely used method in higher vertebrates exists. Therefore, we aimed at establishing the isolation and cultivation of single myofibers from porcine muscle. For the first time, we implemented the isolation of intact myofibers from porcine fibularis tertius muscle by enzymatic digestion and their subsequent cultivation under floating conditions. Confocal microscopy showed intact myofibrill structures in isolated myofibers. Myogenic cells were able to proliferate at their parent myofiber as shown by the increase of myonuclear number during culture. Additionally, the described method can be used to investigate myogenic cells migrated from isolated myofibers. These cells expressed myogenic markers and were able to differentiate. In the future, our method can be used for genetic manipulation of cells at myofibers, investigation of growth factors or pharmacological substances, and determination of interactions between myofibers and associated cells. Working with isolated myofibers has the potential to bridge conventional cell culture and animal experiments. Adapting the method to porcine muscle allows for application possibilities in veterinary medicine as well as in biomedical research, which cannot be addressed in rodent model systems.
Collapse
Affiliation(s)
- Katja Stange
- Institute of Muscle Biology and Growth, Growth and Development Unit, Leibniz Institute for Farm Animal Biology (FBN), Wilhelm-Stahl-Allee 2, 18196, Dummerstorf, Germany
| | - Hellen Elisa Ahrens
- Research Group Stem Cells in Regeneration of Skeletal Muscle, Leibniz Institute on Aging, 07745, Jena, Germany
| | - Julia von Maltzahn
- Research Group Stem Cells in Regeneration of Skeletal Muscle, Leibniz Institute on Aging, 07745, Jena, Germany
| | - Monika Röntgen
- Institute of Muscle Biology and Growth, Growth and Development Unit, Leibniz Institute for Farm Animal Biology (FBN), Wilhelm-Stahl-Allee 2, 18196, Dummerstorf, Germany.
| |
Collapse
|
88
|
Swedlund B, Lescroart F. Cardiopharyngeal Progenitor Specification: Multiple Roads to the Heart and Head Muscles. Cold Spring Harb Perspect Biol 2020; 12:a036731. [PMID: 31818856 PMCID: PMC7397823 DOI: 10.1101/cshperspect.a036731] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
During embryonic development, the heart arises from various sources of undifferentiated mesodermal progenitors, with an additional contribution from ectodermal neural crest cells. Mesodermal cardiac progenitors are plastic and multipotent, but are nevertheless specified to a precise heart region and cell type very early during development. Recent findings have defined both this lineage plasticity and early commitment of cardiac progenitors, using a combination of single-cell and population analyses. In this review, we discuss several aspects of cardiac progenitor specification. We discuss their markers, fate potential in vitro and in vivo, early segregation and commitment, and also intrinsic and extrinsic cues regulating lineage restriction from multipotency to a specific cell type of the heart. Finally, we also discuss the subdivisions of the cardiopharyngeal field, and the shared origins of the heart with other mesodermal derivatives, including head and neck muscles.
Collapse
Affiliation(s)
- Benjamin Swedlund
- Laboratory of Stem Cells and Cancer, Université Libre de Bruxelles, 1070 Brussels, Belgium
| | | |
Collapse
|
89
|
Helmbacher F, Stricker S. Tissue cross talks governing limb muscle development and regeneration. Semin Cell Dev Biol 2020; 104:14-30. [PMID: 32517852 DOI: 10.1016/j.semcdb.2020.05.005] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Revised: 05/07/2020] [Accepted: 05/08/2020] [Indexed: 12/14/2022]
Abstract
For decades, limb development has been a paradigm of three-dimensional patterning. Moreover, as the limb muscles and the other tissues of the limb's musculoskeletal system arise from distinct developmental sources, it has been a prime example of integrative morphogenesis and cross-tissue communication. As the limbs grow, all components of the musculoskeletal system (muscles, tendons, connective tissue, nerves) coordinate their growth and differentiation, ultimately giving rise to a functional unit capable of executing elaborate movement. While the molecular mechanisms governing global three-dimensional patterning and formation of the skeletal structures of the limbs has been a matter of intense research, patterning of the soft tissues is less understood. Here, we review the development of limb muscles with an emphasis on their interaction with other tissue types and the instructive roles these tissues play. Furthermore, we discuss the role of adult correlates of these embryonic accessory tissues in muscle regeneration.
Collapse
Affiliation(s)
| | - Sigmar Stricker
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, 14195, Berlin, Germany.
| |
Collapse
|
90
|
Yue L, Wan R, Luan S, Zeng W, Cheung TH. Dek Modulates Global Intron Retention during Muscle Stem Cells Quiescence Exit. Dev Cell 2020; 53:661-676.e6. [DOI: 10.1016/j.devcel.2020.05.006] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Revised: 04/06/2020] [Accepted: 05/09/2020] [Indexed: 12/21/2022]
|
91
|
Long noncoding RNA SAM promotes myoblast proliferation through stabilizing Sugt1 and facilitating kinetochore assembly. Nat Commun 2020; 11:2725. [PMID: 32483152 PMCID: PMC7264179 DOI: 10.1038/s41467-020-16553-6] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Accepted: 04/30/2020] [Indexed: 01/01/2023] Open
Abstract
The functional study of lncRNAs in skeletal muscle satellite cells (SCs) remains at the infancy stage. Here we identify SAM (Sugt1 asssociated muscle) lncRNA that is enriched in the proliferating myoblasts. Global deletion of SAM has no overt effect on mice but impairs adult muscle regeneration following acute damage; it also exacerbates the chronic injury-induced dystrophic phenotype in mdx mice. Consistently, inducible deletion of SAM in SCs leads to deficiency in muscle regeneration. Further examination reveals that SAM loss results in a cell-autonomous defect in the proliferative expansion of myoblasts. Mechanistically, we find SAM interacts and stabilizes Sugt1, a co-chaperon protein key to kinetochore assembly during cell division. Loss of SAM or Sugt1 both disrupts kinetochore assembly in mitotic cells due to the mislocalization of two components: Dsn1 and Hec1. Altogether, our findings identify SAM as a regulator of SC proliferation through facilitating Sugt1 mediated kinetochore assembly during cell division. Long noncoding RNA SAM (Sugt1 associated muscle) is upregulated in the proliferating myoblast cells. Here the authors investigate SAM knockout mice and suggest that SAM binds and stabilizes Sugt1, a co-chaperone protein that regulates kinetochore assembly.
Collapse
|
92
|
Vyas B, Nandkishore N, Sambasivan R. Vertebrate cranial mesoderm: developmental trajectory and evolutionary origin. Cell Mol Life Sci 2020; 77:1933-1945. [PMID: 31722070 PMCID: PMC11105048 DOI: 10.1007/s00018-019-03373-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2019] [Revised: 10/29/2019] [Accepted: 11/05/2019] [Indexed: 02/06/2023]
Abstract
Vertebrate cranial mesoderm is a discrete developmental unit compared to the mesoderm below the developing neck. An extraordinary feature of the cranial mesoderm is that it includes a common progenitor pool contributing to the chambered heart and the craniofacial skeletal muscles. This striking developmental potential and the excitement it generated led to advances in our understanding of cranial mesoderm developmental mechanism. Remarkably, recent findings have begun to unravel the origin of its distinct developmental characteristics. Here, we take a detailed view of the ontogenetic trajectory of cranial mesoderm and its regulatory network. Based on the emerging evidence, we propose that cranial and posterior mesoderm diverge at the earliest step of the process that patterns the mesoderm germ layer along the anterior-posterior body axis. Further, we discuss the latest evidence and their impact on our current understanding of the evolutionary origin of cranial mesoderm. Overall, the review highlights the findings from contemporary research, which lays the foundation to probe the molecular basis of unique developmental potential and evolutionary origin of cranial mesoderm.
Collapse
Affiliation(s)
- Bhakti Vyas
- Institute for Stem Cell Biology and Regenerative Medicine, GKVK Campus, Bellary Road, Bengaluru, 560065, India
- Manipal Academy of Higher Education, Manipal, 576104, India
| | - Nitya Nandkishore
- Institute for Stem Cell Biology and Regenerative Medicine, GKVK Campus, Bellary Road, Bengaluru, 560065, India
- SASTRA University, Thirumalaisamudram, Thanjavur, 613401, India
| | - Ramkumar Sambasivan
- Indian Institute of Science Education and Research (IISER) Tirupati, Transit Campus, Karakambadi Road, Rami Reddy Nagar, Mangalam, Tirupati, Andhra Pradesh, 517507, India.
| |
Collapse
|
93
|
Kim E, Wu F, Wu X, Choo HJ. Generation of craniofacial myogenic progenitor cells from human induced pluripotent stem cells for skeletal muscle tissue regeneration. Biomaterials 2020; 248:119995. [PMID: 32283390 PMCID: PMC7232788 DOI: 10.1016/j.biomaterials.2020.119995] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Revised: 03/13/2020] [Accepted: 03/20/2020] [Indexed: 12/18/2022]
Abstract
Craniofacial skeletal muscle is composed of approximately 60 muscles, which have critical functions including food uptake, eye movements and facial expressions. Although craniofacial muscles have significantly different embryonic origin, most current skeletal muscle differentiation protocols using human induced pluripotent stem cells (iPSCs) are based on somite-derived limb and trunk muscle developmental pathways. Since the lack of a protocol for craniofacial muscles is a significant gap in the iPSC-derived muscle field, we have developed an optimized protocol to generate craniofacial myogenic precursor cells (cMPCs) from human iPSCs by mimicking key signaling pathways during craniofacial embryonic myogenesis. At each different stage, human iPSC-derived cMPCs mirror the transcription factor expression profiles seen in their counterparts during embryo development. After the bi-potential cranial pharyngeal mesoderm is established, cells are committed to cranial skeletal muscle lineages with inhibition of cardiac lineages and are purified by flow cytometry. Furthermore, identities of Ipsc-derived cMPCs are verified with human primary myoblasts from craniofacial muscles using RNA sequencing. These data suggest that our new method could provide not only in vitro research tools to study muscle specificity of muscular dystrophy but also abundant and reliable cellular resources for tissue engineering to support craniofacial reconstruction surgery.
Collapse
Affiliation(s)
- Eunhye Kim
- Department of Cell Biology, School of Medcine, Emory University, Atlanta, GA, 30322, USA
| | - Fang Wu
- Department of Cell Biology, School of Medcine, Emory University, Atlanta, GA, 30322, USA
| | - Xuewen Wu
- Department of Otolaryngology-Head and Neck Surgery, School of Medicine, Emory University, Atlanta, GA, 30322, USA; Department of Otolaryngology Head and Neck Surgery, Xiangya Hospital of Central South University, Changsha, Hunan, 410008, China
| | - Hyojung J Choo
- Department of Cell Biology, School of Medcine, Emory University, Atlanta, GA, 30322, USA.
| |
Collapse
|
94
|
Dynamics of Asymmetric and Symmetric Divisions of Muscle Stem Cells In Vivo and on Artificial Niches. Cell Rep 2020; 30:3195-3206.e7. [DOI: 10.1016/j.celrep.2020.01.097] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Revised: 11/26/2019] [Accepted: 01/28/2020] [Indexed: 12/13/2022] Open
|
95
|
Adachi N, Bilio M, Baldini A, Kelly RG. Cardiopharyngeal mesoderm origins of musculoskeletal and connective tissues in the mammalian pharynx. Development 2020; 147:147/3/dev185256. [PMID: 32014863 DOI: 10.1242/dev.185256] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Accepted: 12/16/2019] [Indexed: 12/14/2022]
Abstract
Cardiopharyngeal mesoderm (CPM) gives rise to muscles of the head and heart. Using genetic lineage analysis in mice, we show that CPM develops into a broad range of pharyngeal structures and cell types encompassing musculoskeletal and connective tissues. We demonstrate that CPM contributes to medial pharyngeal skeletal and connective tissues associated with both branchiomeric and somite-derived neck muscles. CPM and neural crest cells (NCC) make complementary mediolateral contributions to pharyngeal structures, in a distribution established in the early embryo. We further show that biallelic expression of the CPM regulatory gene Tbx1, haploinsufficient in 22q11.2 deletion syndrome patients, is required for the correct patterning of muscles with CPM-derived connective tissue. Our results suggest that CPM plays a patterning role during muscle development, similar to that of NCC during craniofacial myogenesis. The broad lineage contributions of CPM to pharyngeal structures provide new insights into congenital disorders and evolution of the mammalian pharynx.
Collapse
Affiliation(s)
- Noritaka Adachi
- Aix-Marseille Université, CNRS UMR 7288, IBDM, 13009 Marseille, France
| | - Marchesa Bilio
- CNR Institute of Genetics and Biophysics Adriano Buzzati-Traverso, Via Pietro Castellino 111, 80131 Naples, Italy
| | - Antonio Baldini
- CNR Institute of Genetics and Biophysics Adriano Buzzati-Traverso, Via Pietro Castellino 111, 80131 Naples, Italy.,Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Naples 80131, Italy
| | - Robert G Kelly
- Aix-Marseille Université, CNRS UMR 7288, IBDM, 13009 Marseille, France
| |
Collapse
|
96
|
Amorim NML, Kee A, Coster ACF, Lucas C, Bould S, Daniel S, Weir JM, Mellett NA, Barbour J, Meikle PJ, Cohn RJ, Turner N, Hardeman EC, Simar D. Irradiation impairs mitochondrial function and skeletal muscle oxidative capacity: significance for metabolic complications in cancer survivors. Metabolism 2020; 103:154025. [PMID: 31765667 DOI: 10.1016/j.metabol.2019.154025] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Revised: 11/19/2019] [Accepted: 11/21/2019] [Indexed: 11/16/2022]
Abstract
BACKGROUND Metabolic complications are highly prevalent in cancer survivors treated with irradiation but the underlying mechanisms remain unknown. METHODS Chow or high fat-fed C57Bl/6J mice were irradiated (6Gy) before investigating the impact on whole-body or skeletal muscle metabolism and profiling their lipidomic signature. Using a transgenic mouse model (Tg:Pax7-nGFP), we isolated muscle progenitor cells (satellite cells) and characterised their metabolic functions. We recruited childhood cancer survivors, grouped them based on the use of total body irradiation during their treatment and established their lipidomic profile. RESULTS In mice, irradiation delayed body weight gain and impaired fat pads and muscle weights. These changes were associated with impaired whole-body fat oxidation in chow-fed mice and altered ex vivo skeletal muscle fatty acid oxidation, potentially due to a reduction in oxidative fibres and reduced mitochondrial enzyme activity. Irradiation led to fasting hyperglycaemia and impaired glucose uptake in isolated skeletal muscles. Cultured satellite cells from irradiated mice showed decreased fatty acid oxidation and reduced glucose uptake, recapitulating the host metabolic phenotype. Irradiation resulted in a remodelling of lipid species in skeletal muscles, with the extensor digitorum longus muscle being particularly affected. A large number of lipid species were reduced, with several of these species showing a positive correlation with mitochondrial enzymes activity. In cancer survivors exposed to irradiation, we found a similar decrease in systemic levels of most lipid species, and lipid species that increased were positively correlated with insulin resistance (HOMA-IR). CONCLUSION Irradiation leads to long-term alterations in body composition, and lipid and carbohydrate metabolism in skeletal muscle, and affects muscle progenitor cells. Such changes result in persistent impairment of metabolic functions, providing a new mechanism for the increased prevalence of metabolic diseases reported in irradiated individuals. In this context, changes in the lipidomic signature in response to irradiation could be of diagnostic value.
Collapse
Affiliation(s)
- Nadia M L Amorim
- Cellular and Genetic Medicine Unit, School of Medical Sciences, UNSW Sydney, Sydney, Australia
| | - Anthony Kee
- Cellular and Genetic Medicine Unit, School of Medical Sciences, UNSW Sydney, Sydney, Australia
| | - Adelle C F Coster
- School of Mathematics and Statistics, UNSW Sydney, Sydney, Australia
| | - Christine Lucas
- Cellular and Genetic Medicine Unit, School of Medical Sciences, UNSW Sydney, Sydney, Australia
| | - Sarah Bould
- Cellular and Genetic Medicine Unit, School of Medical Sciences, UNSW Sydney, Sydney, Australia
| | - Sara Daniel
- Mechanisms of Disease and Translational Research, School of Medical Sciences, UNSW Sydney, Sydney, Australia
| | - Jacquelyn M Weir
- Metabolomics Laboratory, Baker IDI, Heart and Diabetes Institute, Melbourne, Australia
| | - Natalie A Mellett
- Metabolomics Laboratory, Baker IDI, Heart and Diabetes Institute, Melbourne, Australia
| | - Jayne Barbour
- Mitochondrial Bioenergetics Lab, Department of Pharmacology, School of Medical Sciences, UNSW Sydney, Sydney, Australia
| | - Peter J Meikle
- Metabolomics Laboratory, Baker IDI, Heart and Diabetes Institute, Melbourne, Australia
| | - Richard J Cohn
- School of Women's and Children's Health, UNSW Sydney, Randwick, Australia; Kids Cancer Centre, Sydney Children's Hospital Network, Randwick, Australia
| | - Nigel Turner
- Mitochondrial Bioenergetics Lab, Department of Pharmacology, School of Medical Sciences, UNSW Sydney, Sydney, Australia
| | - Edna C Hardeman
- Cellular and Genetic Medicine Unit, School of Medical Sciences, UNSW Sydney, Sydney, Australia.
| | - David Simar
- Mechanisms of Disease and Translational Research, School of Medical Sciences, UNSW Sydney, Sydney, Australia.
| |
Collapse
|
97
|
Tey SR, Robertson S, Lynch E, Suzuki M. Coding Cell Identity of Human Skeletal Muscle Progenitor Cells Using Cell Surface Markers: Current Status and Remaining Challenges for Characterization and Isolation. Front Cell Dev Biol 2019; 7:284. [PMID: 31828070 PMCID: PMC6890603 DOI: 10.3389/fcell.2019.00284] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Accepted: 11/01/2019] [Indexed: 12/12/2022] Open
Abstract
Skeletal muscle progenitor cells (SMPCs), also called myogenic progenitors, have been studied extensively in recent years because of their promising therapeutic potential to preserve and recover skeletal muscle mass and function in patients with cachexia, sarcopenia, and neuromuscular diseases. SMPCs can be utilized to investigate the mechanisms of natural and pathological myogenesis via in vitro modeling and in vivo experimentation. While various types of SMPCs are currently available from several sources, human pluripotent stem cells (PSCs) offer an efficient and cost-effective method to derive SMPCs. As human PSC-derived cells often display varying heterogeneity in cell types, cell enrichment using cell surface markers remains a critical step in current procedures to establish a pure population of SMPCs. Here we summarize the cell surface markers currently being used to detect human SMPCs, describing their potential application for characterizing, identifying and isolating human PSC-derived SMPCs. To date, several positive and negative markers have been used to enrich human SMPCs from differentiated PSCs by cell sorting. A careful analysis of current findings can broaden our understanding and reveal potential uses for these surface markers with SMPCs.
Collapse
Affiliation(s)
- Sin-Ruow Tey
- Department of Comparative Biosciences, University of Wisconsin, Madison, WI, United States
| | - Samantha Robertson
- Department of Comparative Biosciences, University of Wisconsin, Madison, WI, United States
| | - Eileen Lynch
- Department of Comparative Biosciences, University of Wisconsin, Madison, WI, United States
| | - Masatoshi Suzuki
- Department of Comparative Biosciences, University of Wisconsin, Madison, WI, United States.,The Stem Cell and Regenerative Medicine Center, University of Wisconsin, Madison, WI, United States
| |
Collapse
|
98
|
Penaloza JS, Pappas MP, Hagen HR, Xie N, Chan SSK. Single-cell RNA-seq analysis of Mesp1-induced skeletal myogenic development. Biochem Biophys Res Commun 2019; 520:284-290. [PMID: 31590918 DOI: 10.1016/j.bbrc.2019.09.140] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Accepted: 09/30/2019] [Indexed: 12/21/2022]
Abstract
The Mesp1 lineage contributes to cardiac, hematopoietic and skeletal myogenic development. Interestingly, muscle stem cells residing in craniofacial skeletal muscles primarily arise from Mesp1+ progenitors, but those in trunk and limb skeletal muscles do not. To gain insights into the difference between the head and trunk/limb muscle developmental processes, we studied Mesp1+ skeletal myogenic derivatives via single-cell RNA-seq and other strategies. Using a doxycycline-inducible Mesp1-expressing mouse embryonic stem cell line, we found that the development of Mesp1-induced skeletal myogenic progenitors can be characterized by dynamic expression of PDGFRα and VCAM1. Single-cell RNA-seq analysis further revealed the heterogeneous nature of these Mesp1+ derivatives, spanning pluripotent and mesodermal to mesenchymal and skeletal myogenic. We subsequently reconstructed the single-cell trajectories of these subpopulations. Our data thereby provide a cell fate projection of Mesp1-induced skeletal myogenesis.
Collapse
Affiliation(s)
| | | | | | - Ning Xie
- Department of Pediatrics, Minneapolis, MN, 55455, USA.
| | - Sunny S K Chan
- Department of Pediatrics, Minneapolis, MN, 55455, USA; Stem Cell Institute, University of Minnesota, Minneapolis, MN, 55455, USA.
| |
Collapse
|
99
|
Ageing affects DNA methylation drift and transcriptional cell-to-cell variability in mouse muscle stem cells. Nat Commun 2019; 10:4361. [PMID: 31554804 PMCID: PMC6761124 DOI: 10.1038/s41467-019-12293-4] [Citation(s) in RCA: 150] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Accepted: 09/03/2019] [Indexed: 12/29/2022] Open
Abstract
Age-related tissue alterations have been associated with a decline in stem cell number and function. Although increased cell-to-cell variability in transcription or epigenetic marks has been proposed to be a major hallmark of ageing, little is known about the molecular diversity of stem cells during ageing. Here we present a single cell multi-omics study of mouse muscle stem cells, combining single-cell transcriptome and DNA methylome profiling. Aged cells show a global increase of uncoordinated transcriptional heterogeneity biased towards genes regulating cell-niche interactions. We find context-dependent alterations of DNA methylation in aged stem cells. Importantly, promoters with increased methylation heterogeneity are associated with increased transcriptional heterogeneity of the genes they drive. These results indicate that epigenetic drift, by accumulation of stochastic DNA methylation changes in promoters, is associated with the degradation of coherent transcriptional networks during stem cell ageing. Furthermore, our observations also shed light on the mechanisms underlying the DNA methylation clock. Age-related tissue alterations have been associated with a decline in stem cell number and function. Here the authors report a single cell multi-omics study of mouse muscle stem cells, combining single cell transcriptome and DNA methylome profiling and find that aged cells have a global increase of uncoordinated transcriptional heterogeneity biased towards genes regulating cell-niche interactions.
Collapse
|
100
|
Comai G, Heude E, Mella S, Paisant S, Pala F, Gallardo M, Langa F, Kardon G, Gopalakrishnan S, Tajbakhsh S. A distinct cardiopharyngeal mesoderm genetic hierarchy establishes antero-posterior patterning of esophagus striated muscle. eLife 2019; 8:e47460. [PMID: 31535973 PMCID: PMC6752947 DOI: 10.7554/elife.47460] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Accepted: 08/27/2019] [Indexed: 02/06/2023] Open
Abstract
In most vertebrates, the upper digestive tract is composed of muscularized jaws linked to the esophagus that permits food ingestion and swallowing. Masticatory and esophagus striated muscles (ESM) share a common cardiopharyngeal mesoderm (CPM) origin, however ESM are unusual among striated muscles as they are established in the absence of a primary skeletal muscle scaffold. Using mouse chimeras, we show that the transcription factors Tbx1 and Isl1 are required cell-autonomously for myogenic specification of ESM progenitors. Further, genetic loss-of-function and pharmacological studies point to MET/HGF signaling for antero-posterior migration of esophagus muscle progenitors, where Hgf ligand is expressed in adjacent smooth muscle cells. These observations highlight the functional relevance of a smooth and striated muscle progenitor dialogue for ESM patterning. Our findings establish a Tbx1-Isl1-Met genetic hierarchy that uniquely regulates esophagus myogenesis and identify distinct genetic signatures that can be used as framework to interpret pathologies arising within CPM derivatives.
Collapse
Affiliation(s)
- Glenda Comai
- Department of Developmental and Stem Cell BiologyInstitut PasteurParisFrance
- CNRS UMR 3738ParisFrance
| | - Eglantine Heude
- Department of Developmental and Stem Cell BiologyInstitut PasteurParisFrance
- CNRS UMR 3738ParisFrance
- Department Adaptation du VivantCNRS/MNHN UMR 7221, Muséum national d’Histoire naturelleParisFrance
| | - Sebastian Mella
- Department of Developmental and Stem Cell BiologyInstitut PasteurParisFrance
- CNRS UMR 3738ParisFrance
| | - Sylvain Paisant
- Department of Developmental and Stem Cell BiologyInstitut PasteurParisFrance
- CNRS UMR 3738ParisFrance
| | - Francesca Pala
- Department of Developmental and Stem Cell BiologyInstitut PasteurParisFrance
- CNRS UMR 3738ParisFrance
- Laboratory of Clinical Immunology and Microbiology (LCIM)National Institutes of HealthBethesdaUnited States
| | - Mirialys Gallardo
- Department of Human GeneticsUniversity of UtahSalt Lake CityUnited States
| | - Francina Langa
- Mouse Genetics Engineering CenterInstitut PasteurParisFrance
| | - Gabrielle Kardon
- Department of Human GeneticsUniversity of UtahSalt Lake CityUnited States
| | - Swetha Gopalakrishnan
- Department of Developmental and Stem Cell BiologyInstitut PasteurParisFrance
- CNRS UMR 3738ParisFrance
- Institute of Biotechnology, HiLIFEUniversity of HelsinkiHelsinkiFinland
| | - Shahragim Tajbakhsh
- Department of Developmental and Stem Cell BiologyInstitut PasteurParisFrance
- CNRS UMR 3738ParisFrance
| |
Collapse
|