51
|
Pluripotent stem cell-derived myogenic progenitors remodel their molecular signature upon in vivo engraftment. Proc Natl Acad Sci U S A 2019; 116:4346-4351. [PMID: 30760602 DOI: 10.1073/pnas.1808303116] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Optimal cell-based therapies for the treatment of muscle degenerative disorders should not only regenerate fibers but provide a quiescent satellite cell pool ensuring long-term maintenance and regeneration. Conditional expression of Pax3/Pax7 in differentiating pluripotent stem cells (PSCs) allows the generation of myogenic progenitors endowed with enhanced regenerative capacity. To identify the molecular determinants underlying their regenerative potential, we performed transcriptome analyses of these cells along with primary myogenic cells from several developmental stages. Here we show that in vitro-generated PSC-derived myogenic progenitors possess a molecular signature similar to embryonic/fetal myoblasts. However, compared with fetal myoblasts, following transplantation they show superior myofiber engraftment and ability to seed the satellite cell niche, respond to multiple reinjuries, and contribute to long-term regeneration. Upon engraftment, the transcriptome of reisolated Pax3/Pax7-induced PSC-derived myogenic progenitors changes toward a postnatal molecular signature, particularly in genes involved in extracellular matrix remodeling. These findings demonstrate that Pax3/Pax7-induced myogenic progenitors remodel their molecular signature and functionally mature upon in vivo exposure to the adult muscle environment.
Collapse
|
52
|
Song JY, Pineault KM, Wellik DM. Development, repair, and regeneration of the limb musculoskeletal system. Curr Top Dev Biol 2019; 132:451-486. [PMID: 30797517 DOI: 10.1016/bs.ctdb.2018.12.011] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The limb musculoskeletal system provides a primary means for locomotion, manipulation of objects and protection for most vertebrate organisms. Intricate integration of the bone, tendon and muscle tissues are required for function. These three tissues arise largely independent of one another, but the connections formed during later development are maintained throughout life and are re-established following injury. Each of these tissues also have mesenchymal stem/progenitor cells that function in maintenance and repair. Here in, we will review the major events in the development of limb skeleton, tendon, and muscle tissues, their response to injury, and discuss current knowledge regarding resident progenitor/stem cells within each tissue that participate in development, repair, and regeneration in vivo.
Collapse
Affiliation(s)
- Jane Y Song
- Program in Cell and Molecular Biology Program, University of Michigan, Ann Arbor, MI, United States
| | - Kyriel M Pineault
- Department of Cell & Regenerative Biology, University of Wisconsin, Madison, WI, United States
| | - Deneen M Wellik
- Department of Cell & Regenerative Biology, University of Wisconsin, Madison, WI, United States.
| |
Collapse
|
53
|
Prasad MS, Charney RM, García-Castro MI. Specification and formation of the neural crest: Perspectives on lineage segregation. Genesis 2019; 57:e23276. [PMID: 30576078 PMCID: PMC6570420 DOI: 10.1002/dvg.23276] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2018] [Revised: 12/17/2018] [Accepted: 12/18/2018] [Indexed: 12/21/2022]
Abstract
The neural crest is a fascinating embryonic population unique to vertebrates that is endowed with remarkable differentiation capacity. Thought to originate from ectodermal tissue, neural crest cells generate neurons and glia of the peripheral nervous system, and melanocytes throughout the body. However, the neural crest also generates many ectomesenchymal derivatives in the cranial region, including cell types considered to be of mesodermal origin such as cartilage, bone, and adipose tissue. These ectomesenchymal derivatives play a critical role in the formation of the vertebrate head, and are thought to be a key attribute at the center of vertebrate evolution and diversity. Further, aberrant neural crest cell development and differentiation is the root cause of many human pathologies, including cancers, rare syndromes, and birth malformations. In this review, we discuss the current findings of neural crest cell ontogeny, and consider tissue, cell, and molecular contributions toward neural crest formation. We further provide current perspectives into the molecular network involved during the segregation of the neural crest lineage.
Collapse
Affiliation(s)
- Maneeshi S Prasad
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, California
| | - Rebekah M Charney
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, California
| | - Martín I García-Castro
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, California
| |
Collapse
|
54
|
Chang CN, Singh AJ, Gross MK, Kioussi C. Requirement of Pitx2 for skeletal muscle homeostasis. Dev Biol 2019; 445:90-102. [PMID: 30414844 PMCID: PMC6289786 DOI: 10.1016/j.ydbio.2018.11.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Revised: 11/05/2018] [Accepted: 11/05/2018] [Indexed: 12/24/2022]
Abstract
Skeletal muscle is generated by the successive incorporation of primary (embryonic), secondary (fetal), and tertiary (adult) fibers into muscle. Conditional excision of Pitx2 function by an MCKCre driver resulted in animals with histological and ultrastructural defects in P30 muscles and fibers, respectively. Mutant muscle showed severe reduction in mitochondria and FoxO3-mediated mitophagy. Both oxidative and glycolytic energy metabolism were reduced. Conditional excision was limited to fetal muscle fibers after the G1-G0 transition and resulted in altered MHC, Rac1, MEF2a, and alpha-tubulin expression within these fibers. The onset of excision, monitored by a nuclear reporter gene, was observed as early as E16. Muscle at this stage was already severely malformed, but appeared to recover by P30 by the expansion of adjoining larger fibers. Our studies demonstrate that the homeodomain transcription factor Pitx2 has a postmitotic role in maintaining skeletal muscle integrity and energy homeostasis in fetal muscle fibers.
Collapse
Affiliation(s)
- Chih-Ning Chang
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Corvallis, OR 97331, USA; Molecular Cell Biology Graduate Program, Oregon State University, Corvallis, OR 97331, USA
| | - Arun J Singh
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Corvallis, OR 97331, USA
| | - Michael K Gross
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Corvallis, OR 97331, USA
| | - Chrissa Kioussi
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Corvallis, OR 97331, USA.
| |
Collapse
|
55
|
Cho DS, Doles JD. Skeletal Muscle Progenitor Cell Heterogeneity. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1169:179-193. [PMID: 31487024 DOI: 10.1007/978-3-030-24108-7_9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Tissue-specific stem cells contribute to adult tissue maintenance, repair, and regeneration. In skeletal muscle, many different mononuclear cell types are capable of giving rise to differentiated muscle. Of these tissue stem-like cells, satellite cells (SCs) are the most studied muscle stem cell population and are widely considered the main cellular source driving muscle repair and regeneration in adult tissue. Within the satellite cell pool, many distinct subpopulations exist, each exhibiting differential abilities to exit quiescence, expand, differentiate, and self-renew. In this chapter, we discuss the different stem cell types that can give rise to skeletal muscle tissue and then focus on satellite cell heterogeneity during the process of myogenesis/muscle regeneration. Finally, we highlight emerging opportunities to better characterize muscle stem cell heterogeneity, which will ultimately deepen our appreciation of stem cells in muscle development, repair/regeneration, aging, and disease.
Collapse
Affiliation(s)
- Dong Seong Cho
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, USA
| | - Jason D Doles
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
56
|
Heude E, Tesarova M, Sefton EM, Jullian E, Adachi N, Grimaldi A, Zikmund T, Kaiser J, Kardon G, Kelly RG, Tajbakhsh S. Unique morphogenetic signatures define mammalian neck muscles and associated connective tissues. eLife 2018; 7:40179. [PMID: 30451684 PMCID: PMC6310459 DOI: 10.7554/elife.40179] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Accepted: 11/17/2018] [Indexed: 12/16/2022] Open
Abstract
In vertebrates, head and trunk muscles develop from different mesodermal populations and are regulated by distinct genetic networks. Neck muscles at the head-trunk interface remain poorly defined due to their complex morphogenesis and dual mesodermal origins. Here, we use genetically modified mice to establish a 3D model that integrates regulatory genes, cell populations and morphogenetic events that define this transition zone. We show that the evolutionary conserved cucullaris-derived muscles originate from posterior cardiopharyngeal mesoderm, not lateral plate mesoderm, and we define new boundaries for neural crest and mesodermal contributions to neck connective tissue. Furthermore, lineage studies and functional analysis of Tbx1- and Pax3-null mice reveal a unique developmental program for somitic neck muscles that is distinct from that of somitic trunk muscles. Our findings unveil the embryological and developmental requirements underlying tetrapod neck myogenesis and provide a blueprint to investigate how muscle subsets are selectively affected in some human myopathies.
Collapse
Affiliation(s)
- Eglantine Heude
- Department of Developmental and Stem Cell Biology, Institut Pasteur, Paris, France.,CNRS UMR 3738, Paris, France
| | - Marketa Tesarova
- Central European Institute of Technology, Brno University of Technology, Brno, Czech Republic
| | - Elizabeth M Sefton
- Department of Human Genetics, University of Utah, Salt Lake City, United States
| | - Estelle Jullian
- Aix-Marseille Université, CNRS UMR 7288, IBDM, Marseille, France
| | - Noritaka Adachi
- Aix-Marseille Université, CNRS UMR 7288, IBDM, Marseille, France
| | - Alexandre Grimaldi
- Department of Developmental and Stem Cell Biology, Institut Pasteur, Paris, France.,CNRS UMR 3738, Paris, France
| | - Tomas 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
| | - Gabrielle Kardon
- Department of Human Genetics, University of Utah, Salt Lake City, United States
| | - Robert G Kelly
- Aix-Marseille Université, CNRS UMR 7288, IBDM, Marseille, France
| | - Shahragim Tajbakhsh
- Department of Developmental and Stem Cell Biology, Institut Pasteur, Paris, France.,CNRS UMR 3738, Paris, France
| |
Collapse
|
57
|
Zecchini S, Giovarelli M, Perrotta C, Morisi F, Touvier T, Di Renzo I, Moscheni C, Bassi MT, Cervia D, Sandri M, Clementi E, De Palma C. Autophagy controls neonatal myogenesis by regulating the GH-IGF1 system through a NFE2L2- and DDIT3-mediated mechanism. Autophagy 2018; 15:58-77. [PMID: 30081710 DOI: 10.1080/15548627.2018.1507439] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Macroautophagy/autophagy is emerging as an important process in adult muscle stem cells functions: it regulates metabolic reprogramming during activation from a quiescent state, maintains stemness and prevents senescence. We now show that autophagy is specifically required for neonatal myogenesis and muscle development. Specific deletion of Atg7 in PAX7+ (paired box 7) precursors led in mice to a dwarf phenotype, with an effect restricted to the neonatal phase of muscle development. Atg7 knockdown suppressed neonatal satellite cell (nSC) proliferation and differentiation, downregulating the GH-IGF1 functions. When we disrupted autophagy, NFE2L2/NRF2 (nuclear factor, erythroid 2 like 2) accumulated in muscle and nSCs and negatively modulated DDIT3/CHOP (DNA-damage inducible transcript 3) expression. Lower levels of DDIT3 were responsible for reduced GHR expression leading to impaired local production of IGF1. Our results conclusively identify a novel autophagy-dependent pathway that regulates nSC behavior and indicate that autophagy is required for skeletal muscle development in the neonatal phase. Abbreviations: AKT/protein kinase B: Thymoma viral proto-oncogene; ASCs: adult stem cells; ATF4: activating transcription factor 4; ATG7: autophagy related 7; BAT: brown adipose tissue; BMP: bone morphogenetic protein; CEBPB: CCAAT/enhancer binding protein (C/EBP), beta; CSA: cross sectional area; CTNNB1: catenin (cadherin associated protein), beta 1; DDIT3: DNA-damage inducible transcript 3; DM: differentiation medium; E: embryonic stage; EIF2AK3/PERK; EIF4EBP1: eukaryotic translation initiation factor 2 alpha kinase 3; eukaryotic translation initiation factor 4E binding protein 1; ER: endoplasmic reticulum; FGF21: fibroblast growth factor 21; GH: growth hormone; GHR: growth hormone receptor; HSCs: hematopoietic stem cells; IGF1: insulin-like growth factor 1; ITGAM: integrin alpha M; KEAP1: kelch-like ECH-associated protein 1; LY6A/Sca-1; MAP1LC3: lymphocyte antigen 6 complex, locus A; microtubule-associated protein 1 light chain 3; MAPK1/ERK2: mitogen-activated protein kinase 1; MAPK3/ERK1: mitogen-activated protein kinase 3; miRNAs: microRNAs; MSCs: mesenchymal stem cells; MTOR: mechanistic target of rapamycin kinase; mtUPR: mitochondrial unfolded protein response; MYF5: myogenic factor 5; MYH: myosin, heavy polypeptide; MYOD1: myogenic differentiation 1; MYOG: myogenin; NFE2L2: nuclear factor, erythroid derived 2, like 2; nSC: neonatal satellite cells; NSCs: neuronal stem cells; P: postnatal day; PAX7: paired box 7; PECAM1: platelet/endothelial cell adhesion molecule 1; PPARG: peroxisome proliferator activated receptor gamma; PTPRC: protein tyrosine phosphatase, receptor type, C; ROS: reactive oxygen species; RPS6: ribosomal protein S6; SCs: adult satellite cells; SQSTM1: sequestosome 1; STAT5: signal transducer and activator of transcription 5; TGFB1: transforming growth factor beta 1; WAT: white adipose tissue; WT: wild type.
Collapse
Affiliation(s)
- Silvia Zecchini
- a Unit of Clinical Pharmacology , University Hospital "Luigi Sacco"-ASST Fatebenefratelli Sacco , Milano , Italy
| | - Matteo Giovarelli
- b Department of Biomedical and Clinical Sciences "Luigi Sacco" , Università degli Studi di Milano , Milano , Italy
| | - Cristiana Perrotta
- b Department of Biomedical and Clinical Sciences "Luigi Sacco" , Università degli Studi di Milano , Milano , Italy
| | - Federica Morisi
- c Division of Genetics and Cell Biology , IRCCS Ospedale San Raffaele , Milano , Italy
| | - Thierry Touvier
- d Biology of Myelin Unit, Division of Genetics and Cell Biology , IRCCS Ospedale San Raffaele , Milano , Italy
| | - Ilaria Di Renzo
- b Department of Biomedical and Clinical Sciences "Luigi Sacco" , Università degli Studi di Milano , Milano , Italy
| | - Claudia Moscheni
- b Department of Biomedical and Clinical Sciences "Luigi Sacco" , Università degli Studi di Milano , Milano , Italy
| | - Maria Teresa Bassi
- e Laboratory of Molecular Biology , IRCCS Eugenio Medea , Bosisio Parini , Italy
| | - Davide Cervia
- f Department for Innovation in Biological, Agro-food and Forest systems , Università degli Studi della Tuscia , Viterbo , Italy
| | - Marco Sandri
- g Department of Biomedical Science , University of Padova , Padova , Italy.,h Laboratory of Molecular Biology , Venetian Institute of Molecular Medicine , Padova , Italy
| | - Emilio Clementi
- e Laboratory of Molecular Biology , IRCCS Eugenio Medea , Bosisio Parini , Italy.,i Department of Biomedical and Clinical Sciences "Luigi Sacco" , Università degli Studi di Milano , Milano , Italy
| | - Clara De Palma
- a Unit of Clinical Pharmacology , University Hospital "Luigi Sacco"-ASST Fatebenefratelli Sacco , Milano , Italy.,b Department of Biomedical and Clinical Sciences "Luigi Sacco" , Università degli Studi di Milano , Milano , Italy
| |
Collapse
|
58
|
Tichy ED, Sidibe DK, Greer CD, Oyster NM, Rompolas P, Rosenthal NA, Blau HM, Mourkioti F. A robust Pax7EGFP mouse that enables the visualization of dynamic behaviors of muscle stem cells. Skelet Muscle 2018; 8:27. [PMID: 30139374 PMCID: PMC6107960 DOI: 10.1186/s13395-018-0169-7] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Accepted: 07/04/2018] [Indexed: 01/10/2023] Open
Abstract
Background Pax7 is a transcription factor involved in the specification and maintenance of muscle stem cells (MuSCs). Upon injury, MuSCs leave their quiescent state, downregulate Pax7 and differentiate, contributing to skeletal muscle regeneration. In the majority of regeneration studies, MuSCs are isolated by fluorescence-activated sorting (FACS), based on cell surface markers. It is known that MuSCs are a heterogeneous population and only a small percentage of isolated cells are true stem cells that are able to self-renew. A strong Pax7 reporter line would be valuable to study the in vivo behavior of Pax7-expressing stem cells. Methods We generated and characterized the muscle properties of a new transgenic Pax7EGFP mouse. Utilizing traditional immunofluorescence assays, we analyzed whole embryos and muscle sections by fluorescence microscopy, in addition to whole skeletal muscles by 2-photon microscopy, to detect the specificity of EGFP expression. Skeletal muscles from Pax7EGFP mice were also evaluated in steady state and under injury conditions. Finally, MuSCs-derived from Pax7EGFP and control mice were sorted and analyzed by FACS and their myogenic activity was comparatively examined. Results Our studies provide a new Pax7 reporter line with robust EGFP expression, detectable by both flow cytometry and fluorescence microscopy. Pax7EGFP-derived MuSCs have identical properties to that of wild-type MuSCs, both in vitro and in vivo, excluding any positional effect due to the transgene insertion. Furthermore, we demonstrated high specificity of EGFP to label MuSCs in a temporal manner that recapitulates the reported Pax7 expression pattern. Interestingly, immunofluorescence analysis showed that the robust expression of EGFP marks cells in the satellite cell position of adult muscles in fixed and live tissues. Conclusions This mouse could be an invaluable tool for the study of a variety of questions related to MuSC biology, including but not limited to population heterogeneity, polarity, aging, regeneration, and motility, either by itself or in combination with mice harboring additional genetic alterations. Electronic supplementary material The online version of this article (10.1186/s13395-018-0169-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Elisia D Tichy
- Department of Orthopaedic Surgery, Perelman School of Medicine, The University of Pennsylvania, Philadelphia, PA, USA
| | - David K Sidibe
- Department of Orthopaedic Surgery, Perelman School of Medicine, The University of Pennsylvania, Philadelphia, PA, USA
| | - Christopher D Greer
- Department of Orthopaedic Surgery, Perelman School of Medicine, The University of Pennsylvania, Philadelphia, PA, USA.,Cell and Molecular Biology Graduate Program, The University of Pennsylvania, Philadelphia, PA, USA
| | - Nicholas M Oyster
- Department of Orthopaedic Surgery, Perelman School of Medicine, The University of Pennsylvania, Philadelphia, PA, USA
| | - Panteleimon Rompolas
- Cell and Molecular Biology Graduate Program, The University of Pennsylvania, Philadelphia, PA, USA.,Department of Dermatology, Institute for Regenerative Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Nadia A Rosenthal
- The Jackson Laboratory, Bar Harbor, ME, USA.,Sackler School of Graduate Biomedical Sciences, Tufts University, Boston, MA, USA.,The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA.,Australian Regenerative Medicine Institute, Monash University, Melbourne, VIC, Australia.,National Heart and Lung Institute, Imperial College London, London, UK
| | - Helen M Blau
- Baxter Laboratory for Stem Cell Biology, Department of Microbiology and Immunology, Institute for Stem Cell Biology and Regenerative Medicine, Stanford School of Medicine, Stanford, CA, USA
| | - Foteini Mourkioti
- Department of Orthopaedic Surgery, Perelman School of Medicine, The University of Pennsylvania, Philadelphia, PA, USA. .,Cell and Molecular Biology Graduate Program, The University of Pennsylvania, Philadelphia, PA, USA. .,Department of Cell and Developmental Biology, Penn Institute of Regenerative Medicine, Musculoskeletal Regeneration Program, Perelman School of Medicine, The University of Pennsylvania, Philadelphia, PA, USA. .,Musculoskeletal Regeneration Program, Department of Orthopaedic Surgery and Cell and Developmental Biology, Penn Institute of Regenerative Medicine, The University of Pennsylvania, 3450 Hamilton Walk, 112A Stemmler Hall, Philadelphia, PA, 19104-6081, USA.
| |
Collapse
|
59
|
Kitajima Y, Ono Y. Visualization of PAX7 protein dynamics in muscle satellite cells in a YFP knock-in-mouse line. Skelet Muscle 2018; 8:26. [PMID: 30139390 PMCID: PMC6108100 DOI: 10.1186/s13395-018-0174-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Accepted: 08/01/2018] [Indexed: 11/13/2022] Open
Abstract
Background Satellite cells are residential muscle stem cells that express a paired box protein, PAX7. Results Here, we report a knock-in mouse line expressing a PAX7-enhanced yellow fluorescent protein (YFP) fusion protein that enables visualization of PAX7 protein dynamics in living satellite cells through YFP fluorescence. The YFP fluorescence signals in Pax7-YFP knock-in mice clearly recapitulated the endogenous expression of PAX7 protein in satellite cells. YFP+ satellite cells were efficiently isolated from muscle tissues by fluorescence-activated cell sorting. Homozygous Pax7-YFP knock-in mice (Pax7YFP/YFP) were viable, grew and regenerated muscle normally, and Pax7YFP/YFP mouse-derived satellite cells proliferated, differentiated, and self-renewed as efficiently as those from wild-type (Pax7+/+) mice. Conclusions Taken together, our Pax7-YFP mouse line is a useful tool to aid the development of stem-cell-based therapies for muscle diseases. Electronic supplementary material The online version of this article (10.1186/s13395-018-0174-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Yasuo Kitajima
- Musculoskeletal Molecular Biology Research Group, Basic and Translational Research Center for Hard Tissue Disease, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1 Sakamoto, Nagasaki, 852-8588, Japan.,Japan Society for the Promotion of Science, 5-3-1 Kojimachi, Chiyoda-ku, Tokyo, 102-0083, Japan
| | - Yusuke Ono
- Musculoskeletal Molecular Biology Research Group, Basic and Translational Research Center for Hard Tissue Disease, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1 Sakamoto, Nagasaki, 852-8588, Japan. .,Agency for Medical Research and Development (AMED), 1-7-1 Otemachi, Chiyodaku, Tokyo, 100-0004, Japan.
| |
Collapse
|
60
|
Barton-Owen TB, Ferrier DEK, Somorjai IML. Pax3/7 duplicated and diverged independently in amphioxus, the basal chordate lineage. Sci Rep 2018; 8:9414. [PMID: 29925900 PMCID: PMC6010424 DOI: 10.1038/s41598-018-27700-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2018] [Accepted: 06/06/2018] [Indexed: 01/06/2023] Open
Abstract
The Pax3/7 transcription factor family is integral to developmental gene networks contributing to important innovations in vertebrate evolution, including the neural crest. The basal chordate lineage of amphioxus is ideally placed to understand the dynamics of the gene regulatory network evolution that produced these novelties. We report here the discovery that the cephalochordate lineage possesses two Pax3/7 genes, Pax3/7a and Pax3/7b. The tandem duplication is ancestral to all extant amphioxus, occurring in both Asymmetron and Branchiostoma, but originated after the split from the lineage leading to vertebrates. The two paralogues are differentially expressed during embryonic development, particularly in neural and somitic tissues, suggesting distinct regulation. Our results have implications for the study of amphioxus regeneration, neural plate and crest evolution, and differential tandem paralogue evolution.
Collapse
Affiliation(s)
- Thomas B Barton-Owen
- University of St Andrews, Gatty Marine Laboratory, Scottish Oceans Institute, East Sands, St Andrews, Fife, KY16 8LB, UK.,University of St Andrews, Biomedical Sciences Research Complex, North Haugh, St Andrews, Fife, KY16 9ST, UK
| | - David E K Ferrier
- University of St Andrews, Gatty Marine Laboratory, Scottish Oceans Institute, East Sands, St Andrews, Fife, KY16 8LB, UK
| | - Ildikó M L Somorjai
- University of St Andrews, Gatty Marine Laboratory, Scottish Oceans Institute, East Sands, St Andrews, Fife, KY16 8LB, UK. .,University of St Andrews, Biomedical Sciences Research Complex, North Haugh, St Andrews, Fife, KY16 9ST, UK.
| |
Collapse
|
61
|
Pla P, Monsoro-Burq AH. The neural border: Induction, specification and maturation of the territory that generates neural crest cells. Dev Biol 2018; 444 Suppl 1:S36-S46. [PMID: 29852131 DOI: 10.1016/j.ydbio.2018.05.018] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2018] [Revised: 05/23/2018] [Accepted: 05/23/2018] [Indexed: 11/17/2022]
Abstract
The neural crest is induced at the edge between the neural plate and the nonneural ectoderm, in an area called the neural (plate) border, during gastrulation and neurulation. In recent years, many studies have explored how this domain is patterned, and how the neural crest is induced within this territory, that also participates to the prospective dorsal neural tube, the dorsalmost nonneural ectoderm, as well as placode derivatives in the anterior area. This review highlights the tissue interactions, the cell-cell signaling and the molecular mechanisms involved in this dynamic spatiotemporal patterning, resulting in the induction of the premigratory neural crest. Collectively, these studies allow building a complex neural border and early neural crest gene regulatory network, mostly composed by transcriptional regulations but also, more recently, including novel signaling interactions.
Collapse
Affiliation(s)
- Patrick Pla
- Univ. Paris Sud, Université Paris Saclay, CNRS UMR 3347, INSERM U1021, Centre Universitaire, 15, rue Georges Clémenceau, F-91405 Orsay, France; Institut Curie Research Division, PSL Research University, CNRS UMR 3347, INSERM U1021, F-91405 Orsay, France
| | - Anne H Monsoro-Burq
- Univ. Paris Sud, Université Paris Saclay, CNRS UMR 3347, INSERM U1021, Centre Universitaire, 15, rue Georges Clémenceau, F-91405 Orsay, France; Institut Curie Research Division, PSL Research University, CNRS UMR 3347, INSERM U1021, F-91405 Orsay, France; Institut Universitaire de France, F-75005, Paris.
| |
Collapse
|
62
|
Chang CN, Kioussi C. Location, Location, Location: Signals in Muscle Specification. J Dev Biol 2018; 6:E11. [PMID: 29783715 PMCID: PMC6027348 DOI: 10.3390/jdb6020011] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Revised: 05/11/2018] [Accepted: 05/15/2018] [Indexed: 12/15/2022] Open
Abstract
Muscles control body movement and locomotion, posture and body position and soft tissue support. Mesoderm derived cells gives rise to 700 unique muscles in humans as a result of well-orchestrated signaling and transcriptional networks in specific time and space. Although the anatomical structure of skeletal muscles is similar, their functions and locations are specialized. This is the result of specific signaling as the embryo grows and cells migrate to form different structures and organs. As cells progress to their next state, they suppress current sequence specific transcription factors (SSTF) and construct new networks to establish new myogenic features. In this review, we provide an overview of signaling pathways and gene regulatory networks during formation of the craniofacial, cardiac, vascular, trunk, and limb skeletal muscles.
Collapse
Affiliation(s)
- Chih-Ning Chang
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Corvallis, OR 97331, USA.
- Molecular Cell Biology Graduate Program, Oregon State University, Corvallis, OR 97331, USA.
| | - Chrissa Kioussi
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Corvallis, OR 97331, USA.
- Molecular Cell Biology Graduate Program, Oregon State University, Corvallis, OR 97331, USA.
| |
Collapse
|
63
|
Boudjadi S, Chatterjee B, Sun W, Vemu P, Barr FG. The expression and function of PAX3 in development and disease. Gene 2018; 666:145-157. [PMID: 29730428 DOI: 10.1016/j.gene.2018.04.087] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2017] [Revised: 04/26/2018] [Accepted: 04/27/2018] [Indexed: 12/27/2022]
Abstract
The PAX3 gene encodes a member of the PAX family of transcription factors that is characterized by a highly conserved paired box motif. The PAX3 protein is a transcription factor consisting of an N-terminal DNA binding domain (containing a paired box and homeodomain) and a C-terminal transcriptional activation domain. This protein is expressed during development of skeletal muscle, central nervous system and neural crest derivatives, and regulates expression of target genes that impact on proliferation, survival, differentiation and motility in these lineages. Germline mutations of the murine Pax3 and human PAX3 genes cause deficiencies in these developmental lineages and result in the Splotch phenotype and Waardenburg syndrome, respectively. Somatic genetic rearrangements that juxtapose the PAX3 DNA binding domain to the transcriptional activation domain of other transcription factors deregulate PAX3 function and contribute to the pathogenesis of the soft tissue cancers alveolar rhabdomyosarcoma and biphenotypic sinonasal sarcoma. The wild-type PAX3 protein is also expressed in other cancers related to developmental lineages that normally express this protein and exerts phenotypic effects related to its normal developmental role.
Collapse
Affiliation(s)
- Salah Boudjadi
- Laboratory of Pathology, National Cancer Institute, Bethesda, MD, USA
| | | | - Wenyue Sun
- Laboratory of Pathology, National Cancer Institute, Bethesda, MD, USA
| | - Prasantha Vemu
- Laboratory of Pathology, National Cancer Institute, Bethesda, MD, USA
| | - Frederic G Barr
- Laboratory of Pathology, National Cancer Institute, Bethesda, MD, USA.
| |
Collapse
|
64
|
Yamamoto M, Legendre NP, Biswas AA, Lawton A, Yamamoto S, Tajbakhsh S, Kardon G, Goldhamer DJ. Loss of MyoD and Myf5 in Skeletal Muscle Stem Cells Results in Altered Myogenic Programming and Failed Regeneration. Stem Cell Reports 2018; 10:956-969. [PMID: 29478898 PMCID: PMC5918368 DOI: 10.1016/j.stemcr.2018.01.027] [Citation(s) in RCA: 83] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Revised: 01/22/2018] [Accepted: 01/23/2018] [Indexed: 11/22/2022] Open
Abstract
MyoD and Myf5 are fundamental regulators of skeletal muscle lineage determination in the embryo, and their expression is induced in satellite cells following muscle injury. MyoD and Myf5 are also expressed by satellite cell precursors developmentally, although the relative contribution of historical and injury-induced expression to satellite cell function is unknown. We show that satellite cells lacking both MyoD and Myf5 (double knockout [dKO]) are maintained with aging in uninjured muscle. However, injured muscle fails to regenerate and dKO satellite cell progeny accumulate in damaged muscle but do not undergo muscle differentiation. dKO satellite cell progeny continue to express markers of myoblast identity, although their myogenic programming is labile, as demonstrated by dramatic morphological changes and increased propensity for non-myogenic differentiation. These data demonstrate an absolute requirement for either MyoD or Myf5 in muscle regeneration and indicate that their expression after injury stabilizes myogenic identity and confers the capacity for muscle differentiation. MyoD or Myf5 expression in satellite cells is essential for muscle regeneration Satellite cells lacking both regulatory genes exhibit labile myogenic programming A single functional allele of either MyoD or Myf5 can support muscle regeneration Satellite cells lacking both MyoD and Myf5 are maintained with aging
Collapse
Affiliation(s)
- Masakazu Yamamoto
- Department of Molecular & Cell Biology, University of Connecticut Stem Cell Institute, University of Connecticut, 91 N. Eagleville Road, Storrs, CT 06269, USA
| | - Nicholas P Legendre
- Department of Molecular & Cell Biology, University of Connecticut Stem Cell Institute, University of Connecticut, 91 N. Eagleville Road, Storrs, CT 06269, USA
| | - Arpita A Biswas
- Department of Molecular & Cell Biology, University of Connecticut Stem Cell Institute, University of Connecticut, 91 N. Eagleville Road, Storrs, CT 06269, USA
| | - Alexander Lawton
- Department of Molecular & Cell Biology, University of Connecticut Stem Cell Institute, University of Connecticut, 91 N. Eagleville Road, Storrs, CT 06269, USA
| | - Shoko Yamamoto
- Department of Molecular & Cell Biology, University of Connecticut Stem Cell Institute, University of Connecticut, 91 N. Eagleville Road, Storrs, CT 06269, USA
| | - Shahragim Tajbakhsh
- Institut Pasteur, Stem Cells & Development, CNRS URA 2578, 25 rue du Dr. Roux, 75724 Paris Cedex 15, France
| | - Gabrielle Kardon
- Department of Human Genetics, University of Utah, 15 North 2030 East, Salt Lake City, UT 84112, USA
| | - David J Goldhamer
- Department of Molecular & Cell Biology, University of Connecticut Stem Cell Institute, University of Connecticut, 91 N. Eagleville Road, Storrs, CT 06269, USA.
| |
Collapse
|
65
|
The axolotl genome and the evolution of key tissue formation regulators. Nature 2018; 554:50-55. [PMID: 29364872 DOI: 10.1038/nature25458] [Citation(s) in RCA: 320] [Impact Index Per Article: 53.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2017] [Accepted: 12/13/2017] [Indexed: 02/08/2023]
Abstract
Salamanders serve as important tetrapod models for developmental, regeneration and evolutionary studies. An extensive molecular toolkit makes the Mexican axolotl (Ambystoma mexicanum) a key representative salamander for molecular investigations. Here we report the sequencing and assembly of the 32-gigabase-pair axolotl genome using an approach that combined long-read sequencing, optical mapping and development of a new genome assembler (MARVEL). We observed a size expansion of introns and intergenic regions, largely attributable to multiplication of long terminal repeat retroelements. We provide evidence that intron size in developmental genes is under constraint and that species-restricted genes may contribute to limb regeneration. The axolotl genome assembly does not contain the essential developmental gene Pax3. However, mutation of the axolotl Pax3 paralogue Pax7 resulted in an axolotl phenotype that was similar to those seen in Pax3-/- and Pax7-/- mutant mice. The axolotl genome provides a rich biological resource for developmental and evolutionary studies.
Collapse
|
66
|
Schubert FR, Singh AJ, Afoyalan O, Kioussi C, Dietrich S. To roll the eyes and snap a bite - function, development and evolution of craniofacial muscles. Semin Cell Dev Biol 2018; 91:31-44. [PMID: 29331210 DOI: 10.1016/j.semcdb.2017.12.013] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Revised: 12/15/2017] [Accepted: 12/18/2017] [Indexed: 02/06/2023]
Abstract
Craniofacial muscles, muscles that move the eyes, control facial expression and allow food uptake and speech, have long been regarded as a variation on the general body muscle scheme. However, evidence has accumulated that the function of head muscles, their developmental anatomy and the underlying regulatory cascades are distinct. This article reviews the key aspects of craniofacial muscle and muscle stem cell formation and discusses how this differs from the trunk programme of myogenesis; we show novel RNAseq data to support this notion. We also trace the origin of head muscle in the chordate ancestors of vertebrates and discuss links with smooth-type muscle in the primitive chordate pharynx. We look out as to how the special properties of head muscle precursor and stem cells, in particular their competence to contribute to the heart, could be exploited in regenerative medicine.
Collapse
Affiliation(s)
- Frank R Schubert
- Institute of Biomedical and Biomolecular Sciences, University of Portsmouth, Portsmouth, UK
| | - Arun J Singh
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Corvallis, OR, 97331, USA
| | - Oluwatomisin Afoyalan
- Institute of Biomedical and Biomolecular Sciences, University of Portsmouth, Portsmouth, UK
| | - Chrissa Kioussi
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Corvallis, OR, 97331, USA
| | - Susanne Dietrich
- Institute of Biomedical and Biomolecular Sciences, University of Portsmouth, Portsmouth, UK.
| |
Collapse
|
67
|
Hernández-Hernández JM, García-González EG, Brun CE, Rudnicki MA. The myogenic regulatory factors, determinants of muscle development, cell identity and regeneration. Semin Cell Dev Biol 2017; 72:10-18. [PMID: 29127045 DOI: 10.1016/j.semcdb.2017.11.010] [Citation(s) in RCA: 341] [Impact Index Per Article: 48.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Revised: 11/04/2017] [Accepted: 11/06/2017] [Indexed: 10/18/2022]
Abstract
The Myogenic Regulatory Factors (MRFs) Myf5, MyoD, myogenin and MRF4 are members of the basic helix-loop-helix family of transcription factors that control the determination and differentiation of skeletal muscle cells during embryogenesis and postnatal myogenesis. The dynamics of their temporal and spatial expression as well as their biochemical properties have allowed the identification of a precise and hierarchical relationship between the four MRFs. This relationship establishes the myogenic lineage as well as the maintenance of the terminal myogenic phenotype. The application of genome-wide technologies has provided important new information as to how the MRFs function to activate muscle gene expression. Application of combined functional genomics technologies along with single cell lineage tracing strategies will allow a deeper understanding of the mechanisms mediating myogenic determination, cell differentiation and muscle regeneration.
Collapse
Affiliation(s)
- J Manuel Hernández-Hernández
- The Sprott Centre for Stem Cell Research, Regenerative Medicine Program, Ottawa Hospital Research Institute, 501 Smyth Road, Ottawa, ON, K1H 8L6, Canada; Departments of Medicine and Cellular and Molecular Medicine, University of Ottawa, 451 Smyth Road, Ottawa, ON, K1H 8M5, Canada
| | - Estela G García-González
- The Sprott Centre for Stem Cell Research, Regenerative Medicine Program, Ottawa Hospital Research Institute, 501 Smyth Road, Ottawa, ON, K1H 8L6, Canada; Departments of Medicine and Cellular and Molecular Medicine, University of Ottawa, 451 Smyth Road, Ottawa, ON, K1H 8M5, Canada
| | - Caroline E Brun
- The Sprott Centre for Stem Cell Research, Regenerative Medicine Program, Ottawa Hospital Research Institute, 501 Smyth Road, Ottawa, ON, K1H 8L6, Canada; Departments of Medicine and Cellular and Molecular Medicine, University of Ottawa, 451 Smyth Road, Ottawa, ON, K1H 8M5, Canada
| | - Michael A Rudnicki
- The Sprott Centre for Stem Cell Research, Regenerative Medicine Program, Ottawa Hospital Research Institute, 501 Smyth Road, Ottawa, ON, K1H 8L6, Canada; Departments of Medicine and Cellular and Molecular Medicine, University of Ottawa, 451 Smyth Road, Ottawa, ON, K1H 8M5, Canada.
| |
Collapse
|
68
|
Effects of insulin like growth factors on early embryonic chick limb myogenesis. PLoS One 2017; 12:e0185775. [PMID: 28972999 PMCID: PMC5626492 DOI: 10.1371/journal.pone.0185775] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Accepted: 09/19/2017] [Indexed: 11/19/2022] Open
Abstract
Limb muscles derive from pax3 expressing precursor cells that migrate from the hypaxial somite into the developing limb bud. Once there they begin to differentiate and express muscle determination genes such as MyoD. This process is regulated by a combination of inductive or inhibitory signals including Fgf18, retinoic acid, HGF, Notch and IGFs. IGFs are well known to affect late stages of muscle development and to promote both proliferation and differentiation. We examined their roles in early stage limb bud myogenesis using chicken embryos as an experimental model. Grafting beads soaked in purified recombinant IGF-I, IGF-II or small molecule inhibitors of specific signaling pathways into developing chick embryo limbs showed that both IGF-I and IGF-II induce expression of the early stage myogenic markers pax3 and MyoD as well as myogenin. Their effects on pax3 and MyoD expression were blocked by inhibitors of both the IGF type I receptor (picropodophyllotoxin, PPP) and MEK (U0126). The PI3K inhibitor LY294002 blocked IGF-II, but not IGF-I, induction of pax3 mRNA as well as the IGF-I, but not IGF-II, induction of MyoD mRNA. In addition SU5402, an FGFR/ VEGFR inhibitor, blocked the induction of MyoD by both IGFs but had no effect on pax3 induction, suggesting a role for FGF or VEGF signaling in their induction of MyoD. This was confirmed by in situ hybridization showing that FGF18, a known regulator of MyoD in limb myoblasts, was induced by IGF-I. In addition to their well-known effects on later stages of myogenesis via their induction of myogenin expression, both IGF-I and IGF-II induced pax3 and MyoD expression in developing chick embryos, indicating that they also regulate early stages of myogenesis. The data suggests that the IGFs may have slightly different effects on IGF1R signal transduction via PI3K and that their stimulatory effects on MyoD expression may be indirect, possibly via induction of FGF18 expression.
Collapse
|
69
|
Abstract
Skeletal muscle is the largest tissue in the body and loss of its function or its regenerative properties results in debilitating musculoskeletal disorders. Understanding the mechanisms that drive skeletal muscle formation will not only help to unravel the molecular basis of skeletal muscle diseases, but also provide a roadmap for recapitulating skeletal myogenesis in vitro from pluripotent stem cells (PSCs). PSCs have become an important tool for probing developmental questions, while differentiated cell types allow the development of novel therapeutic strategies. In this Review, we provide a comprehensive overview of skeletal myogenesis from the earliest premyogenic progenitor stage to terminally differentiated myofibers, and discuss how this knowledge has been applied to differentiate PSCs into muscle fibers and their progenitors in vitro.
Collapse
Affiliation(s)
- Jérome Chal
- Department of Pathology, Brigham and Women's Hospital, 77 Avenue Louis Pasteur, Boston, MA 02115, USA.,Department of Genetics, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA.,Harvard Stem Cell Institute, 77 Avenue Louis Pasteur, Boston, MA 02115, USA
| | - Olivier Pourquié
- Department of Pathology, Brigham and Women's Hospital, 77 Avenue Louis Pasteur, Boston, MA 02115, USA .,Department of Genetics, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA.,Harvard Stem Cell Institute, 77 Avenue Louis Pasteur, Boston, MA 02115, USA.,Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), CNRS (UMR 7104), Inserm U964, Université de Strasbourg, 67400 Illkirch-Graffenstaden, France
| |
Collapse
|
70
|
Rajgara RF, Lala-Tabbert N, Marchildon F, Lamarche É, MacDonald JK, Scott DA, Blais A, Skerjanc IS, Wiper-Bergeron N. SOX7 Is Required for Muscle Satellite Cell Development and Maintenance. Stem Cell Reports 2017; 9:1139-1151. [PMID: 28943254 PMCID: PMC5639291 DOI: 10.1016/j.stemcr.2017.08.014] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Revised: 08/22/2017] [Accepted: 08/23/2017] [Indexed: 01/26/2023] Open
Abstract
Satellite cells are skeletal-muscle-specific stem cells that are activated by injury to proliferate, differentiate, and fuse to enable repair. SOX7, a member of the SRY-related HMG-box family of transcription factors is expressed in quiescent satellite cells. To elucidate SOX7 function in skeletal muscle, we knocked down Sox7 expression in embryonic stem cells and primary myoblasts and generated a conditional knockout mouse in which Sox7 is excised in PAX3+ cells. Loss of Sox7 in embryonic stem cells reduced Pax3 and Pax7 expression. In vivo, conditional knockdown of Sox7 reduced the satellite cell population from birth, reduced myofiber caliber, and impaired regeneration after acute injury. Although Sox7-deficient primary myoblasts differentiated normally, impaired myoblast fusion and increased sensitivity to apoptosis in culture and in vivo were observed. Taken together, these results indicate that SOX7 is dispensable for myogenesis but is necessary to promote satellite cell development and survival.
Collapse
Affiliation(s)
- Rashida F Rajgara
- Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ottawa, ON K1H 8M5, Canada; Graduate Program in Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Neena Lala-Tabbert
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - François Marchildon
- Graduate Program in Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Émilie Lamarche
- Graduate Program in Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Jennifer K MacDonald
- Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Daryl A Scott
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Alexandre Blais
- Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ottawa, ON K1H 8M5, Canada; Ottawa Institute of Systems Biology, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Ilona S Skerjanc
- Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Nadine Wiper-Bergeron
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada.
| |
Collapse
|
71
|
Haynes P, Kernan K, Zhou SL, Miller DG. Expression patterns of FSHD-causing DUX4 and myogenic transcription factors PAX3 and PAX7 are spatially distinct in differentiating human stem cell cultures. Skelet Muscle 2017. [PMID: 28637492 PMCID: PMC5480156 DOI: 10.1186/s13395-017-0130-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Background Facioscapulohumeral muscular dystrophy (FSHD) is most commonly inherited in an autosomal dominant pattern and caused by the abnormal expression of DUX4 in skeletal muscle. The DUX4 transcription factor has DNA binding domains similar to several paired class homeotic transcription factors, but only myogenic factors PAX3 and PAX7 rescue cell viability when co-expressed with DUX4 in mouse myoblasts. This observation suggests competition for DNA binding sites in satellite cells might limit muscle repair and may be one aspect of DUX4-associated myotoxicity. The competition hypothesis requires that DUX4 and PAX3/7 be expressed in the same cells at some point during development or in adult tissues. We modeled myogenesis using human isogenic iPS and ES cells and examined expression patterns of DUX4, PAX3, and PAX7 to determine if conditions that promote PAX3 and PAX7 expression in cell culture also promote DUX4 expression in the same cells. Methods Isogenic iPSCs were generated from human fibroblasts of two FSHD-affected individuals with somatic mosaicism. Clones containing the shortened FSHD-causing D4Z4 array or the long non-pathogenic array were isolated from the same individuals. We also examined myogenesis in commercially available hES cell lines derived from FSHD-affected and non-affected embryos. DUX4, PAX3, and PAX7 messenger RNAs (mRNAs) were quantified during a 40-day differentiation protocol, and antibodies were used to identify cell types in different stages of differentiation to determine if DUX4 and PAX3 or PAX7 are present in the same cells. Results Human iPS and ES cells differentiated into skeletal myocytes as evidenced by Titin positive multinucleated fibers appearing toward the end of a 40-day differentiation protocol. PAX3 and PAX7 were expressed at similar times during differentiation, and DUX4 positive nuclei were seen at terminal stages of differentiation in cells containing the short D4Z4 arrays. Nuclei that expressed both DUX4 and PAX3, or DUX4 and PAX7 were not observed after examining immunostained nuclei at five different time points during myogenic differentiation of pluripotent cells. Conclusions We conclude that DUX4, PAX3, and PAX7 have distinct expression patterns during myogenic differentiation of stem cells. Our findings are consistent with the hypothesis that muscle damage in FSHD is due to DUX4-mediated toxicity causing destruction of terminally differentiated myofibers. While these studies examine DUX4, PAX3, and PAX7 expression patterns during stem cell myogenesis, they should not be generalized to tissue repair in adult muscle tissue. Electronic supplementary material The online version of this article (doi:10.1186/s13395-017-0130-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Premi Haynes
- Division of Genetic Medicine, Department of Pediatrics, University of Washington, Campus Box 358056, 850 Republican Street, Room N416, Seattle, WA, 98109, USA
| | - Kelly Kernan
- Division of Genetic Medicine, Department of Pediatrics, University of Washington, Campus Box 358056, 850 Republican Street, Room N416, Seattle, WA, 98109, USA
| | - Suk-Lin Zhou
- Division of Genetic Medicine, Department of Pediatrics, University of Washington, Campus Box 358056, 850 Republican Street, Room N416, Seattle, WA, 98109, USA
| | - Daniel G Miller
- Division of Genetic Medicine, Department of Pediatrics, University of Washington, Campus Box 358056, 850 Republican Street, Room N416, Seattle, WA, 98109, USA.
| |
Collapse
|
72
|
Zhu XJ, Yuan X, Wang M, Fang Y, Liu Y, Zhang X, Yang X, Li Y, Li J, Li F, Dai ZM, Qiu M, Zhang Z, Zhang Z. A Wnt/Notch/Pax7 signaling network supports tissue integrity in tongue development. J Biol Chem 2017; 292:9409-9419. [PMID: 28438836 DOI: 10.1074/jbc.m117.789438] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2017] [Revised: 04/21/2017] [Indexed: 01/07/2023] Open
Abstract
The tongue is one of the major structures involved in human food intake and speech. Tongue malformations such as aglossia, microglossia, and ankyloglossia are congenital birth defects, greatly affecting individuals' quality of life. However, the molecular basis of the tissue-tissue interactions that ensure tissue morphogenesis to form a functional tongue remains largely unknown. Here we show that ShhCre -mediated epithelial deletion of Wntless (Wls), the key regulator for intracellular Wnt trafficking, leads to lingual hypoplasia in mice. Disruption of epithelial Wnt production by Wls deletion in epithelial cells led to a failure in lingual epidermal stratification and loss of the lamina propria and the underlying superior longitudinal muscle in developing mouse tongues. These defective phenotypes resulted from a reduction in epithelial basal cells positive for the basal epidermal marker protein p63 and from impaired proliferation and differentiation in connective tissue and paired box 3 (Pax3)- and Pax7-positive muscle progenitor cells. We also found that epithelial Wnt production is required for activation of the Notch signaling pathway, which promotes proliferation of myogenic progenitor cells. Notch signaling in turn negatively regulated Wnt signaling during tongue morphogenesis. We further show that Pax7 is a direct Notch target gene in the embryonic tongue. In summary, our findings demonstrate a key role for the lingual epithelial signals in supporting the integrity of the lamina propria and muscular tissue during tongue development and that a Wnt/Notch/Pax7 genetic hierarchy is involved in this development.
Collapse
Affiliation(s)
- Xiao-Jing Zhu
- From the Institute of Life Sciences, College of Life and Environmental Science, Key Laboratory of Mammalian Organogenesis and Regeneration, Hangzhou Normal University, Zhejiang 310036, China and
| | - Xueyan Yuan
- From the Institute of Life Sciences, College of Life and Environmental Science, Key Laboratory of Mammalian Organogenesis and Regeneration, Hangzhou Normal University, Zhejiang 310036, China and
| | - Min Wang
- From the Institute of Life Sciences, College of Life and Environmental Science, Key Laboratory of Mammalian Organogenesis and Regeneration, Hangzhou Normal University, Zhejiang 310036, China and
| | - Yukun Fang
- From the Institute of Life Sciences, College of Life and Environmental Science, Key Laboratory of Mammalian Organogenesis and Regeneration, Hangzhou Normal University, Zhejiang 310036, China and
| | - Yudong Liu
- From the Institute of Life Sciences, College of Life and Environmental Science, Key Laboratory of Mammalian Organogenesis and Regeneration, Hangzhou Normal University, Zhejiang 310036, China and
| | - Xiaoyun Zhang
- From the Institute of Life Sciences, College of Life and Environmental Science, Key Laboratory of Mammalian Organogenesis and Regeneration, Hangzhou Normal University, Zhejiang 310036, China and
| | - Xueqin Yang
- From the Institute of Life Sciences, College of Life and Environmental Science, Key Laboratory of Mammalian Organogenesis and Regeneration, Hangzhou Normal University, Zhejiang 310036, China and
| | - Yan Li
- From the Institute of Life Sciences, College of Life and Environmental Science, Key Laboratory of Mammalian Organogenesis and Regeneration, Hangzhou Normal University, Zhejiang 310036, China and
| | - Jianying Li
- From the Institute of Life Sciences, College of Life and Environmental Science, Key Laboratory of Mammalian Organogenesis and Regeneration, Hangzhou Normal University, Zhejiang 310036, China and
| | - Feixue Li
- From the Institute of Life Sciences, College of Life and Environmental Science, Key Laboratory of Mammalian Organogenesis and Regeneration, Hangzhou Normal University, Zhejiang 310036, China and
| | - Zhong-Min Dai
- From the Institute of Life Sciences, College of Life and Environmental Science, Key Laboratory of Mammalian Organogenesis and Regeneration, Hangzhou Normal University, Zhejiang 310036, China and
| | - Mengsheng Qiu
- From the Institute of Life Sciences, College of Life and Environmental Science, Key Laboratory of Mammalian Organogenesis and Regeneration, Hangzhou Normal University, Zhejiang 310036, China and
| | - Ze Zhang
- the Department of Ophthalmology, Tulane Medical Center, Tulane University, New Orleans, Louisiana 70115
| | - Zunyi Zhang
- From the Institute of Life Sciences, College of Life and Environmental Science, Key Laboratory of Mammalian Organogenesis and Regeneration, Hangzhou Normal University, Zhejiang 310036, China and
| |
Collapse
|
73
|
Jiao S, Wu Z, Tan X, Sui Y, Wang L, You F. Characterization of pax3a and pax3b genes in artificially induced polyploid and gynogenetic olive flounder (Paralichthys olivaceus) during embryogenesis. FISH PHYSIOLOGY AND BIOCHEMISTRY 2017; 43:385-395. [PMID: 27677482 DOI: 10.1007/s10695-016-0294-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/03/2016] [Accepted: 09/21/2016] [Indexed: 06/06/2023]
Abstract
Although chromosome set manipulation techniques including polyploidy induction and gynogentic induction in flatfish are becoming increasingly mature, there exists a poor understanding of their effects on embryonic development. PAX3 plays crucial roles during embryonic myogenesis and neurogenesis. In olive flounder (Paralichthys olivaceus), there are two duplicated pax3 genes (pax3a, pax3b), and both of them are expressed in the brain and muscle regions with some subtle regional differences. We utilized pax3a and pax3b as indicators to preliminarily investigate whether chromosome set manipulation affects embryonic neurogenesis and myogenesis using whole-mount in situ hybridization. In the polyploid induction groups, 94 % of embryos in the triploid induction group had normal pax3a/3b expression patterns; however, 45 % of embryos in the tetraploid induction group showed abnormal pax3a/3b expression patterns from the tailbud formation stage to the hatching stage. Therefore, the artificial induction of triploidy and tetraploidy had a small or a moderate effect on flounder embryonic myogenesis and neurogenesis, respectively. In the gynogenetic induction groups, 87 % of embryos in the meiogynogenetic diploid induction group showed normal pax3a/3b expression patterns. However, almost 100 % of embryos in the gynogenetic haploid induction group and 63 % of embryos in the mitogynogenetic diploid induction group showed abnormal pax3a/3b expression patterns. Therefore, the induction of gynogenetic haploidy and mitogynogenetic diploidy had large effects on flounder embryonic myogenesis and neurogenesis. In conclusion, the differential expression of pax3a and pax3b may provide new insights for consideration of fish chromosome set manipulation.
Collapse
Affiliation(s)
- Shuang Jiao
- Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, 7 Nanhai Road, Qingdao, 266071, Shandong, People's Republic of China
- Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, 7 Nanhai Road, Qingdao, 266071, Shandong, People's Republic of China
| | - Zhihao Wu
- Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, 7 Nanhai Road, Qingdao, 266071, Shandong, People's Republic of China
- Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, 7 Nanhai Road, Qingdao, 266071, Shandong, People's Republic of China
| | - Xungang Tan
- Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, 7 Nanhai Road, Qingdao, 266071, Shandong, People's Republic of China
- Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, 7 Nanhai Road, Qingdao, 266071, Shandong, People's Republic of China
| | - Yulei Sui
- Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, 7 Nanhai Road, Qingdao, 266071, Shandong, People's Republic of China
- Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, 7 Nanhai Road, Qingdao, 266071, Shandong, People's Republic of China
- University of Chinese Academy of Sciences, Beijing, 100049, People's Republic of China
| | - Lijuan Wang
- Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, 7 Nanhai Road, Qingdao, 266071, Shandong, People's Republic of China
- Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, 7 Nanhai Road, Qingdao, 266071, Shandong, People's Republic of China
- University of Chinese Academy of Sciences, Beijing, 100049, People's Republic of China
| | - Feng You
- Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, 7 Nanhai Road, Qingdao, 266071, Shandong, People's Republic of China.
- Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, 7 Nanhai Road, Qingdao, 266071, Shandong, People's Republic of China.
| |
Collapse
|
74
|
Nassari S, Duprez D, Fournier-Thibault C. Non-myogenic Contribution to Muscle Development and Homeostasis: The Role of Connective Tissues. Front Cell Dev Biol 2017; 5:22. [PMID: 28386539 PMCID: PMC5362625 DOI: 10.3389/fcell.2017.00022] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Accepted: 03/07/2017] [Indexed: 12/22/2022] Open
Abstract
Skeletal muscles belong to the musculoskeletal system, which is composed of bone, tendon, ligament and irregular connective tissue, and closely associated with motor nerves and blood vessels. The intrinsic molecular signals regulating myogenesis have been extensively investigated. However, muscle development, homeostasis and regeneration require interactions with surrounding tissues and the cellular and molecular aspects of this dialogue have not been completely elucidated. During development and adult life, myogenic cells are closely associated with the different types of connective tissue. Connective tissues are defined as specialized (bone and cartilage), dense regular (tendon and ligament) and dense irregular connective tissue. The role of connective tissue in muscle morphogenesis has been investigated, thanks to the identification of transcription factors that characterize the different types of connective tissues. Here, we review the development of the various connective tissues in the context of the musculoskeletal system and highlight their important role in delivering information necessary for correct muscle morphogenesis, from the early step of myoblast differentiation to the late stage of muscle maturation. Interactions between muscle and connective tissue are also critical in the adult during muscle regeneration, as impairment of the regenerative potential after injury or in neuromuscular diseases results in the progressive replacement of the muscle mass by fibrotic tissue. We conclude that bi-directional communication between muscle and connective tissue is critical for a correct assembly of the musculoskeletal system during development as well as to maintain its homeostasis in the adult.
Collapse
Affiliation(s)
- Sonya Nassari
- Developmental Biology Laboratory, IBPS, Centre National de la Recherche Scientifique UMR7622, Institut National de la Santé Et de la Recherche Médicale U1156, Université Pierre et Marie Curie, Sorbonne Universités Paris, France
| | - Delphine Duprez
- Developmental Biology Laboratory, IBPS, Centre National de la Recherche Scientifique UMR7622, Institut National de la Santé Et de la Recherche Médicale U1156, Université Pierre et Marie Curie, Sorbonne Universités Paris, France
| | - Claire Fournier-Thibault
- Developmental Biology Laboratory, IBPS, Centre National de la Recherche Scientifique UMR7622, Institut National de la Santé Et de la Recherche Médicale U1156, Université Pierre et Marie Curie, Sorbonne Universités Paris, France
| |
Collapse
|
75
|
Abstract
Skeletal muscle stem cells (MuSCs) derived from the somatic mesoderm play a critical role in successful muscle regeneration following injury and trauma. MuSCs have been found to undergo rapid changes in metabolism following a change in cell state, such as that which occurs during the transition from quiescence to an actively proliferating state. There is mounting evidence that metabolism is critically important in the regulation of quiescence, activation, and differentiation and thus the development of new techniques that aim to further probe the metabolism of MuSCs is essential. The Seahorse XF Bioanalyzer is a powerful tool that simultaneously measures the extracellular rate of change in oxygen partial pressure and pH, providing a method to measure mitochondrial respiration and lactate production. In this chapter, we describe the use of key metabolic inhibitors that allow for the investigation of mitochondrial substrate utilization in primary MuSCs.
Collapse
Affiliation(s)
- C Hai Ly
- Stem Cell Metabolism and Regenerative Medicine Group, Basic and Clinical Myology Laboratory, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - James G Ryall
- Stem Cell Metabolism and Regenerative Medicine Group, Basic and Clinical Myology Laboratory, Department of Physiology, School of Biomedical Sciences, The University of Melbourne, Melbourne, VIC, 3010, Australia.
| |
Collapse
|
76
|
Helinska A, Krupa M, Archacka K, Czerwinska AM, Streminska W, Janczyk-Ilach K, Ciemerych MA, Grabowska I. Myogenic potential of mouse embryonic stem cells lacking functional Pax7 tested in vitro by 5-azacitidine treatment and in vivo in regenerating skeletal muscle. Eur J Cell Biol 2016; 96:47-60. [PMID: 28017376 DOI: 10.1016/j.ejcb.2016.12.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2016] [Revised: 12/05/2016] [Accepted: 12/05/2016] [Indexed: 12/25/2022] Open
Abstract
Regeneration of skeletal muscle relies on the presence of satellite cells. Satellite cells deficiency accompanying some degenerative diseases is the reason for the search for the "replacement cells" that can be used in the muscle therapies. Due to their unique properties embryonic stem cells (ESCs), as well as myogenic cells derived from them, are considered as a promising source of therapeutic cells. Among the factors crucial for the specification of myogenic precursor cells is Pax7 that sustains proper function of satellite cells. In our previous studies we showed that ESCs lacking functional Pax7 are able to form myoblasts in vitro when differentiated within embryoid bodies and their outgrowths. In the current study we showed that ESCs lacking functional Pax7, cultured in vitro in monolayer in the medium supplemented with horse serum and 5azaC, expressed higher levels of factors associated with myogenesis, such as Pdgfra, Pax3, Myf5, and MyoD. Importantly, skeletal myosin immunolocalization confirmed that myogenic differentiation of ESCs was more effective in case of cells lacking Pax7. Our in vivo studies showed that ESCs transplanted into regenerating skeletal muscles were detectable at day 7 of regeneration and the number of Pax7-/- ESCs detected was significantly higher than of control cells. Our results support the concept that lack of functional Pax7 promotes proliferation of differentiating ESCs and for this reason more of them can turn into myogenic lineage.
Collapse
Affiliation(s)
- Anita Helinska
- Department of Cytology, Institute of Zoology, Faculty of Biology, University of Warsaw, Poland
| | - Maciej Krupa
- Department of Cytology, Institute of Zoology, Faculty of Biology, University of Warsaw, Poland
| | - Karolina Archacka
- Department of Cytology, Institute of Zoology, Faculty of Biology, University of Warsaw, Poland
| | - Areta M Czerwinska
- Department of Cytology, Institute of Zoology, Faculty of Biology, University of Warsaw, Poland
| | - Wladyslawa Streminska
- Department of Cytology, Institute of Zoology, Faculty of Biology, University of Warsaw, Poland
| | - Katarzyna Janczyk-Ilach
- Department of Cytology, Institute of Zoology, Faculty of Biology, University of Warsaw, Poland
| | - Maria A Ciemerych
- Department of Cytology, Institute of Zoology, Faculty of Biology, University of Warsaw, Poland
| | - Iwona Grabowska
- Department of Cytology, Institute of Zoology, Faculty of Biology, University of Warsaw, Poland.
| |
Collapse
|
77
|
Besson V, Kyryachenko S, Janich P, Benitah SA, Marazzi G, Sassoon D. Expression Analysis of the Stem Cell Marker Pw1/Peg3 Reveals a CD34 Negative Progenitor Population in the Hair Follicle. Stem Cells 2016; 35:1015-1027. [PMID: 27862634 DOI: 10.1002/stem.2540] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Revised: 09/10/2016] [Accepted: 10/16/2016] [Indexed: 12/30/2022]
Abstract
Pw1/Peg3 is a parentally imprinted gene expressed in adult stem cells in every tissue thus far examined including the stem cells of the hair follicle. Using a Pw1/Peg3 reporter mouse, we carried out a detailed dissection of the stem cells in the bulge, which is a major stem cell compartment of the hair follicle in mammalian skin. We observed that PW1/Peg3 expression initiates upon placode formation during fetal development, coincident with the establishment of the bulge stem cells. In the adult, we observed that PW1/Peg3 expression is found in both CD34+ and CD34- populations of bulge stem cells. We demonstrate that both populations can give rise to new hair follicles, reconstitute their niche, and self-renew. These results demonstrate that PW1/Peg3 is a reliable marker of the full population of follicle stem cells and reveal a novel CD34- bulge stem-cell population. Stem Cells 2017;35:1015-1027.
Collapse
Affiliation(s)
- Vanessa Besson
- Stem Cells and Regenerative Medicine, Institute of Cardiometabolism and Nutrition (ICAN), UMRS 1166 INSERM, University of Pierre and Marie Curie Paris VI, Paris, France
| | - Sergiy Kyryachenko
- Stem Cells and Regenerative Medicine, Institute of Cardiometabolism and Nutrition (ICAN), UMRS 1166 INSERM, University of Pierre and Marie Curie Paris VI, Paris, France
| | - Peggy Janich
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Salvador A Benitah
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, Spain.,ICREA, Catalan Institution for Research and Advanced Studies, Barcelona, Spain.,Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain
| | - Giovanna Marazzi
- Stem Cells and Regenerative Medicine, Institute of Cardiometabolism and Nutrition (ICAN), UMRS 1166 INSERM, University of Pierre and Marie Curie Paris VI, Paris, France
| | - David Sassoon
- Stem Cells and Regenerative Medicine, Institute of Cardiometabolism and Nutrition (ICAN), UMRS 1166 INSERM, University of Pierre and Marie Curie Paris VI, Paris, France
| |
Collapse
|
78
|
Pipalia TG, Koth J, Roy SD, Hammond CL, Kawakami K, Hughes SM. Cellular dynamics of regeneration reveals role of two distinct Pax7 stem cell populations in larval zebrafish muscle repair. Dis Model Mech 2016; 9:671-84. [PMID: 27149989 PMCID: PMC4920144 DOI: 10.1242/dmm.022251] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2015] [Accepted: 04/27/2016] [Indexed: 12/25/2022] Open
Abstract
Heterogeneity of stem cells or their niches is likely to influence tissue regeneration. Here we reveal stem/precursor cell diversity during wound repair in larval zebrafish somitic body muscle using time-lapse 3D confocal microscopy on reporter lines. Skeletal muscle with incision wounds rapidly regenerates both slow and fast muscle fibre types. A swift immune response is followed by an increase in cells at the wound site, many of which express the muscle stem cell marker Pax7. Pax7(+) cells proliferate and then undergo terminal differentiation involving Myogenin accumulation and subsequent loss of Pax7 followed by elongation and fusion to repair fast muscle fibres. Analysis of pax7a and pax7b transgenic reporter fish reveals that cells expressing each of the duplicated pax7 genes are distinctly localised in uninjured larvae. Cells marked by pax7a only or by both pax7a and pax7b enter the wound rapidly and contribute to muscle wound repair, but each behaves differently. Low numbers of pax7a-only cells form nascent fibres. Time-lapse microscopy revealed that the more numerous pax7b-marked cells frequently fuse to pre-existing fibres, contributing more strongly than pax7a-only cells to repair of damaged fibres. pax7b-marked cells are more often present in rows of aligned cells that are observed to fuse into a single fibre, but more rarely contribute to nascent regenerated fibres. Ablation of a substantial portion of nitroreductase-expressing pax7b cells with metronidazole prior to wounding triggered rapid pax7a-only cell accumulation, but this neither inhibited nor augmented pax7a-only cell-derived myogenesis and thus altered the cellular repair dynamics during wound healing. Moreover, pax7a-only cells did not regenerate pax7b cells, suggesting a lineage distinction. We propose a modified founder cell and fusion-competent cell model in which pax7a-only cells initiate fibre formation and pax7b cells contribute to fibre growth. This newly discovered cellular complexity in muscle wound repair raises the possibility that distinct populations of myogenic cells contribute differentially to repair in other vertebrates.
Collapse
Affiliation(s)
- Tapan G Pipalia
- Randall Division of Cell and Molecular Biophysics, Guy's Campus, King's College London, London SE1 1UL, UK
| | - Jana Koth
- Randall Division of Cell and Molecular Biophysics, Guy's Campus, King's College London, London SE1 1UL, UK Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, Oxford University, Oxford OX3 9DS, UK
| | - Shukolpa D Roy
- Randall Division of Cell and Molecular Biophysics, Guy's Campus, King's College London, London SE1 1UL, UK
| | - Christina L Hammond
- Randall Division of Cell and Molecular Biophysics, Guy's Campus, King's College London, London SE1 1UL, UK
| | - Koichi Kawakami
- Division of Molecular and Developmental Biology, National Institute of Genetics, and Department of Genetics, SOKENDAI (The Graduate University for Advanced Studies), Mishima, Shizuoka 411-8540, Japan
| | - Simon M Hughes
- Randall Division of Cell and Molecular Biophysics, Guy's Campus, King's College London, London SE1 1UL, UK
| |
Collapse
|
79
|
Berti F, Nogueira JM, Wöhrle S, Sobreira DR, Hawrot K, Dietrich S. Time course and side-by-side analysis of mesodermal, pre-myogenic, myogenic and differentiated cell markers in the chicken model for skeletal muscle formation. J Anat 2016; 227:361-82. [PMID: 26278933 PMCID: PMC4560570 DOI: 10.1111/joa.12353] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/12/2015] [Indexed: 12/11/2022] Open
Abstract
The chicken is a well-established model for amniote (including human) skeletal muscle formation because the developmental anatomy of chicken skeletal muscle matches that of mammals. The accessibility of the chicken in the egg as well as the sequencing of its genome and novel molecular techniques have raised the profile of this model. Over the years, a number of regulatory and marker genes have been identified that are suited to monitor the progress of skeletal myogenesis both in wildtype and in experimental embryos. However, in the various studies, differing markers at different stages of development have been used. Moreover, contradictory results on the hierarchy of regulatory factors are now emerging, and clearly, factors need to be able to cooperate. Thus, a reference paper describing in detail and side-by-side the time course of marker gene expression during avian myogenesis is needed. We comparatively analysed onset and expression patterns of the key markers for the chicken immature paraxial mesoderm, for muscle-competent cells, for cells committed to myogenesis and for cells entering terminal differentiation. We performed this analysis from stages when the first paraxial mesoderm is being laid down to the stage when mesoderm formation comes to a conclusion. Our data show that, although the sequence of marker gene expression is the same at the various stages of development, the timing of the expression onset is quite different. Moreover, marker gene expression in myogenic cells being deployed from the dorsomedial and ventrolateral lips of the dermomyotome is different from those being deployed from the rostrocaudal lips, suggesting different molecular programs. Furthermore, expression of Myosin Heavy Chain genes is overlapping but different along the length of a myotube. Finally, Mef2c is the most likely partner of Mrf proteins, and, in contrast to the mouse and more alike frog and zebrafish fish, chicken Mrf4 is co-expressed with MyoG as cells enter terminal differentiation.
Collapse
Affiliation(s)
- Federica Berti
- Institute for Biomedical and Biomolecular Science (IBBS), School of Pharmacy and Biomedical Sciences, University of Portsmouth, Portsmouth, UK
| | - Júlia Meireles Nogueira
- Institute for Biomedical and Biomolecular Science (IBBS), School of Pharmacy and Biomedical Sciences, University of Portsmouth, Portsmouth, UK.,Instituto de Ciências Biológicas, Departamento de Morfologia, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Minas Gerais, Brazil
| | - Svenja Wöhrle
- Institute for Biomedical and Biomolecular Science (IBBS), School of Pharmacy and Biomedical Sciences, University of Portsmouth, Portsmouth, UK
| | - Débora Rodrigues Sobreira
- Institute for Biomedical and Biomolecular Science (IBBS), School of Pharmacy and Biomedical Sciences, University of Portsmouth, Portsmouth, UK.,Department of Human Genetics, University of Chicago, Chicago, IL, USA
| | - Katarzyna Hawrot
- Institute for Biomedical and Biomolecular Science (IBBS), School of Pharmacy and Biomedical Sciences, University of Portsmouth, Portsmouth, UK
| | - Susanne Dietrich
- Institute for Biomedical and Biomolecular Science (IBBS), School of Pharmacy and Biomedical Sciences, University of Portsmouth, Portsmouth, UK
| |
Collapse
|
80
|
Dumont NA, Bentzinger CF, Sincennes MC, Rudnicki MA. Satellite Cells and Skeletal Muscle Regeneration. Compr Physiol 2016; 5:1027-59. [PMID: 26140708 DOI: 10.1002/cphy.c140068] [Citation(s) in RCA: 425] [Impact Index Per Article: 53.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Skeletal muscles are essential for vital functions such as movement, postural support, breathing, and thermogenesis. Muscle tissue is largely composed of long, postmitotic multinucleated fibers. The life-long maintenance of muscle tissue is mediated by satellite cells, lying in close proximity to the muscle fibers. Muscle satellite cells are a heterogeneous population with a small subset of muscle stem cells, termed satellite stem cells. Under homeostatic conditions all satellite cells are poised for activation by stimuli such as physical trauma or growth signals. After activation, satellite stem cells undergo symmetric divisions to expand their number or asymmetric divisions to give rise to cohorts of committed satellite cells and thus progenitors. Myogenic progenitors proliferate, and eventually differentiate through fusion with each other or to damaged fibers to reconstitute fiber integrity and function. In the recent years, research has begun to unravel the intrinsic and extrinsic mechanisms controlling satellite cell behavior. Nonetheless, an understanding of the complex cellular and molecular interactions of satellite cells with their dynamic microenvironment remains a major challenge, especially in pathological conditions. The goal of this review is to comprehensively summarize the current knowledge on satellite cell characteristics, functions, and behavior in muscle regeneration and in pathological conditions.
Collapse
Affiliation(s)
- Nicolas A Dumont
- Sprott Centre for Stem Cell Research, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | - C Florian Bentzinger
- Sprott Centre for Stem Cell Research, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada.,Nestlé Institute of Health Sciences, EPFL Campus, Lausanne, Switzerland
| | - Marie-Claude Sincennes
- Sprott Centre for Stem Cell Research, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | - Michael A Rudnicki
- Sprott Centre for Stem Cell Research, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada.,Faculty of Medicine, Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario, Canada
| |
Collapse
|
81
|
Abou-Khalil R, Yang F, Lieu S, Julien A, Perry J, Pereira C, Relaix F, Miclau T, Marcucio R, Colnot C. Role of muscle stem cells during skeletal regeneration. Stem Cells 2016; 33:1501-11. [PMID: 25594525 DOI: 10.1002/stem.1945] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2014] [Accepted: 12/07/2014] [Indexed: 01/20/2023]
Abstract
Although the importance of muscle in skeletal regeneration is well recognized clinically, the mechanisms by which muscle supports bone repair have remained elusive. Muscle flaps are often used to cover the damaged bone after traumatic injury yet their contribution to bone healing is not known. Here, we show that direct bone-muscle interactions are required for periosteum activation and callus formation, and that muscle grafts provide a source of stem cells for skeletal regeneration. We investigated the role of satellite cells, the muscle stem cells. Satellite cells loss in Pax7(-/-) mice and satellite cell ablation in Pax7(Cre) (ERT) (2/) (+) ;DTA(f/f) mice impaired bone regeneration. Although satellite cells did not contribute as a large source of cells endogenously, they exhibited a potential to contribute to bone repair after transplantation. The fracture healing phenotype in Pax7(Cre) (ERT) (2/) (+) ;DTA(f/f) mice was associated with decreased bone morphogenetic proteins (BMPs), insulin-like growth factor 1, and fibroblast growth factor 2 expression that are normally upregulated in response to fracture in satellite cells. Exogenous rhBMP2 improved bone healing in Pax7(Cre) (ERT) (2/) (+) ;DTA(f/f) mice further supporting the role of satellite cells as a source of growth factors. These results provide the first functional evidence for a direct contribution of muscle to bone regeneration with important clinical implications as it may impact the use of muscle flaps, muscle stem cells, and growth factors in orthopedic applications.
Collapse
Affiliation(s)
- Rana Abou-Khalil
- INSERM UMR1163, Université Paris Descartes-Sorbonne Paris Cité, Institut Imagine, Hôpital Necker Enfants Malades, Paris, France
| | | | | | | | | | | | | | | | | | | |
Collapse
|
82
|
Cesarini E, Mozzetta C, Marullo F, Gregoretti F, Gargiulo A, Columbaro M, Cortesi A, Antonelli L, Di Pelino S, Squarzoni S, Palacios D, Zippo A, Bodega B, Oliva G, Lanzuolo C. Lamin A/C sustains PcG protein architecture, maintaining transcriptional repression at target genes. J Cell Biol 2016; 211:533-51. [PMID: 26553927 PMCID: PMC4639869 DOI: 10.1083/jcb.201504035] [Citation(s) in RCA: 83] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Reduction of lamin A/C, which is evolutionarily required for the modulation of Polycomb group (PcG) protein–dependent transcriptional repression by sustaining PcG protein nuclear architecture, leads to PcG protein diffusion and to muscle differentiation. Beyond its role in providing structure to the nuclear envelope, lamin A/C is involved in transcriptional regulation. However, its cross talk with epigenetic factors—and how this cross talk influences physiological processes—is still unexplored. Key epigenetic regulators of development and differentiation are the Polycomb group (PcG) of proteins, organized in the nucleus as microscopically visible foci. Here, we show that lamin A/C is evolutionarily required for correct PcG protein nuclear compartmentalization. Confocal microscopy supported by new algorithms for image analysis reveals that lamin A/C knock-down leads to PcG protein foci disassembly and PcG protein dispersion. This causes detachment from chromatin and defects in PcG protein–mediated higher-order structures, thereby leading to impaired PcG protein repressive functions. Using myogenic differentiation as a model, we found that reduced levels of lamin A/C at the onset of differentiation led to an anticipation of the myogenic program because of an alteration of PcG protein–mediated transcriptional repression. Collectively, our results indicate that lamin A/C can modulate transcription through the regulation of PcG protein epigenetic factors.
Collapse
Affiliation(s)
- Elisa Cesarini
- Consiglio Nazionale delle Ricerche Institute of Cellular Biology and Neurobiology, Istituto di Ricovero e Cura a Carattere Scientifico Fondazione Santa Lucia, 00143 Rome, Italy
| | - Chiara Mozzetta
- Consiglio Nazionale delle Ricerche Institute of Cellular Biology and Neurobiology, Istituto di Ricovero e Cura a Carattere Scientifico Fondazione Santa Lucia, 00143 Rome, Italy
| | - Fabrizia Marullo
- Consiglio Nazionale delle Ricerche Institute of Cellular Biology and Neurobiology, Istituto di Ricovero e Cura a Carattere Scientifico Fondazione Santa Lucia, 00143 Rome, Italy
| | - Francesco Gregoretti
- Consiglio Nazionale delle Ricerche Institute for High Performance Computing and Networking, 80131 Naples, Italy
| | - Annagiusi Gargiulo
- Consiglio Nazionale delle Ricerche Institute of Cellular Biology and Neurobiology, Istituto di Ricovero e Cura a Carattere Scientifico Fondazione Santa Lucia, 00143 Rome, Italy
| | - Marta Columbaro
- Struttura Complessa Laboratorio Biologia Cellulare Muscoloscheletrica, Istituto Ortopedico Rizzoli, 40136 Bologna, Italy
| | - Alice Cortesi
- Istituto Nazionale Genetica Molecolare Romeo ed Enrica Invernizzi, 20122 Milan, Italy
| | - Laura Antonelli
- Consiglio Nazionale delle Ricerche Institute for High Performance Computing and Networking, 80131 Naples, Italy
| | - Simona Di Pelino
- Istituto Nazionale Genetica Molecolare Romeo ed Enrica Invernizzi, 20122 Milan, Italy
| | - Stefano Squarzoni
- Struttura Complessa Laboratorio Biologia Cellulare Muscoloscheletrica, Istituto Ortopedico Rizzoli, 40136 Bologna, Italy Consiglio Nazionale delle Ricerche Institute of Molecular Genetics, 40136 Bologna, Italy
| | - Daniela Palacios
- Istituto di Ricovero e Cura a Carattere Scientifico Fondazione Santa Lucia, 00143 Rome, Italy
| | - Alessio Zippo
- Istituto Nazionale Genetica Molecolare Romeo ed Enrica Invernizzi, 20122 Milan, Italy
| | - Beatrice Bodega
- Istituto Nazionale Genetica Molecolare Romeo ed Enrica Invernizzi, 20122 Milan, Italy
| | - Gennaro Oliva
- Consiglio Nazionale delle Ricerche Institute for High Performance Computing and Networking, 80131 Naples, Italy
| | - Chiara Lanzuolo
- Consiglio Nazionale delle Ricerche Institute of Cellular Biology and Neurobiology, Istituto di Ricovero e Cura a Carattere Scientifico Fondazione Santa Lucia, 00143 Rome, Italy
| |
Collapse
|
83
|
Mayeuf-Louchart A, Montarras D, Bodin C, Kume T, Vincent SD, Buckingham M. Endothelial cell specification in the somite is compromised in Pax3-positive progenitors of Foxc1/2 conditional mutants, with loss of forelimb myogenesis. Development 2016; 143:872-9. [PMID: 26839363 DOI: 10.1242/dev.128017] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Accepted: 01/23/2016] [Indexed: 11/20/2022]
Abstract
Pax3 and Foxc2 have been shown genetically to mutually repress each other in the mouse somite. Perturbation of this balance in multipotent cells of the dermomyotome influences cell fate; upregulation of Foxc2 favours a vascular fate, whereas higher levels of Pax3 lead to myogenesis. Foxc1 has overlapping functions with Foxc2. In Foxc1/2 double-mutant embryos, somitogenesis is severely affected, precluding analysis of somite derivatives. We have adopted a conditional approach whereby mutations in Foxc1 and Foxc2 genes were targeted to Pax3-expressing cells. Inclusion of a conditional reporter allele in the crosses made it possible to follow cells that had expressed Pax3. At the forelimb level, endothelial and myogenic cells migrate from adjacent somites into the limb bud. This population of endothelial cells is compromised in the double mutant, whereas excessive production of myogenic cells is observed in the trunk. However, strikingly, myogenic progenitors fail to enter the limbs, leading to the absence of skeletal muscle. Pax3-positive migratory myogenic progenitors, marked by expression of Lbx1, are specified in the somite at forelimb level, but endothelial progenitors are absent. The myogenic progenitors do not die, but differentiate prematurely adjacent to the somite. We conclude that the small proportion of somite-derived endothelial cells in the limb is required for the migration of myogenic limb progenitors.
Collapse
Affiliation(s)
- Alicia Mayeuf-Louchart
- CNRS UMR 3738, Department of Developmental and Stem Cell Biology, Institut Pasteur, 28 Rue du Dr Roux, Paris 75015, France
| | - Didier Montarras
- CNRS UMR 3738, Department of Developmental and Stem Cell Biology, Institut Pasteur, 28 Rue du Dr Roux, Paris 75015, France
| | - Catherine Bodin
- CNRS UMR 3738, Department of Developmental and Stem Cell Biology, Institut Pasteur, 28 Rue du Dr Roux, Paris 75015, France
| | - Tsutomu Kume
- Feinberg Cardiovascular Research Institute, Northwestern University School of Medicine, Chicago, IL 60611, USA
| | - Stéphane D Vincent
- CNRS UMR 3738, Department of Developmental and Stem Cell Biology, Institut Pasteur, 28 Rue du Dr Roux, Paris 75015, France
| | - Margaret Buckingham
- CNRS UMR 3738, Department of Developmental and Stem Cell Biology, Institut Pasteur, 28 Rue du Dr Roux, Paris 75015, France
| |
Collapse
|
84
|
Abstract
Congenital hydrocephalus is a common birth-defect whose developmental origins are poorly understood. Pax3-null mutants show defects in myogenesis, neural tube closure, neural crest morphogenesis, and heart development that, consequently, results in embryonic lethality. Here we demonstrate that conditional deletion of the mouse Pax3 transcription factor results in fully-penetrant congenital obstructive hydrocephalus. To identify the role of Pax3 during cranial development, we deleted Pax3 within the neuroepithelium (via Pax7−Cre), in the neural crest (via P0-Cre), and in both the neuroepithelium and the neural crest (via Wnt1-Cre). Only conditional mutants generated using Pax7−Cre or Wnt1-Cre developed early onset congenital hydrocephalus due to stenosis of the third ventricle, suggesting that loss of neuroepithelial Pax3 is sufficient to disturb third ventricle morphogenesis. Dilation of lateral ventricles occurs as early as E14.5, and lineage-mapping revealed that the neuroepithelial cells in the conditional mutants are present, but fail to undergo normal differentiation at the stenotic site. Concomitant with a narrowing of the mutant third ventricle, we detected ectopic apoptosis, reduced proliferation, and abnormal β-catenin localization. Furthermore, consistent with the overlapping expression pattern of Pax3 and Pax7 in early cranial neuroepithelium, we demonstrated a combinatorial role, as compound Pax3/Pax7 heterozygotes display partially-penetrant congenital hydrocephalus. These murine data provide an experimental paradigm underpinning clinical observations of the presence of PAX3 mutations in some hydrocephalic patients.
Collapse
|
85
|
Czerwinska AM, Grabowska I, Archacka K, Bem J, Swierczek B, Helinska A, Streminska W, Fogtman A, Iwanicka-Nowicka R, Koblowska M, Ciemerych MA. Myogenic Differentiation of Mouse Embryonic Stem Cells That Lack a Functional Pax7 Gene. Stem Cells Dev 2016; 25:285-300. [PMID: 26649785 PMCID: PMC4761802 DOI: 10.1089/scd.2015.0162] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
The transcription factor Pax7 plays a key role during embryonic myogenesis and sustains the proper function of satellite cells, which serve as adult skeletal muscle stem cells. Overexpression of Pax7 has been shown to promote the myogenic differentiation of pluripotent stem cells. However, the effects of the absence of functional Pax7 in differentiating embryonic stem cells (ESCs) have not yet been directly tested. Herein, we studied mouse stem cells that lacked a functional Pax7 gene and characterized the differentiation of these stem cells under conditions that promoted the derivation of myoblasts in vitro. We analyzed the expression of myogenic factors, such as myogenic regulatory factors and muscle-specific microRNAs, in wild-type and mutant cells. Finally, we compared the transcriptome of both types of cells and did not find substantial differences in the expression of genes related to the regulation of myogenesis. As a result, we showed that the absence of functional Pax7 does not prevent the in vitro myogenic differentiation of ESCs.
Collapse
Affiliation(s)
- Areta M Czerwinska
- 1 Department of Cytology, Faculty of Biology, Institute of Zoology, University of Warsaw , Warsaw, Poland
| | - Iwona Grabowska
- 1 Department of Cytology, Faculty of Biology, Institute of Zoology, University of Warsaw , Warsaw, Poland
| | - Karolina Archacka
- 1 Department of Cytology, Faculty of Biology, Institute of Zoology, University of Warsaw , Warsaw, Poland
| | - Joanna Bem
- 1 Department of Cytology, Faculty of Biology, Institute of Zoology, University of Warsaw , Warsaw, Poland
| | - Barbara Swierczek
- 1 Department of Cytology, Faculty of Biology, Institute of Zoology, University of Warsaw , Warsaw, Poland
| | - Anita Helinska
- 1 Department of Cytology, Faculty of Biology, Institute of Zoology, University of Warsaw , Warsaw, Poland
| | - Wladyslawa Streminska
- 1 Department of Cytology, Faculty of Biology, Institute of Zoology, University of Warsaw , Warsaw, Poland
| | - Anna Fogtman
- 2 Laboratory of Microarray Analysis, Institute of Biochemistry and Biophysics , Polish Academy of Sciences, Warsaw, Poland
| | - Roksana Iwanicka-Nowicka
- 2 Laboratory of Microarray Analysis, Institute of Biochemistry and Biophysics , Polish Academy of Sciences, Warsaw, Poland .,3 Department of Systems Biology, Faculty of Biology, University of Warsaw , Warsaw, Poland
| | - Marta Koblowska
- 2 Laboratory of Microarray Analysis, Institute of Biochemistry and Biophysics , Polish Academy of Sciences, Warsaw, Poland .,3 Department of Systems Biology, Faculty of Biology, University of Warsaw , Warsaw, Poland
| | - Maria A Ciemerych
- 1 Department of Cytology, Faculty of Biology, Institute of Zoology, University of Warsaw , Warsaw, Poland
| |
Collapse
|
86
|
Wu J, Hunt SD, Xue H, Liu Y, Darabi R. Generation and validation of PAX7 reporter lines from human iPS cells using CRISPR/Cas9 technology. Stem Cell Res 2016; 16:220-8. [PMID: 26826926 DOI: 10.1016/j.scr.2016.01.003] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2015] [Revised: 12/10/2015] [Accepted: 01/12/2016] [Indexed: 12/24/2022] Open
Abstract
Directed differentiation of iPS cells toward various tissue progenitors has been the focus of recent research. Therefore, generation of tissue-specific reporter iPS cell lines provides better understanding of developmental stages in iPS cells. This technical report describes an efficient strategy for generation and validation of knock-in reporter lines in human iPS cells using the Cas9-nickase system. Here, we have generated a knock-in human iPS cell line for the early myogenic lineage specification gene of PAX7. By introduction of site-specific double-stranded breaks (DSB) in the genomic locus of PAX7 using CRISPR/Cas9 nickase pairs, a 2A-GFP reporter with selection markers has been incorporated before the stop codon of the PAX7 gene at the last exon. After positive and negative selection, single cell-derived human iPS clones have been isolated and sequenced for in-frame positioning of the reporter construct. Finally, by using a nuclease-dead Cas9 activator (dCas9-VP160) system, the promoter region of PAX7 has been targeted for transient gene induction to validate the GFP reporter activity. This was confirmed by flow cytometry analysis and immunostaining for PAX7 and GFP. This technical report provides a practical guideline for generation and validation of knock-in reporters using CRISPR/Cas9 system.
Collapse
Affiliation(s)
- Jianbo Wu
- Center for Stem Cell and Regenerative Medicine (CSCRM), The Brown Foundation Institute of Molecular Medicine for the Prevention of Human Diseases (IMM), University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Samuel D Hunt
- Center for Stem Cell and Regenerative Medicine (CSCRM), The Brown Foundation Institute of Molecular Medicine for the Prevention of Human Diseases (IMM), University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Haipeng Xue
- Center for Stem Cell and Regenerative Medicine (CSCRM), The Brown Foundation Institute of Molecular Medicine for the Prevention of Human Diseases (IMM), University of Texas Health Science Center at Houston, Houston, TX 77030, USA; Department of Neurosurgery, The Brown Foundation Institute of Molecular Medicine for the Prevention of Human Diseases (IMM), University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Ying Liu
- Center for Stem Cell and Regenerative Medicine (CSCRM), The Brown Foundation Institute of Molecular Medicine for the Prevention of Human Diseases (IMM), University of Texas Health Science Center at Houston, Houston, TX 77030, USA; Department of Neurosurgery, The Brown Foundation Institute of Molecular Medicine for the Prevention of Human Diseases (IMM), University of Texas Health Science Center at Houston, Houston, TX 77030, USA; The Senator Lloyd & B.A. Bentsen Center for Stroke Research, The Brown Foundation Institute of Molecular Medicine for the Prevention of Human Diseases (IMM), University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Radbod Darabi
- Center for Stem Cell and Regenerative Medicine (CSCRM), The Brown Foundation Institute of Molecular Medicine for the Prevention of Human Diseases (IMM), University of Texas Health Science Center at Houston, Houston, TX 77030, USA.
| |
Collapse
|
87
|
Świerczek B, Ciemerych MA, Archacka K. From pluripotency to myogenesis: a multistep process in the dish. J Muscle Res Cell Motil 2015; 36:363-75. [PMID: 26715014 PMCID: PMC4762919 DOI: 10.1007/s10974-015-9436-y] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2015] [Accepted: 11/30/2015] [Indexed: 12/11/2022]
Abstract
Pluripotent stem cells (PSCs), such as embryonic stem cells or induced pluripotent stem cells are a promising source of cells for regenerative medicine as they can differentiate into all cell types building a mammalian body. However, protocols leading to efficient and safe in vitro generation of desired cell types must be perfected before PSCs can be used in cell therapies or tissue engineering. In vivo, i.e. in developing mouse embryo or teratoma, PSCs can differentiate into skeletal muscle, but in vitro their spontaneous differentiation into myogenic cells is inefficient. Numerous attempts have been undertaken to enhance this process. Many of them involved mimicking the interactions occurring during embryonic myogenesis. The key regulators of embryonic myogenesis, such as Wnts proteins, fibroblast growth factor 2, and retinoic acid, have been tested to improve the frequency of in vitro myogenic differentiation of PSCs. This review summarizes the current state of the art, comparing spontaneous and directed myogenic differentiation of PSCs as well as the protocols developed this far to facilitate this process.
Collapse
Affiliation(s)
- Barbara Świerczek
- Department of Cytology, Faculty of Biology, University of Warsaw, Miecznikowa 1, 02-096, Warsaw, Poland
| | - Maria A Ciemerych
- Department of Cytology, Faculty of Biology, University of Warsaw, Miecznikowa 1, 02-096, Warsaw, Poland
| | - Karolina Archacka
- Department of Cytology, Faculty of Biology, University of Warsaw, Miecznikowa 1, 02-096, Warsaw, Poland.
| |
Collapse
|
88
|
Lu Y, Bradley JS, Siegel PB, Yang N, Johnson SE. Selection for divergent body size alters rates of embryonic skeletal muscle formation and muscle gene expression patterns. Dev Growth Differ 2015; 57:614-24. [PMID: 26660844 DOI: 10.1111/dgd.12250] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2015] [Revised: 10/05/2015] [Accepted: 10/13/2015] [Indexed: 11/28/2022]
Abstract
The impact of divergent selection for body size on embryogenesis is poorly understood. The objective of this experiment was to document skeletal muscle development during embryogenesis in two lines of chickens that display divergent growth as adults. Results reveal that after 54 generations of opposing selection from a common founder population, the embryos from the low weight select (LWS) line develop more rapidly during early embryogenesis than those from the high weight select (HWS) line. Muscle formation during the late embryonic period is more rapid and extensive in the HWS embryo than in the LWS contemporary. Isolated muscle progenitors from embryonic day 10 HWS embryos proliferated more rapidly, forming fibers sooner with a larger size than the LWS cells. The limited myogenic capacity of the LWS progenitor cells is not attributed to altered patterns of expression of Pax7, Pax3 or the myogenic regulatory factor genes. Members of the fibroblast growth factor family are potent mitogens and inhibitors of myoblast differentiation. Transcript abundance of FGF2 and FGF4 was measured in cultures of HWS and LWS progenitors as a function of time. The pattern of expression of FGF4 was similar between HWS and LWS with a large increase between days 1 and 3 followed by a reduction at day 5 of culture. Expression of FGF2 in LWS muscle cells did not change while a significant reduction in FGF2 expression was observed by day 5 in the HWS. Our results indicate that divergent selection for postnatal growth has altered embryonic development.
Collapse
Affiliation(s)
- Yue Lu
- National Engineering Laboratory for Animal Breeding and MOA Key Laboratory of Animal Genetics and Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China.,Department of Animal and Poultry Sciences, Virginia Polytechnic and State University, Blacksburg, Virginia, 24061, USA
| | - Jennifer S Bradley
- Department of Animal and Poultry Sciences, Virginia Polytechnic and State University, Blacksburg, Virginia, 24061, USA
| | - Paul B Siegel
- Department of Animal and Poultry Sciences, Virginia Polytechnic and State University, Blacksburg, Virginia, 24061, USA
| | - Ning Yang
- National Engineering Laboratory for Animal Breeding and MOA Key Laboratory of Animal Genetics and Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Sally E Johnson
- Department of Animal and Poultry Sciences, Virginia Polytechnic and State University, Blacksburg, Virginia, 24061, USA
| |
Collapse
|
89
|
Buckingham M, Relaix F. PAX3 and PAX7 as upstream regulators of myogenesis. Semin Cell Dev Biol 2015; 44:115-25. [PMID: 26424495 DOI: 10.1016/j.semcdb.2015.09.017] [Citation(s) in RCA: 149] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2015] [Accepted: 09/23/2015] [Indexed: 10/23/2022]
Abstract
Like other subclasses within the PAX transcription factor family, PAX3 and PAX7 play important roles in the emergence of a number of different tissues during development. PAX3 regulates neural crest and, together with its orthologue PAX7, is also expressed in parts of the central nervous system. In this chapter we will focus on their role in skeletal muscle. Both factors are key regulators of myogenesis where Pax3 plays a major role during early skeletal muscle formation in the embryo while Pax7 predominates during post-natal growth and muscle regeneration in the adult. We review the expression and functions of these factors in the myogenic context. We also discuss mechanistic aspects of PAX3/7 function and modulation of their activity by interaction with other proteins, as well as the post-transcriptional and transcriptional regulation of their expression.
Collapse
Affiliation(s)
- Margaret Buckingham
- Department of Developmental and Stem Cell Biology, CNRS URA 2578, Institut Pasteur, 28 rue du Dr Roux, 75015 Paris, France.
| | - Frédéric Relaix
- INSERM U955 IMRB, Team 10, 94000 Creteil, France; UPEC Paris Est-Creteil University, Faculty of Medicine, F-94000 Creteil, France; Etablissement Français du Sang, 94017 Creteil, France; Université Paris Est, Ecole Nationale Veterinaire d'Alfort, 94700 Maison Alfort, France.
| |
Collapse
|
90
|
Monsoro-Burq AH. PAX transcription factors in neural crest development. Semin Cell Dev Biol 2015; 44:87-96. [PMID: 26410165 DOI: 10.1016/j.semcdb.2015.09.015] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2015] [Revised: 09/14/2015] [Accepted: 09/21/2015] [Indexed: 10/23/2022]
Abstract
The nine vertebrate PAX transcription factors (PAX1-PAX9) play essential roles during early development and organogenesis. Pax genes were identified in vertebrates using their homology with the Drosophila melanogaster paired gene DNA-binding domain. PAX1-9 functions are largely conserved throughout vertebrate evolution, in particular during central nervous system and neural crest development. The neural crest is a vertebrate invention, which gives rise to numerous derivatives during organogenesis, including neurons and glia of the peripheral nervous system, craniofacial skeleton and mesenchyme, the heart outflow tract, endocrine and pigment cells. Human and mouse spontaneous mutations as well as experimental analyses have evidenced the critical and diverse functions of PAX factors during neural crest development. Recent studies have highlighted the role of PAX3 and PAX7 in neural crest induction. Additionally, several PAX proteins - PAX1, 3, 7, 9 - regulate cell proliferation, migration and determination in multiple neural crest-derived lineages, such as cardiac, sensory, and enteric neural crest, pigment cells, glia, craniofacial skeleton and teeth, or in organs developing in close relationship with the neural crest such as the thymus and parathyroids. The diverse PAX molecular functions during neural crest formation rely on fine-tuned modulations of their transcriptional transactivation properties. These modulations are generated by multiple means, such as different roles for the various isoforms (formed by alternative splicing), or posttranslational modifications which alter protein-DNA binding, or carefully orchestrated protein-protein interactions with various co-factors which control PAX proteins activity. Understanding these regulations is the key to decipher the versatile roles of PAX transcription factors in neural crest development, differentiation and disease.
Collapse
Affiliation(s)
- Anne H Monsoro-Burq
- Univ. Paris Sud, Université Paris Saclay, Centre Universitaire, 15, rue Georges Clémenceau, F-91405 Orsay, France; Institut Curie Research Division, Centre Universitaire, 15, rue Georges Clémenceau, F-91405 Orsay, France; UMR 3347 CNRS, U1021 Inserm, Université Paris Saclay, Centre Universitaire, 15, rue Georges Clémenceau, F-91405 Orsay, France.
| |
Collapse
|
91
|
A Cranial Mesoderm Origin for Esophagus Striated Muscles. Dev Cell 2015; 34:694-704. [PMID: 26387456 DOI: 10.1016/j.devcel.2015.07.003] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2015] [Revised: 06/08/2015] [Accepted: 07/10/2015] [Indexed: 11/21/2022]
Abstract
The esophagus links the oral cavity to the stomach and facilitates the transfer of bolus. Using genetic tracing and mouse mutants, we demonstrate that esophagus striated muscles (ESMs) are not derived from somites but are of cranial origin. Tbx1 and Isl1 act as key regulators of ESMs, which we now identify as a third derivative of cardiopharyngeal mesoderm that contributes to second heart field derivatives and head muscles. Isl1-derived ESM progenitors colonize the mouse esophagus in an anterior-posterior direction but are absent in the developing chick esophagus, thus providing evolutionary insight into the lack of ESMs in avians. Strikingly, different from other myogenic regions, in which embryonic myogenesis establishes a scaffold for fetal fiber formation, ESMs are established directly by fetal myofibers. We propose that ESM progenitors use smooth muscle as a scaffold, thereby bypassing the embryonic program. These findings have important implications in understanding esophageal dysfunctions, including dysphagia, and congenital disorders, such as DiGeorge syndrome.
Collapse
|
92
|
Lin J, Wang C, Yang C, Fu S, Redies C. Pax3 and Pax7 interact reciprocally and regulate the expression of cadherin-7 through inducing neuron differentiation in the developing chicken spinal cord. J Comp Neurol 2015; 524:940-62. [PMID: 26287727 DOI: 10.1002/cne.23885] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2015] [Revised: 08/11/2015] [Accepted: 08/12/2015] [Indexed: 01/12/2023]
Abstract
Pax3 and Pax7 are closely related transcription factors that are widely expressed in the developing nervous system and somites. In the CNS, both genes are expressed in the dorsal part of the neural tube during development. Pax3 and Pax7 are involved in the sonic hedgehog (Shh) signaling pathway and are inhibited by Shh overexpression. The present study confirms in vivo that Pax3 overexpression represses the expression of Pax7, whereas Pax7 overexpression endogenously enhances and ectopically induces the expression of Pax3 in the developing chicken spinal cord. Overexpression of Pax3 and Pax7 represses the endogenous expression of cadherin-7, a member of the cadherin family of morphogenetic genes, and induces its ectopic expression. The present study also shows that overexpression of Pax3 and Pax7 changes the fate and morphology of cells in the neuroepithelial layer and induces the expression of postmitotic neuronal markers. We show that both Pax3 and Pax7 promote the differentiation of neural progenitor cells into neurons. Furthermore, the downregulation of Pax3 and Pax7 with specific shRNAs results in apoptosis in the developing spinal cord. Collectively, these results suggest that the transcription factors Pax3 and Pax7 play important roles in regulating morphogenesis and cell differentiation in the developing spinal cord.
Collapse
Affiliation(s)
- Juntang Lin
- Institute of Anatomy I, University of Jena School of Medicine, Jena University Hospital, D-07743, Jena, Germany.,College of Life Science and Technology, Xinxiang Medical University, 453003, Xinxiang, China
| | - Congrui Wang
- Institute of Anatomy I, University of Jena School of Medicine, Jena University Hospital, D-07743, Jena, Germany.,College of Life Science and Technology, Xinxiang Medical University, 453003, Xinxiang, China
| | - Ciqing Yang
- College of Life Science and Technology, Xinxiang Medical University, 453003, Xinxiang, China
| | - Sulei Fu
- Institute of Anatomy I, University of Jena School of Medicine, Jena University Hospital, D-07743, Jena, Germany
| | - Christoph Redies
- Institute of Anatomy I, University of Jena School of Medicine, Jena University Hospital, D-07743, Jena, Germany
| |
Collapse
|
93
|
Jiao S, Tan X, Li M, Sui Y, Du SJ, You F. The duplicated paired box protein 7 (pax7) genes differentially transcribed during Japanese flounder (Paralichthys olivaceus) embryogenesis. Comp Biochem Physiol B Biochem Mol Biol 2015; 189:62-8. [PMID: 26275626 DOI: 10.1016/j.cbpb.2015.08.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2015] [Revised: 07/29/2015] [Accepted: 08/04/2015] [Indexed: 11/16/2022]
Abstract
PAX are important regulators of developmental processes. PAX7 plays crucial roles in patterning of the dorsal central nervous system (CNS), neural crest (NC), and skeletal muscle. Here, we identified six spliced isoforms of pax7a and one pax7b and characterized their expression patterns. All of flounder Pax7a-1, Pax7a-2, Pax7a-3, and Pax7b contain a conserved paired domain (PD), an octapeptide motif (OP), and a paired type homeodomain (HD). However, the PD of Pax7a-4 and the HD of Pax7a-5 are not intact, and there is no HD in Pax7a-4 and Pax7a-6. pax7a and pax7b show distinct spatiotemporal expression patterns during embryogenesis. Whole-mount in situ hybridization demonstrates that the expression patterns of pax7a and pax7b are overlapping but distinguishable in the dorsal central nervous system. pax7a is expressed in most part of the brain and the neural tube, while pax7b is expressed exclusively in the diencephalon and the midbrain. In addition, pax7a is also expressed in the cranial NC and the trunk NC. RT-PCR results show that there were different expression patterns between the different isoforms. These results indicate subfunction partitioning of the duplicated pax7 genes. The duplicated pax7 may provide additional flexibility in fine-tuning neurogenesis and somitogenesis.
Collapse
Affiliation(s)
- Shuang Jiao
- Key Laboratory of Experimental Marine Biology, National and Local Joint Engineering Laboratory of Ecological Mariculture, Institute of Oceanology, Chinese Academy of Sciences, 7 Nanhai Road, Qingdao, Shandong 266071, People's Republic of China
| | - Xungang Tan
- Key Laboratory of Experimental Marine Biology, National and Local Joint Engineering Laboratory of Ecological Mariculture, Institute of Oceanology, Chinese Academy of Sciences, 7 Nanhai Road, Qingdao, Shandong 266071, People's Republic of China.
| | - Meijie Li
- Key Laboratory of Experimental Marine Biology, National and Local Joint Engineering Laboratory of Ecological Mariculture, Institute of Oceanology, Chinese Academy of Sciences, 7 Nanhai Road, Qingdao, Shandong 266071, People's Republic of China; University of Chinese Academy of Sciences, Beijing 100049, People's Republic of China
| | - Yulei Sui
- Key Laboratory of Experimental Marine Biology, National and Local Joint Engineering Laboratory of Ecological Mariculture, Institute of Oceanology, Chinese Academy of Sciences, 7 Nanhai Road, Qingdao, Shandong 266071, People's Republic of China; University of Chinese Academy of Sciences, Beijing 100049, People's Republic of China
| | - Shao Jun Du
- Department of Biochemistry and Molecular Biology, School of Medicine, University of Maryland, 701 E. Pratt St., Baltimore, MD 21202, USA
| | - Feng You
- Key Laboratory of Experimental Marine Biology, National and Local Joint Engineering Laboratory of Ecological Mariculture, Institute of Oceanology, Chinese Academy of Sciences, 7 Nanhai Road, Qingdao, Shandong 266071, People's Republic of China
| |
Collapse
|
94
|
Unpeaceful roles of mutant PAX proteins in cancer. Semin Cell Dev Biol 2015; 44:126-34. [DOI: 10.1016/j.semcdb.2015.09.011] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2015] [Revised: 09/10/2015] [Accepted: 09/16/2015] [Indexed: 01/07/2023]
|
95
|
Abstract
Skeletal muscle development has been the focus of intensive study for many decades. Recent advances in genetic manipulation of the mouse have increased our understanding of the cell signalling involved in the development of muscle progenitors which give rise to adult skeletal muscles and their stem cell populations. However, the influence of a vital tissue type – the peripheral nerve—has largely been ignored since its earliest descriptions. Here we carefully describe the timing in which myogenic progenitors expressing Pax3 and Pax7 (the earliest markers of myogenic cells) enter the limb buds of rat and mouse embryos, as well as the spatiotemporal relationship between these progenitors and the ingrowing peripheral nerve. We show that progenitors expressing Pax3 enter the limb bud one full day ahead of the first neurites and that Pax7-expressing progenitors (associated with secondary myogenesis in the limb) are first seen in the limb bud at the time of nerve entry and in close proximity to the nerve. The initial entry of the nerve also coincides with the first expression of myosin heavy chain showing that the first contact between nerves and myogenic cells correlates with the onset of myogenic differentiation. Furthermore, as the nerve grows into the limb, Pax3 expression is progressively replaced by Pax7 expression in myogenic progenitors. These findings indicate that the ingrowing nerve enters the limb presumptive muscle masses earlier than what was generally described and raises the possibility that nerve may influence the differentiation of muscle progenitors in rodent limbs.
Collapse
|
96
|
Dumont NA, Wang YX, Rudnicki MA. Intrinsic and extrinsic mechanisms regulating satellite cell function. Development 2015; 142:1572-81. [PMID: 25922523 PMCID: PMC4419274 DOI: 10.1242/dev.114223] [Citation(s) in RCA: 315] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Muscle stem cells, termed satellite cells, are crucial for skeletal muscle growth and regeneration. In healthy adult muscle, satellite cells are quiescent but poised for activation. During muscle regeneration, activated satellite cells transiently re-enter the cell cycle to proliferate and subsequently exit the cell cycle to differentiate or self-renew. Recent studies have demonstrated that satellite cells are heterogeneous and that subpopulations of satellite stem cells are able to perform asymmetric divisions to generate myogenic progenitors or symmetric divisions to expand the satellite cell pool. Thus, a complex balance between extrinsic cues and intrinsic regulatory mechanisms is needed to tightly control satellite cell cycle progression and cell fate determination. Defects in satellite cell regulation or in their niche, as observed in degenerative conditions such as aging, can impair muscle regeneration. Here, we review recent discoveries of the intrinsic and extrinsic factors that regulate satellite cell behaviour in regenerating and degenerating muscles. Summary: This Review discusses how satellite stem cell behaviour is regulated during regeneration and degeneration by a complex balance between extrinsic cues and intrinsic regulatory mechanisms.
Collapse
Affiliation(s)
- Nicolas A Dumont
- Sprott Centre for Stem Cell Research, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada K1H 8L6
| | - Yu Xin Wang
- Sprott Centre for Stem Cell Research, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada K1H 8L6 Faculty of Medicine, Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario, Canada K1H 8M5
| | - Michael A Rudnicki
- Sprott Centre for Stem Cell Research, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada K1H 8L6 Faculty of Medicine, Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario, Canada K1H 8M5
| |
Collapse
|
97
|
Zalc A, Rattenbach R, Auradé F, Cadot B, Relaix F. Pax3 and Pax7 play essential safeguard functions against environmental stress-induced birth defects. Dev Cell 2015; 33:56-66. [PMID: 25800090 DOI: 10.1016/j.devcel.2015.02.006] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2014] [Revised: 12/11/2014] [Accepted: 02/08/2015] [Indexed: 12/23/2022]
Abstract
Exposure to environmental teratogenic pollutant leads to severe birth defects. However, the biological events underlying these developmental abnormalities remain undefined. Here, we report a molecular link between an environmental stress response pathway and key developmental genes during craniofacial development. Strikingly, mutant mice with impaired Pax3/7 function display severe craniofacial defects. We show that these are associated with an upregulation of the signaling pathway mediated by the Aryl hydrocarbon receptor (AHR), the receptor to 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD), revealing a genetic interaction between Pax3 and AHR signaling. Activation of AHR signaling in Pax3-deficient embryos drives facial mesenchymal cells out of the cell cycle through the upregulation of p21 expression. Accordingly, inhibiting AHR activity rescues the cycling status of these cells and the facial closure of Pax3/7 mutants. Together, our findings demonstrate that the regulation of AHR signaling by Pax3/7 is required to protect against TCDD/AHR-mediated teratogenesis during craniofacial development.
Collapse
Affiliation(s)
- Antoine Zalc
- Sorbonne Universités, UPMC Univ Paris 06, Myology Research Center, INSERM U974, CNRS FRE 3617, Institut de Myologie, 75013 Paris, France; INSERM U955 IMRB, Team 10, 94000 Creteil, France; UPEC Paris Est-Creteil University, Faculty of Medicine, 94000 Creteil, France
| | - Revital Rattenbach
- Sorbonne Universités, UPMC Univ Paris 06, Myology Research Center, INSERM U974, CNRS FRE 3617, Institut de Myologie, 75013 Paris, France; INSERM U955 IMRB, Team 10, 94000 Creteil, France; UPEC Paris Est-Creteil University, Faculty of Medicine, 94000 Creteil, France
| | - Frédéric Auradé
- Sorbonne Universités, UPMC Univ Paris 06, Myology Research Center, INSERM U974, CNRS FRE 3617, Institut de Myologie, 75013 Paris, France
| | - Bruno Cadot
- Sorbonne Universités, UPMC Univ Paris 06, Myology Research Center, INSERM U974, CNRS FRE 3617, Institut de Myologie, 75013 Paris, France
| | - Frédéric Relaix
- Sorbonne Universités, UPMC Univ Paris 06, Myology Research Center, INSERM U974, CNRS FRE 3617, Institut de Myologie, 75013 Paris, France; INSERM U955 IMRB, Team 10, 94000 Creteil, France; UPEC Paris Est-Creteil University, Faculty of Medicine, 94000 Creteil, France; Etablissement Français du Sang, 94017 Creteil, France; Université Paris Est, Ecole Nationale Veterinaire d'Alfort, 94700 Maison-Alfort, France.
| |
Collapse
|
98
|
Gryzińska M, Jakubczak A, Stryjecki R, Jeżewska-Witkowska G. In silico analysis of methylation of the selected genes using computer programs based on various analytical techniques. Biocybern Biomed Eng 2015. [DOI: 10.1016/j.bbe.2014.09.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
99
|
Grifone R, Xie X, Bourgeois A, Saquet A, Duprez D, Shi DL. The RNA-binding protein Rbm24 is transiently expressed in myoblasts and is required for myogenic differentiation during vertebrate development. Mech Dev 2014; 134:1-15. [PMID: 25217815 DOI: 10.1016/j.mod.2014.08.003] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2014] [Revised: 08/05/2014] [Accepted: 08/22/2014] [Indexed: 12/16/2022]
Abstract
RNA-binding proteins (RBP) contribute to gene regulation through post-transcriptional events. Despite the important roles demonstrated for several RBP in regulating skeletal myogenesis in vitro, very few RBP coding genes have been characterized during skeletal myogenesis in vertebrate embryo. In the present study we report that Rbm24, which encodes the RNA-binding motif protein 24, is required for skeletal muscle differentiation in vivo. We show that Rbm24 transcripts are expressed at all sites of skeletal muscle formation during embryogenesis of different vertebrates, including axial, limb and head muscles. Interestingly, we find that Rbm24 protein starts to accumulate in MyoD-positive myoblasts and is transiently expressed at the onset of muscle cell differentiation. It accumulates in myotomal and limb myogenic cells, but not in Pax3-positive progenitor cells. Rbm24 expression is under the direct regulation by MyoD, as demonstrated by in vivo chromatin immunoprecipitation assay. Using morpholino knockdown approach, we further show that Rbm24 is required for somitic myogenic progenitor cells to differentiate into muscle cells during chick somitic myogenesis. Altogether, these results highlight Rbm24 as a novel key regulator of the myogenic differentiation program during vertebrate development.
Collapse
Affiliation(s)
- Raphaëlle Grifone
- Sorbonne Universités, UPMC Univ Paris 06, UMR 7622, Laboratory of Developmental Biology, Paris F-75005, France; CNRS, UMR 7622, Laboratory of Developmental Biology, Paris F-75005, France
| | - Xin Xie
- Sorbonne Universités, UPMC Univ Paris 06, UMR 7622, Laboratory of Developmental Biology, Paris F-75005, France; CNRS, UMR 7622, Laboratory of Developmental Biology, Paris F-75005, France
| | - Adeline Bourgeois
- Sorbonne Universités, UPMC Univ Paris 06, UMR 7622, Laboratory of Developmental Biology, Paris F-75005, France; CNRS, UMR 7622, Laboratory of Developmental Biology, Paris F-75005, France
| | - Audrey Saquet
- Sorbonne Universités, UPMC Univ Paris 06, UMR 7622, Laboratory of Developmental Biology, Paris F-75005, France; CNRS, UMR 7622, Laboratory of Developmental Biology, Paris F-75005, France
| | - Delphine Duprez
- Sorbonne Universités, UPMC Univ Paris 06, UMR 7622, Laboratory of Developmental Biology, Paris F-75005, France; CNRS, UMR 7622, Laboratory of Developmental Biology, Paris F-75005, France
| | - De-Li Shi
- Sorbonne Universités, UPMC Univ Paris 06, UMR 7622, Laboratory of Developmental Biology, Paris F-75005, France; CNRS, UMR 7622, Laboratory of Developmental Biology, Paris F-75005, France.
| |
Collapse
|
100
|
Eng D, Vogel WK, Flann NS, Gross MK, Kioussi C. Genome-Wide Mapping of Chromatin State of Mouse Forelimbs. ACTA ACUST UNITED AC 2014; 6:1-11. [PMID: 25530700 DOI: 10.2147/oab.s59043] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
BACKGROUND Cell types are defined at the molecular level during embryogenesis by a process called pattern formation and created by the selective utilization of combinations of sequence specific transcription factors. Developmental programs define the sets of genes that are available to each particular cell type, and real-time biochemical signaling interactions define the extent to which these sets are used at any given time and place. Gene expression is regulated through the integrated action of many cis-regulatory elements, including core promoters, enhancers, silencers, and insulators. The chromatin state in developing body parts provides a code to cellular populations that direct their cell fates. Chromatin profiling has been a method of choice for mapping regulatory sequences in cells that go through developmental transitions. RESULTS We used antibodies against histone H3 lysine 4 trimethylations (H3K4me3) a modification associated with promoters and open/active chromatin, histone H3 lysine 27 trimethylations (H3K27me3) associated with Polycomb-repressed regions and RNA polymerase II (Pol2) associated with transcriptional initiation to identify the chromatin state signature of the mouse forelimb during mid-gestation, at embryonic day 12 (E12). The families of genes marked included those related to transcriptional regulation and embryogenesis. One third of the marked genes were transcriptionally active while only a small fraction were bivalent marked. Sequence specific transcription factors that were activated were involved in cell specification including bone and muscle formation. CONCLUSION Our results demonstrate that embryonic limb cells do not exhibit the plasticity of the ES cells but are rather programmed for a finer tuning for cell lineage specification.
Collapse
Affiliation(s)
- Diana Eng
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Corvallis, OR 97331, USA
| | - Walter K Vogel
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Corvallis, OR 97331, USA
| | - Nicholas S Flann
- Department of Computer Science, Utah State University, United States, Institute for Systems Biology, Seattle, United States
| | - Michael K Gross
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Corvallis, OR 97331, USA
| | - Chrissa Kioussi
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Corvallis, OR 97331, USA
| |
Collapse
|