1
|
Yong P, Zhang Z, Du S. Ectopic expression of Myomaker and Myomixer in slow muscle cells induces slow muscle fusion and myofiber death. J Genet Genomics 2024:S1673-8527(24)00214-5. [PMID: 39209151 DOI: 10.1016/j.jgg.2024.08.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 08/21/2024] [Accepted: 08/21/2024] [Indexed: 09/04/2024]
Abstract
Zebrafish embryos possess two major types of myofibers, the slow and fast fibers, with distinct patterns of cell fusion. The fast muscle cells can fuse, while the slow muscle cells cannot. Here, we show that myomaker is expressed in both slow and fast muscle precursors, whereas myomixer is exclusive to fast muscle cells. The loss of Prdm1a, a regulator of slow muscle differentiation, results in strong myomaker and myomixer expression and slow muscle cell fusion. This abnormal fusion is further confirmed by the direct ectopic expression of myomaker or myomixer in slow muscle cells of transgenic models. Using the transgenic models, we show that the heterologous fusion between slow and fast muscle cells can alter slow muscle cell migration and gene expression. Furthermore, the overexpression of myomaker and myomixer also disrupts membrane integrity, resulting in muscle cell death. Collectively, this study identifies that the fiber-type-specific expression of fusogenic proteins is critical for preventing inappropriate fusion between slow and fast fibers in fish embryos, highlighting the need for precise regulation of fusogenic gene expression to maintain muscle fiber integrity and specificity.
Collapse
Affiliation(s)
- Pengzheng Yong
- Department of Biochemistry and Molecular Biology, Institute of Marine and Environmental Technology, University of Maryland School of Medicine, Baltimore, MD 21202, United States
| | - Zhanxiong Zhang
- Department of Biochemistry and Molecular Biology, Institute of Marine and Environmental Technology, University of Maryland School of Medicine, Baltimore, MD 21202, United States
| | - Shaojun Du
- Department of Biochemistry and Molecular Biology, Institute of Marine and Environmental Technology, University of Maryland School of Medicine, Baltimore, MD 21202, United States.
| |
Collapse
|
2
|
Oudhoff H, Hisler V, Baumgartner F, Rees L, Grepper D, Jaźwińska A. Skeletal muscle regeneration after extensive cryoinjury of caudal myomeres in adult zebrafish. NPJ Regen Med 2024; 9:8. [PMID: 38378693 PMCID: PMC10879182 DOI: 10.1038/s41536-024-00351-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Accepted: 01/24/2024] [Indexed: 02/22/2024] Open
Abstract
Skeletal muscles can regenerate after minor injuries, but severe structural damage often leads to fibrosis in mammals. Whether adult zebrafish possess the capacity to reproduce profoundly destroyed musculature remains unknown. Here, a new cryoinjury model revealed that several myomeres efficiently regenerated within one month after wounding the zebrafish caudal peduncle. Wound clearance involved accumulation of the selective autophagy receptor p62, an immune response and Collagen XII deposition. New muscle formation was associated with proliferation of Pax7 expressing muscle stem cells, which gave rise to MyoD1 positive myogenic precursors, followed by myofiber differentiation. Monitoring of slow and fast muscles revealed their coordinated replacement in the superficial and profound compartments of the myomere. However, the final boundary between the muscular components was imperfectly recapitulated, allowing myofibers of different identities to intermingle. The replacement of connective with sarcomeric tissues required TOR signaling, as rapamycin treatment impaired new muscle formation, leading to persistent fibrosis. The model of zebrafish myomere restoration may provide new medical perspectives for treatment of traumatic injuries.
Collapse
Affiliation(s)
- Hendrik Oudhoff
- Department of Biology, University of Fribourg, Chemin du Musée 10, 1700, Fribourg, Switzerland
| | - Vincent Hisler
- Department of Biology, University of Fribourg, Chemin du Musée 10, 1700, Fribourg, Switzerland
| | - Florian Baumgartner
- Department of Biology, University of Fribourg, Chemin du Musée 10, 1700, Fribourg, Switzerland
| | - Lana Rees
- Department of Biology, University of Fribourg, Chemin du Musée 10, 1700, Fribourg, Switzerland
| | - Dogan Grepper
- Department of Biology, University of Fribourg, Chemin du Musée 10, 1700, Fribourg, Switzerland
| | - Anna Jaźwińska
- Department of Biology, University of Fribourg, Chemin du Musée 10, 1700, Fribourg, Switzerland.
| |
Collapse
|
3
|
Karuppasamy M, English KG, Henry CA, Manzini MC, Parant JM, Wright MA, Ruparelia AA, Currie PD, Gupta VA, Dowling JJ, Maves L, Alexander MS. Standardization of zebrafish drug testing parameters for muscle diseases. Dis Model Mech 2024; 17:dmm050339. [PMID: 38235578 PMCID: PMC10820820 DOI: 10.1242/dmm.050339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2023] [Accepted: 12/06/2023] [Indexed: 01/19/2024] Open
Abstract
Skeletal muscular diseases predominantly affect skeletal and cardiac muscle, resulting in muscle weakness, impaired respiratory function and decreased lifespan. These harmful outcomes lead to poor health-related quality of life and carry a high healthcare economic burden. The absence of promising treatments and new therapies for muscular disorders requires new methods for candidate drug identification and advancement in animal models. Consequently, the rapid screening of drug compounds in an animal model that mimics features of human muscle disease is warranted. Zebrafish are a versatile model in preclinical studies that support developmental biology and drug discovery programs for novel chemical entities and repurposing of established drugs. Due to several advantages, there is an increasing number of applications of the zebrafish model for high-throughput drug screening for human disorders and developmental studies. Consequently, standardization of key drug screening parameters, such as animal husbandry protocols, drug compound administration and outcome measures, is paramount for the continued advancement of the model and field. Here, we seek to summarize and explore critical drug treatment and drug screening parameters in the zebrafish-based modeling of human muscle diseases. Through improved standardization and harmonization of drug screening parameters and protocols, we aim to promote more effective drug discovery programs.
Collapse
Affiliation(s)
- Muthukumar Karuppasamy
- Division of Neurology, Department of Pediatrics, University of Alabama at Birmingham and Children's of Alabama, Birmingham, AL 35294, USA
| | - Katherine G. English
- Division of Neurology, Department of Pediatrics, University of Alabama at Birmingham and Children's of Alabama, Birmingham, AL 35294, USA
| | - Clarissa A. Henry
- Graduate School of Biomedical Science and Engineering, University of Maine, Orono, ME 04469, USA
- School of Biology and Ecology, University of Maine, Orono, ME 04469, USA
| | - M. Chiara Manzini
- Child Health Institute of New Jersey and Department of Neuroscience and Cell Biology, Rutgers, Robert Wood Johnson Medical School, New Brunswick, NJ 08901, USA
| | - John M. Parant
- Department of Pharmacology and Toxicology, University of Alabama at Birmingham Heersink School of Medicine, Birmingham, AL 35294, USA
| | - Melissa A. Wright
- Department of Pediatrics, Section of Child Neurology, University of Colorado at Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Avnika A. Ruparelia
- Department of Anatomy and Physiology, School of Biomedical Sciences, Faculty of Medicine Dentistry and Health Sciences, University of Melbourne, Melbourne, Victoria 3010, Australia
- Centre for Muscle Research, Department of Anatomy and Physiology, University of Melbourne, Melbourne, Victoria 3010, Australia
- Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria 3800, Australia
| | - Peter D. Currie
- Centre for Muscle Research, Department of Anatomy and Physiology, University of Melbourne, Melbourne, Victoria 3010, Australia
- Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria 3800, Australia
- EMBL Australia, Victorian Node, Monash University, Clayton, Victoria 3800, Australia
| | - Vandana A. Gupta
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - James J. Dowling
- Division of Neurology, The Hospital for Sick Children, Toronto, Ontario M5G 1X8, Canada
- Department of Paediatrics, University of Toronto, Toronto, Ontario M5G 1X8, Canada
- Program for Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Ontario M5G 0A4, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario M5G 0A4, Canada
| | - Lisa Maves
- Center for Developmental Biology and Regenerative Medicine, Seattle Children's Research Institute, Seattle, WA 98101, USA
- Department of Pediatrics, University of Washington, Seattle, WA 98195, USA
| | - Matthew S. Alexander
- Division of Neurology, Department of Pediatrics, University of Alabama at Birmingham and Children's of Alabama, Birmingham, AL 35294, USA
- UAB Center for Exercise Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
- Department of Genetics, University of Alabama at Birmingham, Birmingham, AL 35294, USA
- Civitan International Research Center, University of Alabama at Birmingham, Birmingham, AL 35294, USA
- UAB Center for Neurodegeneration and Experimental Therapeutics (CNET), Birmingham, AL 35294, USA
| |
Collapse
|
4
|
Hau HTA, Kelu JJ, Ochala J, Hughes SM. Slow myosin heavy chain 1 is required for slow myofibril and muscle fibre growth but not for myofibril initiation. Dev Biol 2023; 499:47-58. [PMID: 37121308 PMCID: PMC10713478 DOI: 10.1016/j.ydbio.2023.04.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Revised: 03/31/2023] [Accepted: 04/04/2023] [Indexed: 05/02/2023]
Abstract
Slow myosin heavy chain 1 (Smyhc1) is the major sarcomeric myosin driving early contraction by slow skeletal muscle fibres in zebrafish. New mutant alleles lacking a functional smyhc1 gene move poorly, but recover motility as the later-formed fast muscle fibres of the segmental myotomes mature, and are adult viable. By motility analysis and inhibiting fast muscle contraction pharmacologically, we show that a slow muscle motility defect persists in mutants until about 1 month of age. Breeding onto a genetic background marking slow muscle fibres with EGFP revealed that mutant slow fibres undergo terminal differentiation, migration and fibre formation indistinguishable from wild type but fail to generate large myofibrils and maintain cellular orientation and attachments. In mutants, initial myofibrillar structures with 1.67 μm periodic actin bands fail to mature into the 1.96 μm sarcomeres observed in wild type, despite the presence of alternative myosin heavy chain molecules. The poorly-contractile mutant slow muscle cells generate numerous cytoplasmic organelles, but fail to grow and bundle myofibrils or to increase in cytoplasmic volume despite passive movements imposed by fast muscle. The data show that both slow myofibril maturation and cellular volume increase depend on the function of a specific myosin isoform and suggest that appropriate force production regulates muscle fibre growth.
Collapse
Affiliation(s)
- Hoi-Ting A Hau
- Randall Centre for Cell and Molecular Biophysics, School of Basic and Medical Biosciences, King's College London, SE1 1UL, UK; Centre for Human & Applied Physiological Sciences, School of Basic and Medical Biosciences, King's College London, SE1 1UL, UK
| | - Jeffrey J Kelu
- Randall Centre for Cell and Molecular Biophysics, School of Basic and Medical Biosciences, King's College London, SE1 1UL, UK
| | - Julien Ochala
- Centre for Human & Applied Physiological Sciences, School of Basic and Medical Biosciences, King's College London, SE1 1UL, UK
| | - Simon M Hughes
- Randall Centre for Cell and Molecular Biophysics, School of Basic and Medical Biosciences, King's College London, SE1 1UL, UK.
| |
Collapse
|
5
|
Paolocci E, Zaccolo M. Compartmentalised cAMP signalling in the primary cilium. Front Physiol 2023; 14:1187134. [PMID: 37256063 PMCID: PMC10226274 DOI: 10.3389/fphys.2023.1187134] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Accepted: 04/25/2023] [Indexed: 06/01/2023] Open
Abstract
cAMP is a universal second messenger that relies on precise spatio-temporal regulation to control varied, and often opposing, cellular functions. This is achieved via selective activation of effectors embedded in multiprotein complexes, or signalosomes, that reside at distinct subcellular locations. cAMP is also one of many pathways known to operate within the primary cilium. Dysfunction of ciliary signaling leads to a class of diseases known as ciliopathies. In Autosomal Dominant Polycystic Kidney Disease (ADPKD), a ciliopathy characterized by the formation of fluid-filled kidney cysts, upregulation of cAMP signaling is known to drive cystogenesis. For decades it has been debated whether the primary cilium is an independent cAMP sub-compartment, or whether it shares a diffusible pool of cAMP with the cell body. Recent studies now suggest it is a specific pool of cAMP generated in the cilium that propels cyst formation in ADPKD, supporting the notion that this antenna-like organelle is a compartment within which cAMP signaling occurs independently from cAMP signaling in the bulk cytosol. Here we present examples of cAMP function in the cilium which suggest this mysterious organelle is home to more than one cAMP signalosome. We review evidence that ciliary membrane localization of G-Protein Coupled Receptors (GPCRs) determines their downstream function and discuss how optogenetic tools have contributed to establish that cAMP generated in the primary cilium can drive cystogenesis.
Collapse
|
6
|
Ikeda T, Inamori K, Kawanishi T, Takeda H. Reemployment of Kupffer's vesicle cells into axial and paraxial mesoderm via transdifferentiation. Dev Growth Differ 2022; 64:163-177. [PMID: 35129208 DOI: 10.1111/dgd.12774] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 01/17/2022] [Accepted: 01/25/2022] [Indexed: 01/25/2023]
Abstract
Kupffer's vesicle (KV) in the teleost embryo is a fluid-filled vesicle surrounded by a layer of epithelial cells with rotating primary cilia. KV transiently acts as the left-right organizer and degenerates after the establishment of left-right asymmetric gene expression. Previous labelling experiments in zebrafish embryos indicated that descendants of KV-epithelial cells are incorporated into mesodermal tissues after the collapse of KV. However, the overall picture of their differentiation potency had been unclear due to the lack of suitable genetic tools and molecular analyses. In the present study, we established a novel zebrafish transgenic line with a promoter of dand5, in which all KV-epithelial cells and their descendants are specifically labelled until the larval stage. We found that KV-epithelial cells undergo epithelial-mesenchymal transition upon KV collapse and infiltrate into adjacent mesodermal progenitors, the presomitic mesoderm and chordoneural hinge. Once incorporated, the descendants of KV-epithelial cells expressed distinct mesodermal differentiation markers and contributed to the mature populations such as the axial muscles and notochordal sheath through normal developmental process. These results indicate that differentiated KV-epithelial cells possess unique plasticity in that they are reemployed into mesodermal lineages through transdifferentiation after they complete their initial role in KV.
Collapse
Affiliation(s)
- Takafumi Ikeda
- Laboratory of Embryology, Department of Biological Sciences, Graduate School of Science, The University of Tokyo, Tokyo, Japan
| | - Kiichi Inamori
- Laboratory of Embryology, Department of Biological Sciences, Graduate School of Science, The University of Tokyo, Tokyo, Japan
| | - Toru Kawanishi
- Laboratory of Embryology, Department of Biological Sciences, Graduate School of Science, The University of Tokyo, Tokyo, Japan
| | - Hiroyuki Takeda
- Laboratory of Embryology, Department of Biological Sciences, Graduate School of Science, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
7
|
Root ZD, Allen C, Gould C, Brewer M, Jandzik D, Medeiros DM. A Comprehensive Analysis of Fibrillar Collagens in Lamprey Suggests a Conserved Role in Vertebrate Musculoskeletal Evolution. Front Cell Dev Biol 2022; 10:809979. [PMID: 35242758 PMCID: PMC8887668 DOI: 10.3389/fcell.2022.809979] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Accepted: 01/18/2022] [Indexed: 12/03/2022] Open
Abstract
Vertebrates have distinct tissues which are not present in invertebrate chordates nor other metazoans. The rise of these tissues also coincided with at least one round of whole-genome duplication as well as a suite of lineage-specific segmental duplications. Understanding whether novel genes lead to the origin and diversification of novel cell types, therefore, is of great importance in vertebrate evolution. Here we were particularly interested in the evolution of the vertebrate musculoskeletal system, the muscles and connective tissues that support a diversity of body plans. A major component of the musculoskeletal extracellular matrix (ECM) is fibrillar collagens, a gene family which has been greatly expanded upon in vertebrates. We thus asked whether the repertoire of fibrillar collagens in vertebrates reflects differences in the musculoskeletal system. To test this, we explored the diversity of fibrillar collagens in lamprey, a jawless vertebrate which diverged from jawed vertebrates (gnathostomes) more than five hundred million years ago and has undergone its own gene duplications. Some of the principal components of vertebrate hyaline cartilage are the fibrillar collagens type II and XI, but their presence in cartilage development across all vertebrate taxa has been disputed. We particularly emphasized the characterization of genes in the lamprey hyaline cartilage, testing if its collagen repertoire was similar to that in gnathostomes. Overall, we discovered thirteen fibrillar collagens from all known gene subfamilies in lamprey and were able to identify several lineage-specific duplications. We found that, while the collagen loci have undergone rearrangement, the Clade A genes have remained linked with the hox clusters, a phenomenon also seen in gnathostomes. While the lamprey muscular tissue was largely similar to that seen in gnathostomes, we saw considerable differences in the larval lamprey skeletal tissue, with distinct collagen combinations pertaining to different cartilage types. Our gene expression analyses were unable to identify type II collagen in the sea lamprey hyaline cartilage nor any other fibrillar collagen during chondrogenesis at the stages observed, meaning that sea lamprey likely no longer require these genes during early cartilage development. Our findings suggest that fibrillar collagens were multifunctional across the musculoskeletal system in the last common ancestor of vertebrates and have been largely conserved, but these genes alone cannot explain the origin of novel cell types.
Collapse
Affiliation(s)
- Zachary D Root
- Department of Ecology and Evolutionary Biology, University of Colorado, Boulder, CO, United States
| | - Cara Allen
- Department of Ecology and Evolutionary Biology, University of Colorado, Boulder, CO, United States
| | - Claire Gould
- Department of Ecology and Evolutionary Biology, University of Colorado, Boulder, CO, United States
| | - Margaux Brewer
- Department of Ecology and Evolutionary Biology, University of Colorado, Boulder, CO, United States
| | - David Jandzik
- Department of Ecology and Evolutionary Biology, University of Colorado, Boulder, CO, United States.,Department of Zoology, Comenius University in Bratislava, Bratislava, Slovakia
| | - Daniel M Medeiros
- Department of Ecology and Evolutionary Biology, University of Colorado, Boulder, CO, United States
| |
Collapse
|
8
|
Della Gaspera B, Weill L, Chanoine C. Evolution of Somite Compartmentalization: A View From Xenopus. Front Cell Dev Biol 2022; 9:790847. [PMID: 35111756 PMCID: PMC8802780 DOI: 10.3389/fcell.2021.790847] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Accepted: 11/26/2021] [Indexed: 11/13/2022] Open
Abstract
Somites are transitory metameric structures at the basis of the axial organization of vertebrate musculoskeletal system. During evolution, somites appear in the chordate phylum and compartmentalize mainly into the dermomyotome, the myotome, and the sclerotome in vertebrates. In this review, we summarized the existing literature about somite compartmentalization in Xenopus and compared it with other anamniote and amniote vertebrates. We also present and discuss a model that describes the evolutionary history of somite compartmentalization from ancestral chordates to amniote vertebrates. We propose that the ancestral organization of chordate somite, subdivided into a lateral compartment of multipotent somitic cells (MSCs) and a medial primitive myotome, evolves through two major transitions. From ancestral chordates to vertebrates, the cell potency of MSCs may have evolved and gave rise to all new vertebrate compartments, i.e., the dermomyome, its hypaxial region, and the sclerotome. From anamniote to amniote vertebrates, the lateral MSC territory may expand to the whole somite at the expense of primitive myotome and may probably facilitate sclerotome formation. We propose that successive modifications of the cell potency of some type of embryonic progenitors could be one of major processes of the vertebrate evolution.
Collapse
|
9
|
Eve A. An interview with Michael Barresi. Development 2021. [DOI: 10.1242/dev.200011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Michael Barresi is Professor of Biological Sciences at Smith College, Northampton, MA, USA, where he uses the zebrafish to understand central nervous system development. Michael is also Program Director of the ‘Student Scientists’ outreach project and has made significant contributions to teaching developmental biology, including being co-author and illustrator of the textbook Developmental Biology, producing developmental documentaries and starting the Online Developmental Teaching Forums. He was awarded the 2021 Viktor Hamburger Outstanding Educator Prize from the Society of Developmental Biology (SDB). We caught up with Michael over Teams to hear more about his career and love of learning.
Collapse
|
10
|
Truong ME, Bilekova S, Choksi SP, Li W, Bugaj LJ, Xu K, Reiter JF. Vertebrate cells differentially interpret ciliary and extraciliary cAMP. Cell 2021; 184:2911-2926.e18. [PMID: 33932338 DOI: 10.1016/j.cell.2021.04.002] [Citation(s) in RCA: 69] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2020] [Revised: 02/08/2021] [Accepted: 03/31/2021] [Indexed: 12/12/2022]
Abstract
Hedgehog pathway components and select G protein-coupled receptors (GPCRs) localize to the primary cilium, an organelle specialized for signal transduction. We investigated whether cells distinguish between ciliary and extraciliary GPCR signaling. To test whether ciliary and extraciliary cyclic AMP (cAMP) convey different information, we engineered optogenetic and chemogenetic tools to control the subcellular site of cAMP generation. Generating equal amounts of ciliary and cytoplasmic cAMP in zebrafish and mammalian cells revealed that ciliary cAMP, but not cytoplasmic cAMP, inhibited Hedgehog signaling. Modeling suggested that the distinct geometries of the cilium and cell body differentially activate local effectors. The search for effectors identified a ciliary pool of protein kinase A (PKA). Blocking the function of ciliary PKA, but not extraciliary PKA, activated Hedgehog signal transduction and reversed the effects of ciliary cAMP. Therefore, cells distinguish ciliary and extraciliary cAMP using functionally and spatially distinct pools of PKA, and different subcellular pools of cAMP convey different information.
Collapse
Affiliation(s)
- Melissa E Truong
- Department of Biochemistry and Biophysics, Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Sara Bilekova
- Department of Biochemistry and Biophysics, Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA 94158, USA; Ludwig-Maximilians-Universität München, Munich 80539, Germany
| | - Semil P Choksi
- Department of Biochemistry and Biophysics, Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Wan Li
- Department of Chemistry, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Lukasz J Bugaj
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Ke Xu
- Department of Chemistry, University of California, Berkeley, Berkeley, CA 94720, USA; Chan Zuckerberg Biohub, San Francisco, San Francisco, CA 94158, USA
| | - Jeremy F Reiter
- Department of Biochemistry and Biophysics, Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA 94158, USA; Chan Zuckerberg Biohub, San Francisco, San Francisco, CA 94158, USA.
| |
Collapse
|
11
|
Hromowyk KJ, Talbot JC, Martin BL, Janssen PML, Amacher SL. Cell fusion is differentially regulated in zebrafish post-embryonic slow and fast muscle. Dev Biol 2020; 462:85-100. [PMID: 32165147 PMCID: PMC7225055 DOI: 10.1016/j.ydbio.2020.03.005] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Revised: 02/08/2020] [Accepted: 03/03/2020] [Indexed: 12/13/2022]
Abstract
Skeletal muscle fusion occurs during development, growth, and regeneration. To investigate how muscle fusion compares among different muscle cell types and developmental stages, we studied muscle cell fusion over time in wild-type, myomaker (mymk), and jam2a mutant zebrafish. Using live imaging, we show that embryonic myoblast elongation and fusion correlate tightly with slow muscle cell migration. In wild-type embryos, only fast muscle fibers are multinucleate, consistent with previous work showing that the cell fusion regulator gene mymk is specifically expressed throughout the embryonic fast muscle domain. However, by 3 weeks post-fertilization, slow muscle fibers also become multinucleate. At this late-larval stage, mymk is not expressed in muscle fibers, but is expressed in small cells near muscle fibers. Although previous work showed that both mymk and jam2a are required for embryonic fast muscle cell fusion, we observe that muscle force and function is almost normal in mymk and jam2a mutant embryos, despite the lack of fast muscle multinucleation. We show that genetic requirements change post-embryonically, with jam2a becoming much less important by late-larval stages and mymk now required for muscle fusion and growth in both fast and slow muscle cell types. Correspondingly, adult mymk mutants perform poorly in sprint and endurance tests compared to wild-type and jam2a mutants. We show that adult mymk mutant muscle contains small mononucleate myofibers with average myonuclear domain size equivalent to that in wild type adults. The mymk mutant fibers have decreased Laminin expression and increased numbers of Pax7-positive cells, suggesting that impaired fiber growth and active regeneration contribute to the muscle phenotype. Our findings identify several aspects of muscle fusion that change with time in slow and fast fibers as zebrafish develop beyond embryonic stages.
Collapse
Affiliation(s)
- Kimberly J Hromowyk
- Department of Molecular Genetics and Biological Chemistry and Pharmacology, The Ohio State University, Columbus, OH, 43210, USA; Center for Muscle Health and Neuromuscular Disorders, The Ohio State University and Nationwide Children's Hospital, Columbus, OH, 43210, USA; Molecular, Cellular, and Developmental Biology Graduate Program, The Ohio State University, Columbus, OH, 43210, USA
| | - Jared C Talbot
- Department of Molecular Genetics and Biological Chemistry and Pharmacology, The Ohio State University, Columbus, OH, 43210, USA; Center for Muscle Health and Neuromuscular Disorders, The Ohio State University and Nationwide Children's Hospital, Columbus, OH, 43210, USA.
| | - Brit L Martin
- Center for Muscle Health and Neuromuscular Disorders, The Ohio State University and Nationwide Children's Hospital, Columbus, OH, 43210, USA; Molecular, Cellular, and Developmental Biology Graduate Program, The Ohio State University, Columbus, OH, 43210, USA; Department of Physiology and Cell Biology, The Ohio State University, Columbus, OH, 43210, USA
| | - Paul M L Janssen
- Center for Muscle Health and Neuromuscular Disorders, The Ohio State University and Nationwide Children's Hospital, Columbus, OH, 43210, USA; Department of Physiology and Cell Biology, The Ohio State University, Columbus, OH, 43210, USA; Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH, 43210, USA
| | - Sharon L Amacher
- Department of Molecular Genetics and Biological Chemistry and Pharmacology, The Ohio State University, Columbus, OH, 43210, USA; Center for Muscle Health and Neuromuscular Disorders, The Ohio State University and Nationwide Children's Hospital, Columbus, OH, 43210, USA; Center for RNA Biology, The Ohio State University, Columbus, OH, 43210, USA.
| |
Collapse
|
12
|
Li S, Wen H, Du S. Defective sarcomere organization and reduced larval locomotion and fish survival in slow muscle heavy chain 1 (smyhc1) mutants. FASEB J 2020; 34:1378-1397. [PMID: 31914689 PMCID: PMC6956737 DOI: 10.1096/fj.201900935rr] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Revised: 11/07/2019] [Accepted: 11/09/2019] [Indexed: 11/11/2022]
Abstract
Zebrafish skeletal muscles are broadly divided into slow-twitch and fast-twitch muscle fibers. The slow fibers, which express a slow fiber-specific myosin heavy chain 1 (Smyhc1), are the first group of muscle fibers formed during myogenesis. To uncover Smyhc1 function in muscle growth, we generated three mutant alleles with reading frame shift mutations in the zebrafish smyhc1 gene using CRISPR. The mutants showed shortened sarcomeres with no thick filaments and M-lines in slow fibers of the mutant embryos. However, the formation of slow muscle precursors and expression of other slow muscle genes were not affected and fast muscles appeared normal. The smyhc1 mutant embryos and larvae showed reduced locomotion and food intake. The mutant larvae exhibited increased lethality of incomplete penetrance. Approximately 2/5 of the homozygous mutants were viable and grew into reproductive adults. These adult mutants displayed a typical pattern of slow and fast muscle fiber distribution, and regained normal slow muscle formation. Together, our studies indicate that Smyhc1 is essential for myogenesis in embryonic slow muscles, and loss of Smyhc1 results in defective sarcomere assembly, reduces larval motility and fish survival, but has no visible impact on muscle growth in juvenile and adult zebrafish that escape the larval lethality.
Collapse
Affiliation(s)
- Siping Li
- Institute of Marine and Environmental Technology, Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, MD 21202, USA
- The Key Laboratory of Mariculture, Ministry of Education, Fishery College of Ocean University of China, Qingdao 266003, China
| | - Haishen Wen
- The Key Laboratory of Mariculture, Ministry of Education, Fishery College of Ocean University of China, Qingdao 266003, China
| | - Shaojun Du
- Institute of Marine and Environmental Technology, Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, MD 21202, USA
| |
Collapse
|
13
|
Sagarin KA, Redgrave AC, Mosimann C, Burke AC, Devoto SH. Anterior trunk muscle shows mix of axial and appendicular developmental patterns. Dev Dyn 2019; 248:961-968. [PMID: 31386244 DOI: 10.1002/dvdy.95] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Revised: 07/04/2019] [Accepted: 07/10/2019] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Skeletal muscle in the trunk derives from the somites, paired segments of paraxial mesoderm. Whereas axial musculature develops within the somite, appendicular muscle develops following migration of muscle precursors into lateral plate mesoderm. The development of muscles bridging axial and appendicular systems appears mixed. RESULTS We examine development of three migratory muscle precursor-derived muscles in zebrafish: the sternohyoideus (SH), pectoral fin (PF), and posterior hypaxial (PHM) muscles. We show there is an anterior to posterior gradient to the developmental gene expression and maturation of these three muscles. SH muscle precursors exhibit a long delay between migration and differentiation, PF muscle precursors exhibit a moderate delay in differentiation, and PHM muscle precursors show virtually no delay between migration and differentiation. Using lineage tracing, we show that lateral plate contribution to the PHM muscle is minor, unlike its known extensive contribution to the PF muscle and absence in the ventral extension of axial musculature. CONCLUSIONS We propose that PHM development is intermediate between a migratory muscle mode and an axial muscle mode of development, wherein the PHM differentiates after a very short migration of its precursors and becomes more anterior primarily by elongation of differentiated muscle fibers.
Collapse
Affiliation(s)
| | - Anna C Redgrave
- Department of Biology, Wesleyan University, Middletown, Connecticut.,Biology Department, Boston College, Chestnut Hill, Massachusetts
| | - Christian Mosimann
- Institute of Molecular Life Sciences, University of Zürich, Zürich, Switzerland
| | - Ann C Burke
- Department of Biology, Wesleyan University, Middletown, Connecticut
| | - Stephen H Devoto
- Department of Biology, Wesleyan University, Middletown, Connecticut
| |
Collapse
|
14
|
Characterization of paralogous uncx transcription factor encoding genes in zebrafish. Gene X 2019; 721S:100011. [PMID: 31193955 PMCID: PMC6543554 DOI: 10.1016/j.gene.2019.100011] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Revised: 02/12/2019] [Accepted: 02/15/2019] [Indexed: 12/23/2022] Open
Abstract
The paired-type homeodomain transcription factor Uncx is involved in multiple processes of embryogenesis in vertebrates. Reasoning that zebrafish genes uncx4.1 and uncx are orthologs of mouse Uncx, we studied their genomic environment and developmental expression. Evolutionary analyses indicate the zebrafish uncx genes as being paralogs deriving from teleost-specific whole-genome duplication. Whole-mount in situ mRNA hybridization of uncx transcripts in zebrafish embryos reveals novel expression domains, confirms those previously known, and suggests sub-functionalization of paralogs. Using genetic mutants and pharmacological inhibitors, we investigate the role of signaling pathways on the expression of zebrafish uncx genes in developing somites. In identifying putative functional role(s) of zebrafish uncx genes, we hypothesized that they encode transcription factors that coordinate growth and innervation of somitic muscles. The Uncx4.1 and Uncx genes derive from the teleost-specific whole-genome duplication. Uncx genes are expressed during embryogenesis in unique and overlapping domains. Uncx gene expression during somite differentiation is regulated by FGF signaling. Synteny and expression profiles correlate Uncx genes with axon guidance.
Collapse
Key Words
- AP, antero-posterior
- Ace, acerebellar
- CAMP, conserved ancestral microsyntenic pairs
- CNE, conserved non-coding elements
- CRM, cis-regulatory module
- CS, Corpuscle of Stannius
- CaP, caudal primary motor neuron axons
- Ce, cerebellum
- Development
- Di, diencephalon
- Elfn1, Extracellular Leucine Rich Repeat And Fibronectin Type III Domain Containing 1
- Ey, eye
- FB, forebrain
- FGF, fibroblast growth factor
- Flh, floating head
- HB, hindbrain
- HM, hybridization mix
- Hy, hypothalamus
- MO, morpholino
- Mical, molecule interacting with CasL
- No, notochord
- OP, olfactory placode
- OT, optic tectum
- PA, pharyngeal arches
- PSM, presomitic mesoderm
- SC, spinal cord
- Shh, sonic hedgehog
- Signaling pathway
- So, somites
- Synteny
- TSGD
- TSGD, teleost-specific genome duplication
- Te, telencephalon
- Th, thalamus
- Uncx
- VLP, ventro-lateral-posterior
- WIHC, whole-mount immunohistochemistry
- WISH, whole-mount in situ hybridization
- YE, yolk extension
- Yo, yolk
- Zebrafish
- cyc, cyclops
- fss, fused-somites
- hpf, hours post fertilization
- ptc, patched
- smu, slow-muscle-omitted
- syu, sonic-you
- yot, you-too
Collapse
|
15
|
Bello-Rojas S, Istrate AE, Kishore S, McLean DL. Central and peripheral innervation patterns of defined axial motor units in larval zebrafish. J Comp Neurol 2019; 527:2557-2572. [PMID: 30919953 DOI: 10.1002/cne.24689] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Revised: 03/20/2019] [Accepted: 03/25/2019] [Indexed: 02/06/2023]
Abstract
Spinal motor neurons and the peripheral muscle fibers they innervate form discrete motor units that execute movements of varying force and speed. Subsets of spinal motor neurons also exhibit axon collaterals that influence motor output centrally. Here, we have used in vivo imaging to anatomically characterize the central and peripheral innervation patterns of axial motor units in larval zebrafish. Using early born "primary" motor neurons and their division of epaxial and hypaxial muscle into four distinct quadrants as a reference, we define three distinct types of later born "secondary" motor units. The largest is "m-type" units, which innervate deeper fast-twitch muscle fibers via medial nerves. Next in size are "ms-type" secondaries, which innervate superficial fast-twitch and slow fibers via medial and septal nerves, followed by "s-type" units, which exclusively innervate superficial slow muscle fibers via septal nerves. All types of secondaries innervate up to four axial quadrants. Central axon collaterals are found in subsets of primaries based on soma position and predominantly in secondary fast-twitch units (m, ms) with increasing likelihood based on number of quadrants innervated. Collaterals are labeled by synaptophysin-tagged fluorescent proteins, but not PSD95, consistent with their output function. Also, PSD95 dendrite labeling reveals that larger motor units receive more excitatory synaptic input. Collaterals are largely restricted to the neuropil, however, perisomatic connections are observed between motor units. These observations suggest that recurrent interactions are dominated by motor neurons recruited during stronger movements and set the stage for functional investigations of recurrent motor circuitry in larval zebrafish.
Collapse
Affiliation(s)
- Saul Bello-Rojas
- Interdepartmental Neuroscience Postbaccalaureate Research Education Program, Northwestern University, Evanston, Illinois
| | - Ana E Istrate
- Masters Program in Neurobiology, Northwestern University, Evanston, Illinois
| | - Sandeep Kishore
- Department of Neurobiology, Northwestern University, Evanston, Illinois
| | - David L McLean
- Interdepartmental Neuroscience Postbaccalaureate Research Education Program, Northwestern University, Evanston, Illinois.,Masters Program in Neurobiology, Northwestern University, Evanston, Illinois.,Department of Neurobiology, Northwestern University, Evanston, Illinois
| |
Collapse
|
16
|
Landemaine A, Ramirez-Martinez A, Monestier O, Sabin N, Rescan PY, Olson EN, Gabillard JC. Trout myomaker contains 14 minisatellites and two sequence extensions but retains fusogenic function. J Biol Chem 2019; 294:6364-6374. [PMID: 30819805 DOI: 10.1074/jbc.ra118.006047] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Revised: 02/26/2019] [Indexed: 01/20/2023] Open
Abstract
The formation of new myofibers in vertebrates occurs by myoblast fusion and requires fusogenic activity of the muscle-specific membrane protein myomaker. Here, using in silico (BLAST) genome analyses, we show that the myomaker gene from trout includes 14 minisatellites, indicating that it has an unusual structure compared with those of other animal species. We found that the trout myomaker gene encodes a 434-amino acid (aa) protein, in accordance with its apparent molecular mass (∼40 kDa) observed by immunoblotting. The first half of the trout myomaker protein (1-220 aa) is similar to the 221-aa mouse myomaker protein, whereas the second half (222-234 aa) does not correspond to any known motifs and arises from two protein extensions. The first extension (∼70 aa) apparently appeared with the radiation of the bony fish clade Euteleostei, whereas the second extension (up to 236 aa) is restricted to the superorder Protacanthopterygii (containing salmonids and pike) and corresponds to the insertion of minisatellites having a length of 30 nucleotides. According to gene expression analyses, trout myomaker expression is consistently associated with the formation of new myofibers during embryonic development, postlarval growth, and muscle regeneration. Using cell-mixing experiments, we observed that trout myomaker has retained the ability to drive the fusion of mouse fibroblasts with C2C12 myoblasts. Our work reveals that trout myomaker has fusogenic function despite containing two protein extensions.
Collapse
Affiliation(s)
- Aurélie Landemaine
- From the Institut National de la Recherche Agronomique, UR1037 Laboratory of Fish Physiology and Genomics, 35000 Rennes, France
| | - Andres Ramirez-Martinez
- the Department of Molecular Biology, Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, Texas 75390, and
| | - Olivier Monestier
- Institute of Interdisciplinary Research in Human and Molecular Biology, Université Libre de Bruxelles, 1070 Bruxelles, Belgium
| | - Nathalie Sabin
- From the Institut National de la Recherche Agronomique, UR1037 Laboratory of Fish Physiology and Genomics, 35000 Rennes, France
| | - Pierre-Yves Rescan
- From the Institut National de la Recherche Agronomique, UR1037 Laboratory of Fish Physiology and Genomics, 35000 Rennes, France
| | - Eric N Olson
- the Department of Molecular Biology, Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, Texas 75390, and
| | - Jean-Charles Gabillard
- From the Institut National de la Recherche Agronomique, UR1037 Laboratory of Fish Physiology and Genomics, 35000 Rennes, France,
| |
Collapse
|
17
|
Roy NM, Zambrzycka E, Santangelo J. Butyl benzyl phthalate (BBP) induces caudal defects during embryonic development. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2017; 56:129-135. [PMID: 28934690 DOI: 10.1016/j.etap.2017.09.009] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Revised: 09/11/2017] [Accepted: 09/14/2017] [Indexed: 06/07/2023]
Abstract
Butyl benzyl phthalate (BBP) is commonly added during the manufacturing of plastics to increase flexibility and elasticity. However, BBP leaches off of plastic and environment presence has been detected in soil, groundwater and sediment potentially effecting organisms in the environment. Given the widespread uses of BBP in household, consumer goods and the presence of BBP in the environment, studies on developmental toxicity are needed. Here, we use a zebrafish model to investigate the early developmental toxicity of BBP. We treated gastrula staged embryos with increasing concentrations of BBP and noted concentration-dependent defects in caudal tail development, but the effect was caudal specific with no other developmental defects noted. In situ hybridization studies using muscle and notochord markers show alterations in muscle development and non-linear, kinked notochord staining. A more detailed antibody staining using a myosin specific marker shows disorganized myofibrils and a loss of chevron shaped somites. Furthermore, vascular development in the tail was also disrupted in a concentration dependent manner. We conclude that BBP is toxic to caudal development in zebrafish. The sensitivity of zebrafish during development to environmental toxins and chemicals has been useful in assessing the health of the aquatic environment. The results presented here are a useful early warning system for contamination that could affect human health.
Collapse
Affiliation(s)
- Nicole M Roy
- Department of Biology, Sacred Heart University, 5151 Park Ave, Fairfield, CT, 06825, United States.
| | - Ewelina Zambrzycka
- Department of Biology, Sacred Heart University, 5151 Park Ave, Fairfield, CT, 06825, United States
| | - Jenna Santangelo
- Department of Biology, Sacred Heart University, 5151 Park Ave, Fairfield, CT, 06825, United States
| |
Collapse
|
18
|
Nuclear Pores Regulate Muscle Development and Maintenance by Assembling a Localized Mef2C Complex. Dev Cell 2017; 41:540-554.e7. [PMID: 28586646 DOI: 10.1016/j.devcel.2017.05.007] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2016] [Revised: 04/29/2017] [Accepted: 05/08/2017] [Indexed: 02/08/2023]
Abstract
Nuclear pore complexes (NPCs) are multiprotein channels connecting the nucleus with the cytoplasm. NPCs have been shown to have tissue-specific composition, suggesting that their function can be specialized. However, the physiological roles of NPC composition changes and their impacts on cellular processes remain unclear. Here we show that the addition of the Nup210 nucleoporin to NPCs during myoblast differentiation results in assembly of an Mef2C transcriptional complex required for efficient expression of muscle structural genes and microRNAs. We show that this NPC-localized complex is essential for muscle growth, myofiber maturation, and muscle cell survival and that alterations in its activity result in muscle degeneration. Our findings suggest that NPCs regulate the activity of functional gene groups by acting as scaffolds that promote the local assembly of tissue-specific transcription complexes and show how nuclear pore composition changes can be exploited to regulate gene expression at the nuclear periphery.
Collapse
|
19
|
Roy SD, Williams VC, Pipalia TG, Li K, Hammond CL, Knappe S, Knight RD, Hughes SM. Myotome adaptability confers developmental robustness to somitic myogenesis in response to fibre number alteration. Dev Biol 2017; 431:321-335. [PMID: 28887016 PMCID: PMC5667637 DOI: 10.1016/j.ydbio.2017.08.029] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2017] [Revised: 05/22/2017] [Accepted: 08/26/2017] [Indexed: 12/31/2022]
Abstract
Balancing the number of stem cells and their progeny is crucial for tissue development and repair. Here we examine how cell numbers and overall muscle size are tightly regulated during zebrafish somitic muscle development. Muscle stem/precursor cell (MPCs) expressing Pax7 are initially located in the dermomyotome (DM) external cell layer, adopt a highly stereotypical distribution and thereafter a proportion of MPCs migrate into the myotome. Regional variations in the proliferation and terminal differentiation of MPCs contribute to growth of the myotome. To probe the robustness of muscle size control and spatiotemporal regulation of MPCs, we compared the behaviour of wild type (wt) MPCs with those in mutant zebrafish that lack the muscle regulatory factor Myod. Myodfh261 mutants form one third fewer multinucleate fast muscle fibres than wt and show a significant expansion of the Pax7+ MPC population in the DM. Subsequently, myodfh261 mutant fibres generate more cytoplasm per nucleus, leading to recovery of muscle bulk. In addition, relative to wt siblings, there is an increased number of MPCs in myodfh261 mutants and these migrate prematurely into the myotome, differentiate and contribute to the hypertrophy of existing fibres. Thus, homeostatic reduction of the excess MPCs returns their number to normal levels, but fibre numbers remain low. The GSK3 antagonist BIO prevents MPC migration into the deep myotome, suggesting that canonical Wnt pathway activation maintains the DM in zebrafish, as in amniotes. BIO does not, however, block recovery of the myodfh261 mutant myotome, indicating that homeostasis acts on fibre intrinsic growth to maintain muscle bulk. The findings suggest the existence of a critical window for early fast fibre formation followed by a period in which homeostatic mechanisms regulate myotome growth by controlling fibre size. The feedback controls we reveal in muscle help explain the extremely precise grading of myotome size along the body axis irrespective of fish size, nutrition and genetic variation and may form a paradigm for wider matching of organ size. A critical window for early muscle fibre formation is proposed. Fish lacking MyoD1 form fewer muscle fibres, but have more myogenic stem cells. Stem cell numbers rapidly return to normal during subsequent development. GSK3 activity promotes and MyoD1 delays myoblast migration into the myotome. Compensatory fibre size increase ensures robustness of overall muscle size.
Collapse
Affiliation(s)
- Shukolpa D Roy
- Randall Division of Cell and Molecular Biophysics, New Hunt's House, Guy's Campus, King's College London, London SE1 1UL, UK
| | - Victoria C Williams
- Randall Division of Cell and Molecular Biophysics, New Hunt's House, Guy's Campus, King's College London, London SE1 1UL, UK
| | - Tapan G Pipalia
- Randall Division of Cell and Molecular Biophysics, New Hunt's House, Guy's Campus, King's College London, London SE1 1UL, UK
| | - Kuoyu Li
- Randall Division of Cell and Molecular Biophysics, New Hunt's House, Guy's Campus, King's College London, London SE1 1UL, UK
| | - Christina L Hammond
- Randall Division of Cell and Molecular Biophysics, New Hunt's House, Guy's Campus, King's College London, London SE1 1UL, UK
| | - Stefanie Knappe
- Division of Craniofacial Development and Stem Cell Biology, Guy's Hospital, King's College London, UK
| | - Robert D Knight
- Division of Craniofacial Development and Stem Cell Biology, Guy's Hospital, King's College London, UK
| | - Simon M Hughes
- Randall Division of Cell and Molecular Biophysics, New Hunt's House, Guy's Campus, King's College London, London SE1 1UL, UK.
| |
Collapse
|
20
|
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
|
21
|
Roy NM, Carneiro B, Ochs J. Glyphosate induces neurotoxicity in zebrafish. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2016; 42:45-54. [PMID: 26773362 DOI: 10.1016/j.etap.2016.01.003] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/24/2015] [Revised: 12/29/2015] [Accepted: 01/01/2016] [Indexed: 05/20/2023]
Abstract
Glyphosate based herbicides (GBH) like Roundup(®) are used extensively in agriculture as well as in urban and rural settings as a broad spectrum herbicide. Its mechanism of action was thought to be specific only to plants and thus considered safe and non-toxic. However, mounting evidence suggests that GBHs may not be as safe as once thought as initial studies in frogs suggest that GBHs may be teratogenic. Here we utilize the zebrafish vertebrate model system to study early effects of glyphosate exposure using technical grade glyphosate and the Roundup(®) Classic formulation. We find morphological abnormalities including cephalic and eye reductions and a loss of delineated brain ventricles. Concomitant with structural changes in the developing brain, using in situ hybridization analysis, we detect decreases in genes expressed in the eye, fore and midbrain regions of the brain including pax2, pax6, otx2 and ephA4. However, we do not detect changes in hindbrain expression domains of ephA4 nor exclusive hindbrain markers krox-20 and hoxb1a. Additionally, using a Retinoic Acid (RA) mediated reporter transgenic, we detect no alterations in the RA expression domains in the hindbrain and spinal cord, but do detect a loss of expression in the retina. We conclude that glyphosate and the Roundup(®) formulation is developmentally toxic to the forebrain and midbrain but does not affect the hindbrain after 24 h exposure.
Collapse
Affiliation(s)
- Nicole M Roy
- Department of Biology, Sacred Heart University, Fairfield, CT, United States.
| | - Bruno Carneiro
- Department of Biology, Sacred Heart University, Fairfield, CT, United States
| | - Jeremy Ochs
- Department of Biology, Sacred Heart University, Fairfield, CT, United States
| |
Collapse
|
22
|
5′-flanking sequences of zebrafish fast myosin heavy chain genes regulate unique expression in the anterior, medial subsection and posterior tail somites of the skeletal muscle. Comp Biochem Physiol B Biochem Mol Biol 2016; 191:1-12. [DOI: 10.1016/j.cbpb.2015.08.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2015] [Revised: 08/18/2015] [Accepted: 08/18/2015] [Indexed: 11/18/2022]
|
23
|
Ahammad AS, Asaduzzaman M, Asakawa S, Watabe S, Kinoshita S. Regulation of gene expression mediating indeterminate muscle growth in teleosts. Mech Dev 2015; 137:53-65. [DOI: 10.1016/j.mod.2015.02.006] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2014] [Revised: 01/19/2015] [Accepted: 02/02/2015] [Indexed: 01/13/2023]
|
24
|
Roy NM, DeWolf S, Carneiro B. Evaluation of the developmental toxicity of lead in the Danio rerio body. AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2015; 158:138-148. [PMID: 25438119 DOI: 10.1016/j.aquatox.2014.10.026] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/03/2014] [Revised: 10/30/2014] [Accepted: 10/31/2014] [Indexed: 06/04/2023]
Abstract
Lead has been utilized throughout history and is widely distributed and mobilized globally. Although lead in the environment has been somewhat mitigated, the nature of lead and its extensive uses in the past prohibit it from being completely absent from our environment and exposure to lead is still a public health concern. Most studies regarding lead toxicity have focused on the brain. However, little is found in the literature on the effects of lead in other tissues. Here, we utilize the zebrafish model system to investigate effects of lead exposure during early developmental time windows at 24, 48 and 72 h post fertilization in the body. We analyze whole body, notochord and somatic muscle changes, vascular changes of the body, as well as motor neuron alterations. We find lead exposure induces a curved body phenotype with concomitant changes in somite length, decreased notochord staining and abnormal muscle staining using live and in situ approaches. Furthermore, altered vasculature within the somatic regions, loss and/or alternations of motor neuron extension both dorsally and ventrally from the spinal cord, loss of Rohon-Beard sensory neurons, and increased areas of apoptosis were found. We conclude that lead is developmentally toxic to other areas of the developing embryo, not just the brain.
Collapse
Affiliation(s)
- Nicole M Roy
- Department of Biology, Sacred Heart University, Fairfield CT, United States.
| | - Sarah DeWolf
- Department of Biology, Sacred Heart University, Fairfield CT, United States
| | - Bruno Carneiro
- Department of Biology, Sacred Heart University, Fairfield CT, United States
| |
Collapse
|
25
|
Rost F, Eugster C, Schröter C, Oates AC, Brusch L. Chevron formation of the zebrafish muscle segments. J Exp Biol 2014; 217:3870-82. [PMID: 25267843 PMCID: PMC4213178 DOI: 10.1242/jeb.102202] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2014] [Accepted: 09/04/2014] [Indexed: 01/16/2023]
Abstract
The muscle segments of fish have a folded shape, termed a chevron, which is thought to be optimal for the undulating body movements of swimming. However, the mechanism shaping the chevron during embryogenesis is not understood. Here, we used time-lapse microscopy of developing zebrafish embryos spanning the entire somitogenesis period to quantify the dynamics of chevron shape development. By comparing such time courses with the start of movements in wildtype zebrafish and analysing immobile mutants, we show that the previously implicated body movements do not play a role in chevron formation. Further, the monotonic increase of chevron angle along the anteroposterior axis revealed by our data constrains or rules out possible contributions by previously proposed mechanisms. In particular, we found that muscle pioneers are not required for chevron formation. We put forward a tension-and-resistance mechanism involving interactions between intra-segmental tension and segment boundaries. To evaluate this mechanism, we derived and analysed a mechanical model of a chain of contractile and resisting elements. The predictions of this model were verified by comparison with experimental data. Altogether, our results support the notion that a simple physical mechanism suffices to self-organize the observed spatiotemporal pattern in chevron formation.
Collapse
Affiliation(s)
- Fabian Rost
- Center for Information Services and High-Performance Computing, Technische Universität Dresden, 01062 Dresden, Germany
| | - Christina Eugster
- Max Planck Institute of Molecular Cell Biology and Genetics, 01307 Dresden, Germany
| | - Christian Schröter
- Max Planck Institute of Molecular Cell Biology and Genetics, 01307 Dresden, Germany
| | - Andrew C Oates
- Max Planck Institute of Molecular Cell Biology and Genetics, 01307 Dresden, Germany MRC-National Institute for Medical Research, The Ridgeway, Mill Hill, London NW7 1AA, UK Department of Cell and Developmental Biology, University College London, Gower Street, London WC1E 6BT, UK
| | - Lutz Brusch
- Center for Information Services and High-Performance Computing, Technische Universität Dresden, 01062 Dresden, Germany
| |
Collapse
|
26
|
Subramanian A, Schilling TF. Thrombospondin-4 controls matrix assembly during development and repair of myotendinous junctions. eLife 2014; 3. [PMID: 24941943 PMCID: PMC4096842 DOI: 10.7554/elife.02372] [Citation(s) in RCA: 94] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2014] [Accepted: 06/17/2014] [Indexed: 12/13/2022] Open
Abstract
Tendons are extracellular matrix (ECM)-rich structures that mediate muscle attachments with the skeleton, but surprisingly little is known about molecular mechanisms of attachment. Individual myofibers and tenocytes in Drosophila interact through integrin (Itg) ligands such as Thrombospondin (Tsp), while vertebrate muscles attach to complex ECM fibrils embedded with tenocytes. We show for the first time that a vertebrate thrombospondin, Tsp4b, is essential for muscle attachment and ECM assembly at myotendinous junctions (MTJs). Tsp4b depletion in zebrafish causes muscle detachment upon contraction due to defects in laminin localization and reduced Itg signaling at MTJs. Mutation of its oligomerization domain renders Tsp4b unable to rescue these defects, demonstrating that pentamerization is required for ECM assembly. Furthermore, injected human TSP4 localizes to zebrafish MTJs and rescues muscle detachment and ECM assembly in Tsp4b-deficient embryos. Thus Tsp4 functions as an ECM scaffold at MTJs, with potential therapeutic uses in tendon strengthening and repair. DOI:http://dx.doi.org/10.7554/eLife.02372.001 Tendons, the tough connective tissues that link muscles to bones, are essential for lifting, running and other movements in animals. A matrix of proteins, called the extracellular matrix, connects the cells in a tendon, giving it the strength it needs to prevent muscles from detaching from bones during strenuous activities. To achieve this strength, extracellular matrix proteins bind to one another and to receptors on the muscle cell surface that are linked to its internal scaffolding, thereby organizing other proteins into a structure called a myotendinous junction. However, despite the essential roles of tendons, scientists do not fully understand how this organization occurs, or how it can go awry. Subramanian and Schilling screened zebrafish for genes that are essential for proper muscle attachment, and zeroed in on a gene encoding a protein called Thrombospondin-4b (Tsp4b). A similar protein helps to connect muscle and tendon cells in fruit flies. Without Tsp4b, zebrafish are able to form connections between muscles and tendons, but the muscles detach easily during movement. This weakened connection is caused by disorganization of the proteins in the extracellular matrix, which results in reduced signaling from the muscle cell receptors. When a human form of this protein was injected into zebrafish embryos lacking Tsp4b, it settled into the junctions between muscle and tendon cells. The human protein repaired the detached muscles and restored the proper organization of the matrix. This improved the strength of the muscle-tendon attachment in the treated fish embryos, suggesting that similar injections could also help to strengthen and repair muscles and tendons in people. DOI:http://dx.doi.org/10.7554/eLife.02372.002
Collapse
Affiliation(s)
- Arul Subramanian
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, United States
| | - Thomas F Schilling
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, United States
| |
Collapse
|
27
|
Rossi G, Messina G. Comparative myogenesis in teleosts and mammals. Cell Mol Life Sci 2014; 71:3081-99. [PMID: 24664432 PMCID: PMC4111864 DOI: 10.1007/s00018-014-1604-5] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2013] [Revised: 02/17/2014] [Accepted: 03/06/2014] [Indexed: 01/02/2023]
Abstract
Skeletal myogenesis has been and is currently under extensive study in both mammals and teleosts, with the latter providing a good model for skeletal myogenesis because of their flexible and conserved genome. Parallel investigations of muscle studies using both these models have strongly accelerated the advances in the field. However, when transferring the knowledge from one model to the other, it is important to take into account both their similarities and differences. The main difficulties in comparing mammals and teleosts arise from their different temporal development. Conserved aspects can be seen for muscle developmental origin and segmentation, and for the presence of multiple myogenic waves. Among the divergences, many fish have an indeterminate growth capacity throughout their entire life span, which is absent in mammals, thus implying different post-natal growth mechanisms. This review covers the current state of the art on myogenesis, with a focus on the most conserved and divergent aspects between mammals and teleosts.
Collapse
Affiliation(s)
- Giuliana Rossi
- Department of Biosciences, University of Milan, 20133, Milan, Italy
| | | |
Collapse
|
28
|
Jackson HE, Ingham PW. Control of muscle fibre-type diversity during embryonic development: the zebrafish paradigm. Mech Dev 2013; 130:447-57. [PMID: 23811405 DOI: 10.1016/j.mod.2013.06.001] [Citation(s) in RCA: 70] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2013] [Revised: 06/04/2013] [Accepted: 06/04/2013] [Indexed: 01/01/2023]
Abstract
Vertebrate skeletal muscle is composed of distinct types of fibre that are functionally adapted through differences in their physiological and metabolic properties. An understanding of the molecular basis of fibre-type specification is of relevance to human health and fitness. The zebrafish provides an attractive model for investigating fibre type specification; not only are their rapidly developing embryos optically transparent, but in contrast to amniotes, the embryonic myotome shows a discrete temporal and spatial separation of fibre type ontogeny that simplifies its analysis. Here we review the current state of understanding of muscle fibre type specification and differentiation during embryonic development of the zebrafish, with a particular focus on the roles of the Prdm1a and Sox6 transcription factors, and consider the relevance of these findings to higher vertebrate muscle biology.
Collapse
Affiliation(s)
- Harriet E Jackson
- Institute of Molecular and Cell Biology, 61 Biopolis Drive, Proteos, Singapore 138673, Singapore
| | | |
Collapse
|
29
|
Pistocchi A, Gaudenzi G, Foglia E, Monteverde S, Moreno-Fortuny A, Pianca A, Cossu G, Cotelli F, Messina G. Conserved and divergent functions of Nfix in skeletal muscle development during vertebrate evolution. Development 2013; 140:1528-36. [PMID: 23482488 DOI: 10.1242/dev.076315] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
During mouse skeletal muscle development, the Nfix gene has a pivotal role in regulating fetal-specific transcription. Zebrafish and mice share related programs for muscle development, although zebrafish develops at a much faster rate. In fact, although mouse fetal muscle fibers form after 15 days of development, in fish secondary muscle fibers form by 48 hours post-fertilization in a process that until now has been poorly characterized mechanically. In this work, we studied the zebrafish ortholog Nfix (nfixa) and its role in the proper switch to the secondary myogenic wave. This allowed us to highlight evolutionarily conserved and divergent functions of Nfix. In fact, the knock down of nfixa in zebrafish blocks secondary myogenesis, as in mouse, but also alters primary slow muscle fiber formation. Moreover, whereas Nfix mutant mice are motile, nfixa knockdown zebrafish display impaired motility that probably depends upon disruption of the sarcoplasmic reticulum. We conclude that, during vertebrate evolution, the transcription factor Nfix lost some specific functions, probably as a consequence of the different environment in which teleosts and mammals develop.
Collapse
Affiliation(s)
- Anna Pistocchi
- Department of Biosciences, University of Milan, via Celoria 26, 20133 Milan, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Devakanmalai GS, Zumrut HE, Ozbudak EM. Cited3 activates Mef2c to control muscle cell differentiation and survival. Biol Open 2013; 2:505-14. [PMID: 23789100 PMCID: PMC3654270 DOI: 10.1242/bio.20132550] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2012] [Accepted: 03/25/2013] [Indexed: 01/13/2023] Open
Abstract
Vertebrate muscle development occurs through sequential differentiation of cells residing in somitic mesoderm – a process that is largely governed by transcriptional regulators. Our recent spatiotemporal microarray study in zebrafish has identified functionally uncharacterized transcriptional regulators that are expressed at the initial stages of myogenesis. cited3 is one such novel gene encoding a transcriptional coactivator, which is expressed in the precursors of oxidative slow-twitch myofibers. Our experiments placed cited3 into a gene regulatory network, where it acts downstream of Hedgehog signaling and myoD/myf5 but upstream of mef2c. Knockdown of expression of cited3 by antisense morpholino oligonucleotides impaired muscle cell differentiation and growth, caused muscle cell death and eventually led to total immotility. Transplantation experiments demonstrated that Cited3 cell-autonomously activates the expression of mef2c in slow myofibers, while it non-cell-autonomously regulates expression of structural genes in fast myofibers. Restoring expression of cited3 or mef2c rescued all the cited3 loss-of-function phenotypes. Protein truncation experiments revealed the functional necessity of C-terminally conserved domain of Cited3, which is known to mediate interactions of Cited-family proteins with histone acetylases. Our findings demonstrate that Cited3 is a critical transcriptional coactivator functioning during muscle differentiation and its absence leads to defects in terminal differentiation and survival of muscle cells.
Collapse
|
31
|
Marschallinger J, Obermayer A, Steinbacher P, Stoiber W. The zebrafish myotome contains tonic muscle fibers: morphological characterization and time course of formation. J Morphol 2013; 274:320-30. [PMID: 23280572 DOI: 10.1002/jmor.20095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2012] [Revised: 08/20/2012] [Accepted: 09/24/2012] [Indexed: 11/07/2022]
Abstract
It is long known that the skeletal muscle of teleost fish contains muscle fibers which are in all probability of a tonic type according to morphological criteria. However, the evidence for the existence of teleost tonic fibers is still confined to a very small number of species, and knowledge concerning their ontogeny and possible functions is even more restricted. A remarkable deficit in this context is that it is not even exactly known whether the zebrafish, which is widely used to study vertebrate developmental biology, has such fibers, or how they arise. The present study demonstrates the existence of tonic fibers in the zebrafish myotome. They are identical with a fiber population previously termed "red muscle rim" fibers. A combined histochemical, immunocytochemical, and ultrastructural approach is used to characterize the morphology and development of these fibers. This study provides a basis for using the zebrafish model system in the future research on the developmental regulation and the functions of tonic fibers.
Collapse
Affiliation(s)
- Julia Marschallinger
- Department of Organismic Biology, Division of Zoology and Functional Anatomy, University of Salzburg, Hellbrunnerstr. 34, A-5020 Salzburg, Austria.
| | | | | | | |
Collapse
|
32
|
Asaduzzaman M, Kinoshita S, Bhuiyan SS, Asakawa S, Watabe S. Stimulatory and inhibitory mechanisms of slow muscle-specific myosin heavy chain gene expression in fish: transient and transgenic analysis of torafugu MYH(M86-2) promoter in zebrafish embryos. Exp Cell Res 2012; 319:820-37. [PMID: 23237989 DOI: 10.1016/j.yexcr.2012.11.020] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2012] [Revised: 11/29/2012] [Accepted: 11/30/2012] [Indexed: 12/18/2022]
Abstract
The myosin heavy chain gene, MYHM86-2, exhibited restricted expression in slow muscle fibers of torafugu embryos and larvae, suggesting its functional roles for embryonic and larval muscle development. However, the transcriptional mechanisms involved in its expression are still ambiguous. The present study is the first extensive analysis of slow muscle-specific MYHM86-2 promoter in fish for identifying the cis-elements that are crucial for its expression. Combining both transient transfection and transgenic approaches, we demonstrated that the 2614bp 5'-flanking sequences of MYHM86-2 contain a sufficient promoter activity to drive gene expression specific to superficial slow muscle fibers. By cyclopamine treatment, we also demonstrated that the differentiation of such superficial slow muscle fibers depends on hedgehog signaling activity. The deletion analyses defined an upstream fragment necessary for repressing ectopic MYHM86-2 expression in the fast muscle fibers. The transcriptional mechanism that prevents MYHM86-2 expression in the fast muscle fibers is mediated through Sox6 binding elements. We also demonstrated that Sox6 may function as a transcriptional repressor of MYHM86-2 expression. We further discovered that nuclear factor of activated T cells (NFAT) binding elements plays a key role and myocyte enhancer factor-2 (MEF2) binding elements participate in the transcriptional regulation of MYHM86-2 expression.
Collapse
Affiliation(s)
- Md Asaduzzaman
- Department of Aquatic Bioscience, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Bunkyo, Tokyo 113-8657, Japan
| | | | | | | | | |
Collapse
|
33
|
Asaduzzaman M, Akolkar DB, Kinoshita S, Watabe S. The expression of multiple myosin heavy chain genes during skeletal muscle development of torafugu Takifugu rubripes embryos and larvae. Gene 2012. [PMID: 23201422 DOI: 10.1016/j.gene.2012.10.089] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
In vertebrates, the development-dependent and tissue-specific expression of myosin heavy chain (MYH) genes (MYHs) contributes to the formation of diverged muscle fiber types. The expression patterns of developmentally regulated MYHs have been investigated in certain species of fish. However, the expression profiles of MYHs during torafugu Takifugu rubripes development, although extensively studied in adult tissues, have not been sufficiently studied, and also the expression orders of MYHs during development have remained unclear. In the present study, we comprehensively cloned four MYHs (MYH(M743-2), MYH(M86-2), MYH(M5) and MYH(M2126-1)) from embryos, and two MYHs (MYH(M2528-1) and MYH(M1034)) from larvae, and characterized their expression pattern in relation to developmental stages of torafugu by reverse transcription (RT)-PCR and in situ hybridization. The expression of MYHs from torafugu embryos and larvae appeared sequentially and varied largely in relation to the developmental stage-dependent and fibers-type-specific manners. The transcripts of MYH(M743-2) appeared first in embryos at 3 days post fertilization (dpf) and were localized in the epaxial and hypaxial domains of fast muscle fibers of larval myotome, whereas those of MYH(M5) and MYH(M86-2) in 3 dpf and 4 dpf, respectively, and both were localized in superficial slow and horizontal myoseptum regions. The expression of MYH(M1034) and MYH(M2126-1) was quite low and mostly undetectable. Different MYHs from torafugu embryos and larvae have also been found to be expressed differentially in pectoral fin and craniofacial muscles. Interestingly, the transcripts of MYH(M2528-1) first appeared at 6 dpf and were distinctly expressed at the dorsal and ventral extremes of larval myotome, suggesting its involvement in stratified hyperplasia. The novel involvement of MYH(M2528-1) in mosaic hyperplasia was further confirmed in juvenile torafugu, where the transcripts were expressed in fast fibers with small diameters as well as the inner part of superficial slow fibers.
Collapse
Affiliation(s)
- Md Asaduzzaman
- Department of Aquatic Bioscience, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Bunkyo, Tokyo 113-8657, Japan
| | | | | | | |
Collapse
|
34
|
Bower NI, de la Serrana DG, Cole NJ, Hollway GE, Lee HT, Assinder S, Johnston IA. Stac3 is required for myotube formation and myogenic differentiation in vertebrate skeletal muscle. J Biol Chem 2012; 287:43936-49. [PMID: 23076145 DOI: 10.1074/jbc.m112.361311] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Stac3 was identified as a nutritionally regulated gene from an Atlantic salmon subtractive hybridization library with highest expression in skeletal muscle. Salmon Stac3 mRNA was highly correlated with myogenin and myoD1a expression during differentiation of a salmon primary myogenic culture and was regulated by amino acid availability. In zebrafish embryos, stac3 was initially expressed in myotomal adaxial cells and in fast muscle fibers post-segmentation. Morpholino knockdown resulted in defects in myofibrillar protein assembly, particularly in slow muscle fibers, and decreased levels of the hedgehog receptor patched. The function of Stac3 was further characterized in vitro using the mammalian C2C12 myogenic cell line. Stac3 mRNA expression increased during the differentiation of the C2C12 myogenic cell line. Knockdown of Stac3 by RNAi inhibited myotube formation, and microarray analysis revealed that transcripts involved in cell cycle, focal adhesion, cytoskeleton, and the pro-myogenic factors Igfbp-5 and Igf2 were down-regulated. RNAi-treated cells had suppressed Akt signaling and exogenous insulin-like growth factor (Igf) 2 was unable to rescue the phenotype, however, Igf/Akt signaling was not blocked. Overexpression of Stac3, which results in increased levels of Igfbp-5 mRNA, did not lead to increased differentiation. In synchronized cells, Stac3 mRNA was most abundant during the G(1) phase of the cell cycle. RNAi-treated cells were smaller, had higher proliferation rates and a decreased proportion of cells in G(1) phase when compared with controls, suggesting a role in the G(1) phase checkpoint. These results identify Stac3 as a new gene required for myogenic differentiation and myofibrillar protein assembly in vertebrates.
Collapse
Affiliation(s)
- Neil I Bower
- Scottish Oceans Institute, School of Biology, University of St. Andrews, St. Andrews KY16 8LB, United Kingdom
| | | | | | | | | | | | | |
Collapse
|
35
|
Windner SE, Bird NC, Patterson SE, Doris RA, Devoto SH. Fss/Tbx6 is required for central dermomyotome cell fate in zebrafish. Biol Open 2012; 1:806-14. [PMID: 23213474 PMCID: PMC3507223 DOI: 10.1242/bio.20121958] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2012] [Accepted: 05/29/2012] [Indexed: 12/18/2022] Open
Abstract
The dermomyotome is a pool of progenitor cells on the surface of the myotome. In zebrafish, dermomyotome precursors (anterior border cells, ABCs) can be first identified in the anterior portion of recently formed somites. They must be prevented from undergoing terminal differentiation during segmentation, even while mesodermal cells around them respond to signaling cues and differentiate. T-box containing transcription factors regulate many aspects of mesoderm fate including segmentation and somite patterning. The fused somites (fss) gene is the zebrafish ortholog of tbx6. We demonstrate that in addition to its requirement for segmentation, fss/tbx6 is also required for the specification of ABCs and subsequently the central dermomyotome. The absence of Tbx6-dependent central dermomyotome cells in fss/tbx6 mutants is spatially coincident with a patterning defect in the myotome. Using transgenic fish with a heat-shock inducible tbx6 gene in the fss/tbx6 mutant background, we further demonstrate that ubiquitous fss/tbx6 expression has spatially distinct effects on recovery of the dermomyotome and segment boundaries, suggesting that the mechanism of Fss/Tbx6 action is distinct with respect to dermomyotome development and segmentation. We propose that Fss/Tbx6 is required for preventing myogenic differentiation of central dermomyotome precursors before and after segmentation and that central dermomyotome cells represent a genetically and functionally distinct subpopulation within the zebrafish dermomyotome.
Collapse
Affiliation(s)
- Stefanie Elisabeth Windner
- Department of Biology, Wesleyan University , Middletown, CT 06459 , USA ; Division of Zoology and Functional Anatomy, Department of Organismic Biology, University of Salzburg , A-5020 Salzburg, Austria
| | | | | | | | | |
Collapse
|
36
|
Webb SE, Cheung CCY, Chan CM, Love DR, Miller AL. Application of complementary luminescent and fluorescent imaging techniques to visualize nuclear and cytoplasmic Ca²⁺ signalling during the in vivo differentiation of slow muscle cells in zebrafish embryos under normal and dystrophic conditions. Clin Exp Pharmacol Physiol 2012; 39:78-86. [PMID: 21824171 DOI: 10.1111/j.1440-1681.2011.05582.x] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
1. Evidence is accumulating for a role for Ca²⁺ signalling in the differentiation and development of embryonic skeletal muscle. 2. Imaging of intact, normally developing transgenic zebrafish that express the protein component of the Ca²⁺-sensitive complex aequorin, specifically in skeletal muscle, show that two distinct periods of spontaneous synchronised Ca²⁺ transients occur in the trunk: one at approximately 17.5-19.5 h post-fertilization (h.p.f.; termed signalling period SP1) and the other after approximately 23 h.p.f. (termed SP2). These periods of intense Ca²⁺ signalling activity are separated by a quiet period. 3. Higher-resolution confocal imaging of embryos loaded with the fluorescent Ca²⁺ reporter calcium green-1 dextran shows that the Ca²⁺ signals are generated almost exclusively in the slow muscle cells, the first muscle cells to differentiate, with distinct nuclear and cytoplasmic components. 4. Here, we show that coincidental with the SP1 Ca²⁺ signals, dystrophin becomes localized to the vertical myoseptae of the myotome. Introduction of a dmd morpholino (dmd-MO) resulted in no dystrophin being expressed in the vertical myoseptae, as well as a disruption of myotome morphology and sarcomere organization. In addition, the Ca²⁺ signalling signatures of dmd-MO-injected embryos or homozygous sapje mutant embryos were abnormal such that the frequency, amplitude and timing of the Ca²⁺ signals were altered compared with controls. 5. Our new data suggest that, in addition to a structural role, dystrophin may function in the regulation of [Ca²⁺](i) during the early stages of slow muscle cell differentiation when the Ca²⁺ signals generated in these cells coincide with the first spontaneous contractions of the trunk.
Collapse
Affiliation(s)
- Sarah E Webb
- Division of Life Science and Key State Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong SAR, China
| | | | | | | | | |
Collapse
|
37
|
Transcriptional responses of zebrafish embryos exposed to potential sonic hedgehog pathway interfering compounds deviate from expression profiles of cyclopamine. Reprod Toxicol 2012; 33:254-63. [DOI: 10.1016/j.reprotox.2011.12.010] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2011] [Revised: 12/13/2011] [Accepted: 12/16/2011] [Indexed: 01/08/2023]
|
38
|
Ccdc80-l1 Is involved in axon pathfinding of zebrafish motoneurons. PLoS One 2012; 7:e31851. [PMID: 22384085 PMCID: PMC3285184 DOI: 10.1371/journal.pone.0031851] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2011] [Accepted: 01/19/2012] [Indexed: 11/29/2022] Open
Abstract
Axon pathfinding is a subfield of neural development by which neurons send out axons to reach the correct targets. In particular, motoneurons extend their axons toward skeletal muscles, leading to spontaneous motor activity. In this study, we identified the zebrafish Ccdc80 and Ccdc80-like1 (Ccdc80-l1) proteins in silico on the basis of their high aminoacidic sequence identity with the human CCDC80 (Coiled-Coil Domain Containing 80). We focused on ccdc80-l1 gene that is expressed in nervous and non-nervous tissues, in particular in territories correlated with axonal migration, such as adaxial cells and muscle pioneers. Loss of ccdc80-l1 in zebrafish embryos induced motility issues, although somitogenesis and myogenesis were not impaired. Our results strongly suggest that ccdc80-l1 is involved in axon guidance of primary and secondary motoneurons populations, but not in their proper formation. ccdc80-l1 has a differential role as regards the development of ventral and dorsal motoneurons, and this is consistent with the asymmetric distribution of the transcript. The axonal migration defects observed in ccdc80-l1 loss-of-function embryos are similar to the phenotype of several mutants with altered Hedgehog activity. Indeed, we reported that ccdc80-l1 expression is positively regulated by the Hedgehog pathway in adaxial cells and muscle pioneers. These findings strongly indicate ccdc80-l1 as a down-stream effector of the Hedgehog pathway.
Collapse
|
39
|
Rescan PY, Ralliere C, Lebret V. N-cadherin and M-cadherin are sequentially expressed in myoblast populations contributing to the first and second waves of myogenesis in the trout (Oncorhynchus mykiss). JOURNAL OF EXPERIMENTAL ZOOLOGY PART B-MOLECULAR AND DEVELOPMENTAL EVOLUTION 2012; 318:71-7. [PMID: 22057948 DOI: 10.1002/jez.b.21443] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/28/2011] [Revised: 08/29/2011] [Accepted: 08/30/2011] [Indexed: 11/10/2022]
Abstract
The objective of this study was to investigate the expression of two promyogenic cell surface adhesion receptors, N- and M-cadherin, in developing trout (Oncorhynchus mykiss) somite, taking account of the recent identification of a dermomyotome-like epithelium in teleosts. In situ hybridization showed that N-cadherin was expressed throughout the paraxial mesoderm and nascent somite. As the somite matured, N-cadherin expression disappeared ventrally from the sclerotome, and then mediolaterally from the differentiating slow and fast muscle cells of the embryonic myotome, to become finally restricted to the undifferentiated myogenic precursors forming the dermomyotome-like epithelium that surrounds the embryonic myotome. By contrast, M-cadherin, which was transcribed in the differentiating embryonic myotome, was never expressed in the dermomyotome-like epithelium. In late-stage trout embryos, M-cadherin transcript was only detected at the periphery of the expanding myotome, where muscle cells stemming from the N-cadherin positive dermomyotome-like epithelium differentiate. Collectively, our results support the view that, in trout embryo, N-cadherin is associated with muscle cell immaturity while M-cadherin is associated with muscle cell maturation and differentiation and this during the two successive phases of myogenesis.
Collapse
|
40
|
Bird NC, Windner SE, Devoto SH. Immunocytochemistry to study myogenesis in zebrafish. Methods Mol Biol 2012; 798:153-69. [PMID: 22130836 DOI: 10.1007/978-1-61779-343-1_9] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
During myogenesis, cells gradually transition from mesodermal precursors to myoblasts, myocytes, and then to muscle fibers. The molecular characterization of this process requires the ability to identify each of these cell types and the factors that regulate the transitions between them. The most versatile technique for assaying cell identities in situ is immunocytochemistry, because multiple independent molecular markers of differentiation can be assayed simultaneously. The zebrafish has developed into a popular model for the study of myogenesis, and immunocytochemical techniques have been critical. We have adapted existing protocols to optimize immunocytochemistry in zebrafish, and have tested many antibodies developed against mouse, chick, and frog muscle antigens for their cross-reactivity in zebrafish. Here, we present protocols for whole mount immunocytochemistry on both formaldehyde and Carnoy's fixed embryos as well as on sectioned zebrafish tissue. We include a table of antibodies useful for experiments on the molecular biology of myogenesis in zebrafish.
Collapse
Affiliation(s)
- Nathan C Bird
- Department of Biology, Wesleyan University, Hall-Atwater Laboratories, Middletown, CT, USA
| | | | | |
Collapse
|
41
|
Kettunen P. Calcium imaging in the zebrafish. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2012; 740:1039-71. [PMID: 22453983 DOI: 10.1007/978-94-007-2888-2_48] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
The zebrafish (Danio rerio) has emerged as a new model system during the last three decades. The fact that the zebrafish larva is transparent enables sophisticated in vivo imaging. While being the vertebrate, the reduced complexity of its nervous system and small size make it possible to follow large-scale activity in the whole brain. Its genome is sequenced and many genetic and molecular tools have been developed that simplify the study of gene function. Since the mid 1990s, the embryonic development and neuronal function of the larval, and later, adult zebrafish have been studied using calcium imaging methods. The choice of calcium indicator depends on the desired number of cells to study and cell accessibility. Dextran indicators have been used to label cells in the developing embryo from dye injection into the one-cell stage. Dextrans have also been useful for retrograde labeling of spinal cord neurons and cells in the olfactory system. Acetoxymethyl (AM) esters permit labeling of larger areas of tissue such as the tectum, a region responsible for visual processing. Genetically encoded calcium indicators have been expressed in various tissues by the use of cell-specific promoters. These studies have contributed greatly to our understanding of basic biological principles during development and adulthood, and of the function of disease-related genes in a vertebrate system.
Collapse
Affiliation(s)
- Petronella Kettunen
- Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, Sweden.
| |
Collapse
|
42
|
Steinbacher P, Marschallinger J, Obermayer A, Neuhofer A, Sänger AM, Stoiber W. Temperature-dependent modification of muscle precursor cell behaviour is an underlying reason for lasting effects on muscle cellularity and body growth of teleost fish. ACTA ACUST UNITED AC 2011; 214:1791-801. [PMID: 21562165 DOI: 10.1242/jeb.050096] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Temperature is an important factor influencing teleost muscle growth, including a lasting ('imprinted') influence of embryonic thermal experience throughout all further life. However, little is known about the cellular processes behind this phenomenon. The study reported here used digital morphometry and immunolabelling for Pax7, myogenin and H3P to quantitatively examine the effects of thermal history on muscle precursor cell (MPC) behaviour and muscle growth in pearlfish (Rutilus meidingeri) until the adult stage. Fish were reared at three different temperatures (8.5, 13 and 16°C) until hatching and subsequently kept under the same (ambient) thermal conditions. Cellularity data were combined with a quantitative analysis of Pax7+ MPCs including those that were mitotically active (Pax7+/H3P+) or had entered differentiation (Pax7+/myogenin+). The results demonstrate that at hatching, body lengths, fast and slow muscle cross-sectional areas and fast fibre numbers are lower in fish reared at 8.5 and 13°C than at 16°C. During the larval period, this situation changes in the 13°C-fish, so that these fish are finally the largest. The observed effects can be related to divergent cellular mechanisms at the MPC level that are initiated in the embryo during the imprinting period. Embryos of 16°C-fish have reduced MPC proliferation but increased differentiation, and thus give rise to larger hatchlings. However, their limited MPC reserves finally lead to smaller adults. By contrast, embryos of 13°C-fish and, to a lesser extent, 8.5°-fish, show enhanced MPC proliferation but reduced differentiation, thus leading to smaller hatchlings but allowing for a larger MPC pool that can be used for enhanced post-hatching growth, finally resulting in larger adults.
Collapse
Affiliation(s)
- Peter Steinbacher
- Division of Zoology and Functional Anatomy, Department of Organismic Biology, University of Salzburg, Salzburg, Austria.
| | | | | | | | | | | |
Collapse
|
43
|
Johnston IA, Bower NI, Macqueen DJ. Growth and the regulation of myotomal muscle mass in teleost fish. ACTA ACUST UNITED AC 2011; 214:1617-28. [PMID: 21525308 DOI: 10.1242/jeb.038620] [Citation(s) in RCA: 260] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Teleost muscle first arises in early embryonic life and its development is driven by molecules present in the egg yolk and modulated by environmental stimuli including temperature and oxygen. Several populations of myogenic precursor cells reside in the embryonic somite and external cell layer and contribute to muscle fibres in embryo, larval, juvenile and adult stages. Many signalling proteins and transcription factors essential for these events are known. In all cases, myogenesis involves myoblast proliferation, migration, fusion and terminal differentiation. Maturation of the embryonic muscle is associated with motor innervation and the development of a scaffold of connective tissue and complex myotomal architecture needed to generate swimming behaviour. Adult muscle is a heterogeneous tissue composed of several cell types that interact to affect growth patterns. The development of capillary and lymphatic circulations and extramuscular organs--notably the gastrointestinal, endocrine, neuroendocrine and immune systems--serves to increase information exchange between tissues and with the external environment, adding to the complexity of growth regulation. Teleosts often exhibit an indeterminate growth pattern, with body size and muscle mass increasing until mortality or senescence occurs. The dramatic increase in myotomal muscle mass between embryo and adult requires the continuous production of muscle fibres until 40-50% of the maximum body length is reached. Sarcomeric proteins can be mobilised as a source of amino acids for energy metabolism by other tissues and for gonad generation, requiring the dynamic regulation of muscle mass throughout the life cycle. The metabolic and contractile phenotypes of muscle fibres also show significant plasticity with respect to environmental conditions, migration and spawning. Many genes regulating muscle growth are found as multiple copies as a result of paralogue retention following whole-genome duplication events in teleost lineages. The extent to which indeterminate growth, ectothermy and paralogue preservation have resulted in modifications of the genetic pathways regulating muscle growth in teleosts compared to mammals largely remains unknown. This review describes the use of compensatory growth models, transgenesis and tissue culture to explore the mechanisms of muscle growth in teleosts and provides some perspectives on future research directions.
Collapse
Affiliation(s)
- Ian A Johnston
- Scottish Oceans Institute, School of Biology, University of St Andrews, St Andrews, Fife KY168LB, UK.
| | | | | |
Collapse
|
44
|
Windner SE, Steinbacher P, Obermayer A, Kasiba B, Zweimueller-Mayer J, Stoiber W. Distinct modes of vertebrate hypaxial muscle formation contribute to the teleost body wall musculature. Dev Genes Evol 2011; 221:167-78. [PMID: 21720828 DOI: 10.1007/s00427-011-0369-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2011] [Accepted: 06/14/2011] [Indexed: 11/28/2022]
Abstract
The formation of the body wall musculature in vertebrates is assumed to be initiated by direct ventral extension of the somites/myotomes. This contrasts to the formation of limb muscles and muscles involved in feeding or respiration/ventilation, which are founded by migratory muscle precursors (MMPs) distant to the somites. Here, we present evidence from morphology and expression of molecular markers proposing that the formation of the two muscle layers of the teleost body wall involves both of the above mechanisms: (1) MMPs from somites 5 and 6 found an independent muscle primordium-the so-called posterior hypaxial muscle (PHM)-which subsequently gives rise to the most anterior two segments of the medial obliquus inferioris (OI) muscle. (2) Direct epithelial extension of the hypaxial myotomes generates the OI segments from somite 7 caudalward and the entire lateral obliquus superioris (OS) muscle. The findings are discussed in relation to the evolution of hypaxial myogenic patterning including functional considerations. We hypothesise that the potential of the most anterior somites to generate migratory muscle precursors is a general vertebrate feature that has been differently utilised in the evolution in vertebrate groups.
Collapse
Affiliation(s)
- Stefanie E Windner
- Division of Zoology, Department of Organismic Biology, University of Salzburg, Austria
| | | | | | | | | | | |
Collapse
|
45
|
Webb SE, Miller AL. Visualization of Ca²+ signaling during embryonic skeletal muscle formation in vertebrates. Cold Spring Harb Perspect Biol 2011; 3:cshperspect.a004325. [PMID: 21421918 DOI: 10.1101/cshperspect.a004325] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Dynamic changes in cytosolic and nuclear Ca(2+) concentration are reported to play a critical regulatory role in different aspects of skeletal muscle development and differentiation. Here we review our current knowledge of the spatial dynamics of Ca(2+) signals generated during muscle development in mouse, rat, and Xenopus myocytes in culture, in the exposed myotome of dissected Xenopus embryos, and in intact normally developing zebrafish. It is becoming clear that subcellular domains, either membrane-bound or otherwise, may have their own Ca(2+) signaling signatures. Thus, to understand the roles played by myogenic Ca(2+) signaling, we must consider: (1) the triggers and targets within these signaling domains; (2) interdomain signaling, and (3) how these Ca(2+) signals integrate with other signaling networks involved in myogenesis. Imaging techniques that are currently available to provide direct visualization of these Ca(2+) signals are also described.
Collapse
Affiliation(s)
- Sarah E Webb
- Section of Biochemistry and Cell Biology, and State Key Laboratory of Molecular Neuroscience, Division of Life Science, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong SAR, PRC
| | | |
Collapse
|
46
|
Expression and interaction of muscle-related genes in the lamprey imply the evolutionary scenario for vertebrate skeletal muscle, in association with the acquisition of the neck and fins. Dev Biol 2011; 350:217-27. [DOI: 10.1016/j.ydbio.2010.10.029] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2010] [Revised: 10/19/2010] [Accepted: 10/20/2010] [Indexed: 01/04/2023]
|
47
|
Osborn DPS, Li K, Hinits Y, Hughes SM. Cdkn1c drives muscle differentiation through a positive feedback loop with Myod. Dev Biol 2010; 350:464-75. [PMID: 21147088 DOI: 10.1016/j.ydbio.2010.12.010] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2010] [Revised: 12/01/2010] [Accepted: 12/03/2010] [Indexed: 01/15/2023]
Abstract
Differentiation often requires conversion of analogue signals to a stable binary output through positive feedback. Hedgehog (Hh) signalling promotes myogenesis in the vertebrate somite, in part by raising the activity of muscle regulatory factors (MRFs) of the Myod family above a threshold. Hh is known to enhance MRF expression. Here we show that Hh is also essential at a second step that increases Myod protein activity, permitting it to promote Myogenin expression. Hh acts by inducing expression of cdkn1c (p57(Kip2)) in slow muscle precursor cells, but neither Hh nor Cdkn1c is required for their cell cycle exit. Cdkn1c co-operates with Myod to drive differentiation of several early zebrafish muscle fibre types. Myod in turn up-regulates cdkn1c, thereby providing a positive feedback loop that switches myogenic cells to terminal differentiation.
Collapse
Affiliation(s)
- Daniel P S Osborn
- King's College London, Randall Division for Cell and Molecular Biophysics, London, UK
| | | | | | | |
Collapse
|
48
|
Peterson MT, Henry CA. Hedgehog signaling and laminin play unique and synergistic roles in muscle development. Dev Dyn 2010; 239:905-13. [PMID: 20063418 DOI: 10.1002/dvdy.22204] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
Hedgehog (Hh) signaling and laminin-111, a basement membrane protein, are required for early muscle development. Hh signaling specifies different populations of muscle fibers and laminin-111 is critical for early muscle morphogenesis. However, additional requirements for Hh signaling and laminin during later phases of muscle development are not known. Furthermore, interactions between Hh signaling and laminin in this context are unknown. We used laminin gamma1 mutant zebrafish and cyclopamine to block Hh signal transduction separately and in combination to investigate their functions and interactions. We found that both Hh signaling and laminin are required for normal myosin chain expression. In addition, Hh signaling and laminin act synergistically during fast-twitch fiber elongation: fast muscle cells do not elongate in embryos deficient for both Hh signaling and laminin. Finally, we present evidence that suggests that Hh signaling is indirectly required via slow fiber specification for recovery of fast fiber elongation in laminin gamma1 mutant embryos.
Collapse
Affiliation(s)
- Matthew T Peterson
- School of Biology and Ecology, University of Maine, Orono, Maine 04469, USA
| | | |
Collapse
|
49
|
Abstract
In amniotes, BMP signaling from lateral plate and dorsal neural tube inhibits differentiation of muscle precursors in the dermomyotome. Here, we show that BMPs are expressed adjacent to the dermomyotome during and after segmentation in zebrafish. In addition, downstream BMP pathway members are expressed within the somite during dermomyotome development. We also show that zebrafish dermomyotome is responsive to BMP throughout its development. Ectopic overexpression of Bmp2b increases expression of the muscle precursor marker pax3, and changes the time course of myoD expression. At later stages, overexpression increases the number of Pax7+ myogenic precursors, and delays muscle differentiation, as indicated by decreased numbers of MEF2+ nuclei, decreased number of multi-nucleated muscle fibers, and an increased myotome angle. In addition, we show that while BMP overexpression is sufficient to delay myogenic differentiation, inhibition of BMP does not detectably affect this process, suggesting that other factors redundantly inhibit myogenic differentiation.
Collapse
Affiliation(s)
- Sara E Patterson
- Department of Biology, Wesleyan University, Middletown, Connecticut 06459, USA
| | | | | |
Collapse
|
50
|
Lee CY, Vogeli KM, Kim SH, Chong SW, Jiang YJ, Stainier DYR, Jin SW. Notch signaling functions as a cell-fate switch between the endothelial and hematopoietic lineages. Curr Biol 2009; 19:1616-22. [PMID: 19747827 DOI: 10.1016/j.cub.2009.07.073] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2009] [Revised: 07/21/2009] [Accepted: 07/23/2009] [Indexed: 11/28/2022]
Abstract
Recent studies have begun to elucidate how the endothelial lineage is specified from the nascent mesoderm. However, the molecular mechanisms which regulate this process remain largely unknown. We hypothesized that Notch signaling might play an important role in specifying endothelial progenitors from the mesoderm, given that this pathway acts as a bipotential cell-fate switch on equipotent progenitor populations in other settings. We found that zebrafish embryos with decreased levels of Notch signaling exhibited a significant increase in the number of endothelial cells, whereas embryos with increased levels of Notch signaling displayed a reduced number of endothelial cells. Interestingly, there is a concomitant gain of endothelial cells and loss of erythrocytes in embryos with decreased Notch activity, without an effect on cell proliferation or apoptosis. Lineage-tracing analyses indicate that the ectopic endothelial cells in embryos with decreased Notch activity originate from mesodermal cells that normally produce erythrocyte progenitors. Taken together, our data suggest that Notch signaling negatively regulates the number of endothelial cells by limiting the number of endothelial progenitors within the mesoderm, probably functioning as a cell-fate switch between the endothelial and the hematopoietic lineages.
Collapse
Affiliation(s)
- Christina Y Lee
- McAllister Heart Institute, Department of Cell and Molecular Physiology, and Curriculum in Genetics and Molecular Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | | | | | | | | | | | | |
Collapse
|