1
|
Xue X, Wu X, Fan Y, Han S, Zhang H, Sun Y, Yin Y, Yin M, Chen B, Sun Z, Zhao S, Zhang Q, Liu W, Zhang J, Li J, Shi Y, Xiao Z, Dai J, Zhao Y. Heterogeneous fibroblasts contribute to fibrotic scar formation after spinal cord injury in mice and monkeys. Nat Commun 2024; 15:6321. [PMID: 39060269 PMCID: PMC11282111 DOI: 10.1038/s41467-024-50564-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2024] [Accepted: 06/24/2024] [Indexed: 07/28/2024] Open
Abstract
Spinal cord injury (SCI) leads to fibrotic scar formation at the lesion site, yet the heterogeneity of fibrotic scar remains elusive. Here we show the heterogeneity in distribution, origin, and function of fibroblasts within fibrotic scars after SCI in mice and female monkeys. Utilizing lineage tracing and single-cell RNA sequencing (scRNA-seq), we found that perivascular fibroblasts (PFs), and meningeal fibroblasts (MFs), rather than pericytes/vascular smooth cells (vSMCs), primarily contribute to fibrotic scar in both transection and crush SCI. Crabp2 + /Emb+ fibroblasts (CE-F) derived from meninges primarily localize in the central region of fibrotic scars, demonstrating enhanced cholesterol synthesis and secretion of type I collagen and fibronectin. In contrast, perivascular/pial Lama1 + /Lama2+ fibroblasts (LA-F) are predominantly found at the periphery of the lesion, expressing laminin and type IV collagen and functionally involved in angiogenesis and lipid transport. These findings may provide a comprehensive understanding for remodeling heterogeneous fibrotic scars after SCI.
Collapse
Affiliation(s)
- Xiaoyu Xue
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Xianming Wu
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Yongheng Fan
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100101, China
| | - Shuyu Han
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100101, China
| | - Haipeng Zhang
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100101, China
| | - Yuting Sun
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100101, China
| | - Yanyun Yin
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Man Yin
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100101, China
| | - Bing Chen
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Zheng Sun
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100101, China
| | - Shuaijing Zhao
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100101, China
| | - Qi Zhang
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Weiyuan Liu
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100101, China
| | - Jiaojiao Zhang
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Jiayin Li
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Ya Shi
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Zhifeng Xiao
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China.
| | - Jianwu Dai
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China.
- University of Chinese Academy of Sciences, Beijing, 100101, China.
- Tianjin Key Laboratory of Biomedical Materials, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300192, China.
| | - Yannan Zhao
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China.
| |
Collapse
|
2
|
Nemoz-Billet L, Balland M, Gilquin L, Gillet B, Stévant I, Guillon E, Hughes S, Carpentier G, Vaganay E, Sohm F, Misiak V, Gonzalez-Melo MJ, Koch M, Ghavi-Helm Y, Bretaud S, Ruggiero F. Dual topologies of myotomal collagen XV and Tenascin C act in concert to guide and shape developing motor axons. Proc Natl Acad Sci U S A 2024; 121:e2314588121. [PMID: 38502691 PMCID: PMC10990108 DOI: 10.1073/pnas.2314588121] [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: 08/23/2023] [Accepted: 02/26/2024] [Indexed: 03/21/2024] Open
Abstract
During development, motor axons are guided toward muscle target by various extrinsic cues including extracellular matrix (ECM) proteins whose identities and cellular source remain poorly characterized. Here, using single-cell RNAseq of sorted GFP+ cells from smyhc1:gfp-injected zebrafish embryos, we unravel the slow muscle progenitors (SMP) pseudotemporal trajectory at the single-cell level and show that differentiating SMPs are a major source of ECM proteins. The SMP core-matrisome was characterized and computationally predicted to form a basement membrane-like structure tailored for motor axon guidance, including basement membrane-associated ECM proteins, as collagen XV-B, one of the earliest core-matrisome gene transcribed in differentiating SMPs and the glycoprotein Tenascin C. To investigate how contact-mediated guidance cues are organized along the motor path to exert their function in vivo, we used microscopy-based methods to analyze and quantify motor axon navigation in tnc and col15a1b knock-out fish. We show that motor axon shape and growth rely on the timely expression of the attractive cue Collagen XV-B that locally provides axons with a permissive soft microenvironment and separately organizes the repulsive cue Tenascin C into a unique functional dual topology. Importantly, bioprinted micropatterns that mimic this in vivo ECM topology were sufficient to drive directional motor axon growth. Our study offers evidence that not only the composition of ECM cues but their topology critically influences motor axon navigation in vertebrates with potential applications in regenerative medicine for peripheral nerve injury as regenerating nerves follow their original path.
Collapse
Affiliation(s)
- Laurie Nemoz-Billet
- Institut de Génomique Fonctionnelle de Lyon, Ecole Normale Supérieure de Lyon, UMR5242 CNRS, Université Claude Bernard-Lyon1, National Research Institute for Agriculture, Food and the Environment (INRAe) Unit under Contract (USC) 1370, Lyon69007, France
| | - Martial Balland
- LIPphy: Interdisciplinary Laboratory of Physics, Université Grenoble Alpes, CNRS, GrenobleF-38000, France
| | - Laurent Gilquin
- Institut de Génomique Fonctionnelle de Lyon, Ecole Normale Supérieure de Lyon, UMR5242 CNRS, Université Claude Bernard-Lyon1, National Research Institute for Agriculture, Food and the Environment (INRAe) Unit under Contract (USC) 1370, Lyon69007, France
| | - Benjamin Gillet
- Institut de Génomique Fonctionnelle de Lyon, Ecole Normale Supérieure de Lyon, UMR5242 CNRS, Université Claude Bernard-Lyon1, National Research Institute for Agriculture, Food and the Environment (INRAe) Unit under Contract (USC) 1370, Lyon69007, France
| | - Isabelle Stévant
- Institut de Génomique Fonctionnelle de Lyon, Ecole Normale Supérieure de Lyon, UMR5242 CNRS, Université Claude Bernard-Lyon1, National Research Institute for Agriculture, Food and the Environment (INRAe) Unit under Contract (USC) 1370, Lyon69007, France
| | - Emilie Guillon
- Institut de Génomique Fonctionnelle de Lyon, Ecole Normale Supérieure de Lyon, UMR5242 CNRS, Université Claude Bernard-Lyon1, National Research Institute for Agriculture, Food and the Environment (INRAe) Unit under Contract (USC) 1370, Lyon69007, France
| | - Sandrine Hughes
- Institut de Génomique Fonctionnelle de Lyon, Ecole Normale Supérieure de Lyon, UMR5242 CNRS, Université Claude Bernard-Lyon1, National Research Institute for Agriculture, Food and the Environment (INRAe) Unit under Contract (USC) 1370, Lyon69007, France
| | - Gilles Carpentier
- Gly-CRRET: Glycobiology, Cell Growth and Tissue Repair Research Unit, Laboratoire Gly-CRRET Faculté des Sciences et Technologie, Université Paris Est-Créteil-Val de Marne, Créteil Cedex94010, France
| | - Elisabeth Vaganay
- Institut de Génomique Fonctionnelle de Lyon, Ecole Normale Supérieure de Lyon, UMR5242 CNRS, Université Claude Bernard-Lyon1, National Research Institute for Agriculture, Food and the Environment (INRAe) Unit under Contract (USC) 1370, Lyon69007, France
| | - Frédéric Sohm
- Institut de Génomique Fonctionnelle de Lyon, Ecole Normale Supérieure de Lyon, UMR5242 CNRS, Université Claude Bernard-Lyon1, National Research Institute for Agriculture, Food and the Environment (INRAe) Unit under Contract (USC) 1370, Lyon69007, France
| | - Vladimir Misiak
- LIPphy: Interdisciplinary Laboratory of Physics, Université Grenoble Alpes, CNRS, GrenobleF-38000, France
| | - Mary-Julieth Gonzalez-Melo
- Institut de Génomique Fonctionnelle de Lyon, Ecole Normale Supérieure de Lyon, UMR5242 CNRS, Université Claude Bernard-Lyon1, National Research Institute for Agriculture, Food and the Environment (INRAe) Unit under Contract (USC) 1370, Lyon69007, France
| | - Manuel Koch
- Institute for Experimental Dental Research and Oral Musculoskeletal Biology, Center for Biochemistry, Medical Faculty, University of Cologne, Cologne50931, Germany
| | - Yad Ghavi-Helm
- Institut de Génomique Fonctionnelle de Lyon, Ecole Normale Supérieure de Lyon, UMR5242 CNRS, Université Claude Bernard-Lyon1, National Research Institute for Agriculture, Food and the Environment (INRAe) Unit under Contract (USC) 1370, Lyon69007, France
| | - Sandrine Bretaud
- Institut de Génomique Fonctionnelle de Lyon, Ecole Normale Supérieure de Lyon, UMR5242 CNRS, Université Claude Bernard-Lyon1, National Research Institute for Agriculture, Food and the Environment (INRAe) Unit under Contract (USC) 1370, Lyon69007, France
| | - Florence Ruggiero
- Institut de Génomique Fonctionnelle de Lyon, Ecole Normale Supérieure de Lyon, UMR5242 CNRS, Université Claude Bernard-Lyon1, National Research Institute for Agriculture, Food and the Environment (INRAe) Unit under Contract (USC) 1370, Lyon69007, France
| |
Collapse
|
3
|
Kolb J, Tsata V, John N, Kim K, Möckel C, Rosso G, Kurbel V, Parmar A, Sharma G, Karandasheva K, Abuhattum S, Lyraki O, Beck T, Müller P, Schlüßler R, Frischknecht R, Wehner A, Krombholz N, Steigenberger B, Beis D, Takeoka A, Blümcke I, Möllmert S, Singh K, Guck J, Kobow K, Wehner D. Small leucine-rich proteoglycans inhibit CNS regeneration by modifying the structural and mechanical properties of the lesion environment. Nat Commun 2023; 14:6814. [PMID: 37884489 PMCID: PMC10603094 DOI: 10.1038/s41467-023-42339-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Accepted: 10/04/2023] [Indexed: 10/28/2023] Open
Abstract
Extracellular matrix (ECM) deposition after central nervous system (CNS) injury leads to inhibitory scarring in humans and other mammals, whereas it facilitates axon regeneration in the zebrafish. However, the molecular basis of these different fates is not understood. Here, we identify small leucine-rich proteoglycans (SLRPs) as a contributing factor to regeneration failure in mammals. We demonstrate that the SLRPs chondroadherin, fibromodulin, lumican, and prolargin are enriched in rodent and human but not zebrafish CNS lesions. Targeting SLRPs to the zebrafish injury ECM inhibits axon regeneration and functional recovery. Mechanistically, we find that SLRPs confer mechano-structural properties to the lesion environment that are adverse to axon growth. Our study reveals SLRPs as inhibitory ECM factors that impair axon regeneration by modifying tissue mechanics and structure, and identifies their enrichment as a feature of human brain and spinal cord lesions. These findings imply that SLRPs may be targets for therapeutic strategies to promote CNS regeneration.
Collapse
Affiliation(s)
- Julia Kolb
- Max Planck Institute for the Science of Light, 91058, Erlangen, Germany
- Max-Planck-Zentrum für Physik und Medizin, 91058, Erlangen, Germany
- Department of Biology, Animal Physiology, Friedrich-Alexander-University Erlangen-Nürnberg, 91058, Erlangen, Germany
| | - Vasiliki Tsata
- Experimental Surgery, Clinical and Translational Research Center, Biomedical Research Foundation Academy of Athens, 11527, Athens, Greece
- Center of Basic Research, Biomedical Research Foundation, Academy of Athens, 11527, Athens, Greece
| | - Nora John
- Max Planck Institute for the Science of Light, 91058, Erlangen, Germany
- Max-Planck-Zentrum für Physik und Medizin, 91058, Erlangen, Germany
- Department of Biology, Animal Physiology, Friedrich-Alexander-University Erlangen-Nürnberg, 91058, Erlangen, Germany
| | - Kyoohyun Kim
- Max Planck Institute for the Science of Light, 91058, Erlangen, Germany
- Max-Planck-Zentrum für Physik und Medizin, 91058, Erlangen, Germany
| | - Conrad Möckel
- Max Planck Institute for the Science of Light, 91058, Erlangen, Germany
- Max-Planck-Zentrum für Physik und Medizin, 91058, Erlangen, Germany
- Department of Physics, Friedrich-Alexander-University Erlangen-Nürnberg, 91058, Erlangen, Germany
| | - Gonzalo Rosso
- Max Planck Institute for the Science of Light, 91058, Erlangen, Germany
- Max-Planck-Zentrum für Physik und Medizin, 91058, Erlangen, Germany
| | - Veronika Kurbel
- Department of Neuropathology, Universitätsklinikum Erlangen, Friedrich-Alexander-University Erlangen-Nürnberg, 91054, Erlangen, Germany
| | - Asha Parmar
- Max Planck Institute for the Science of Light, 91058, Erlangen, Germany
- Max-Planck-Zentrum für Physik und Medizin, 91058, Erlangen, Germany
- Department of Physics, Friedrich-Alexander-University Erlangen-Nürnberg, 91058, Erlangen, Germany
| | - Gargi Sharma
- Max Planck Institute for the Science of Light, 91058, Erlangen, Germany
- Department of Medicine 1, Universitätsklinikum Erlangen, Friedrich-Alexander-University Erlangen-Nürnberg, 91054, Erlangen, Germany
| | - Kristina Karandasheva
- Department of Neuropathology, Universitätsklinikum Erlangen, Friedrich-Alexander-University Erlangen-Nürnberg, 91054, Erlangen, Germany
| | - Shada Abuhattum
- Max Planck Institute for the Science of Light, 91058, Erlangen, Germany
- Max-Planck-Zentrum für Physik und Medizin, 91058, Erlangen, Germany
| | - Olga Lyraki
- Max Planck Institute for the Science of Light, 91058, Erlangen, Germany
- Max-Planck-Zentrum für Physik und Medizin, 91058, Erlangen, Germany
- Department of Biology, Animal Physiology, Friedrich-Alexander-University Erlangen-Nürnberg, 91058, Erlangen, Germany
| | - Timon Beck
- Max Planck Institute for the Science of Light, 91058, Erlangen, Germany
- Max-Planck-Zentrum für Physik und Medizin, 91058, Erlangen, Germany
| | - Paul Müller
- Max Planck Institute for the Science of Light, 91058, Erlangen, Germany
- Max-Planck-Zentrum für Physik und Medizin, 91058, Erlangen, Germany
| | - Raimund Schlüßler
- Biotechnology Center, Center for Molecular and Cellular Bioengineering, Technische Universität Dresden, 01307, Dresden, Germany
| | - Renato Frischknecht
- Department of Biology, Animal Physiology, Friedrich-Alexander-University Erlangen-Nürnberg, 91058, Erlangen, Germany
| | - Anja Wehner
- Mass Spectrometry Core Facility, Max Planck Institute of Biochemistry, 82152, Martinsried, Germany
| | - Nicole Krombholz
- Mass Spectrometry Core Facility, Max Planck Institute of Biochemistry, 82152, Martinsried, Germany
| | - Barbara Steigenberger
- Mass Spectrometry Core Facility, Max Planck Institute of Biochemistry, 82152, Martinsried, Germany
| | - Dimitris Beis
- Experimental Surgery, Clinical and Translational Research Center, Biomedical Research Foundation Academy of Athens, 11527, Athens, Greece
- Laboratory of Biological Chemistry, Faculty of Medicine, School of Health Sciences, University of Ioannina, 45110, Ioannina, Greece
| | - Aya Takeoka
- VIB-Neuroelectronics Research Flanders, 3001, Leuven, Belgium
- Department of Neuroscience and Leuven Brain Institute, KU Leuven, 3000, Leuven, Belgium
| | - Ingmar Blümcke
- Department of Neuropathology, Universitätsklinikum Erlangen, Friedrich-Alexander-University Erlangen-Nürnberg, 91054, Erlangen, Germany
| | - Stephanie Möllmert
- Max Planck Institute for the Science of Light, 91058, Erlangen, Germany
- Max-Planck-Zentrum für Physik und Medizin, 91058, Erlangen, Germany
| | - Kanwarpal Singh
- Max Planck Institute for the Science of Light, 91058, Erlangen, Germany
- Max-Planck-Zentrum für Physik und Medizin, 91058, Erlangen, Germany
- Department of Physics, Friedrich-Alexander-University Erlangen-Nürnberg, 91058, Erlangen, Germany
| | - Jochen Guck
- Max Planck Institute for the Science of Light, 91058, Erlangen, Germany
- Max-Planck-Zentrum für Physik und Medizin, 91058, Erlangen, Germany
- Department of Physics, Friedrich-Alexander-University Erlangen-Nürnberg, 91058, Erlangen, Germany
| | - Katja Kobow
- Department of Neuropathology, Universitätsklinikum Erlangen, Friedrich-Alexander-University Erlangen-Nürnberg, 91054, Erlangen, Germany
| | - Daniel Wehner
- Max Planck Institute for the Science of Light, 91058, Erlangen, Germany.
- Max-Planck-Zentrum für Physik und Medizin, 91058, Erlangen, Germany.
| |
Collapse
|
4
|
Lencer E, Rains A, Binne E, Prekeris R, Artinger KB. Mutations in cdon and boc affect trunk neural crest cell migration and slow-twitch muscle development in zebrafish. Development 2023; 150:dev201304. [PMID: 37390228 PMCID: PMC10357035 DOI: 10.1242/dev.201304] [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: 10/04/2022] [Accepted: 06/22/2023] [Indexed: 07/02/2023]
Abstract
The transmembrane proteins cdon and boc are implicated in regulating hedgehog signaling during vertebrate development. Recent work showing roles for these genes in axon guidance and neural crest cell migration suggest that cdon and boc may play additional functions in regulating directed cell movements. We use newly generated and existing mutants to investigate a role for cdon and boc in zebrafish neural crest cell migration. We find that single mutant embryos exhibit normal neural crest phenotypes, but that neural crest migration is strikingly disrupted in double cdon;boc mutant embryos. We further show that this migration phenotype is associated with defects in the differentiation of slow-twitch muscle cells, and the loss of a Col1a1a-containing extracellular matrix, suggesting that neural crest defects may be a secondary consequence to defects in mesoderm development. Combined, our data add to a growing literature showing that cdon and boc act synergistically to promote hedgehog signaling during vertebrate development, and suggest that the zebrafish can be used to study the function of hedgehog receptor paralogs.
Collapse
Affiliation(s)
- Ezra Lencer
- Department of Cell and Developmental Biology, University of Colorado, Anschutz Medical Campus Aurora, CO 80045, USA
- Department of Craniofacial Biology, University of Colorado, Anschutz Medical Campus Aurora, CO 80045, USA
| | - Addison Rains
- Department of Craniofacial Biology, University of Colorado, Anschutz Medical Campus Aurora, CO 80045, USA
- Cell Biology, Stem Cells and Development Graduate Program, University of Colorado, Anschutz Medical Campus Aurora, CO 80045, USA
| | - Erin Binne
- Department of Craniofacial Biology, University of Colorado, Anschutz Medical Campus Aurora, CO 80045, USA
| | - Rytis Prekeris
- Department of Cell and Developmental Biology, University of Colorado, Anschutz Medical Campus Aurora, CO 80045, USA
| | - Kristin B. Artinger
- Department of Craniofacial Biology, University of Colorado, Anschutz Medical Campus Aurora, CO 80045, USA
- Department of Diagnostic and Biological Sciences, University of Minnesota School of Dentistry, Minneapolis, MN 55455, USA
| |
Collapse
|
5
|
Nemoz-Billet L, Bretaud S, Ruggiero F. [The role of extracellular matrix in the regeneration of motor nerves]. Med Sci (Paris) 2021; 37 Hors série n° 1:11-14. [PMID: 34878386 DOI: 10.1051/medsci/2021183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
The motor neurons (MN) form the ultimate route to convey the commands from the central nervous system to muscles. During development, MN extend axons that follow stereotyped trajectories to their muscle targets, guided by various attractive and repulsive molecular cues. Extracellular matrix (ECM) is a major source of guidance cues, but its role in axonal development and regeneration remains poorly documented. Regenerating axons are able to return to their synaptic target following their original trajectory. The same guidance cues could be thus involved in motor nerve regeneration. Zebrafish has become a popular model system in understanding the development of the peripheral nervous system. Thanks to the generation of fluorescent transgenic lines and the optical transparency of embryos and larvae, it allows direct visualization of axonogenesis. Additionally, and contrary to humans, its remarkable capacity to regenerate makes it well suited for the study of nerve regeneration. A laser method to ablate nerves in living zebrafish larvae has been developed in our laboratory that, combined with the use of the fluorescent mnx1:gfp zebrafish transgenic line, allows the follow up of the dynamics of the nerve regeneration process. To study the role of ECM proteins present in the axonal path, mutant lines for different ECM proteins (already available in our laboratory or generated in mnx1:gfp fish using CRISPR-Cas9 method) will be used to analyze their role during the regeneration process. These mutant lines for ECM will be crossed with existing fluorescent transgenic lines to visualize different cell types involved in the nerve regeneration, such as macrophages (mfap4:mcherry), neutrophils (mpx:gfp) or even Schwann cells (sox10:mrfp). Overall, this study will depict the role of ECM in nerve regeneration and will provide essential knowledge for the development of new biomaterials to promote the regeneration of injured motor nerves.
Collapse
Affiliation(s)
- Laurie Nemoz-Billet
- Institut de Génomique Fonctionnelle de Lyon, Université de Lyon, ENS de Lyon, CNRS UMR 5242, 46 allée d'Italie, Lyon F-69364, France
| | - Sandrine Bretaud
- Institut de Génomique Fonctionnelle de Lyon, Université de Lyon, ENS de Lyon, CNRS UMR 5242, 46 allée d'Italie, Lyon F-69364, France
| | - Florence Ruggiero
- Institut de Génomique Fonctionnelle de Lyon, Université de Lyon, ENS de Lyon, CNRS UMR 5242, 46 allée d'Italie, Lyon F-69364, France
| |
Collapse
|
6
|
Tsata V, Möllmert S, Schweitzer C, Kolb J, Möckel C, Böhm B, Rosso G, Lange C, Lesche M, Hammer J, Kesavan G, Beis D, Guck J, Brand M, Wehner D. A switch in pdgfrb + cell-derived ECM composition prevents inhibitory scarring and promotes axon regeneration in the zebrafish spinal cord. Dev Cell 2021; 56:509-524.e9. [PMID: 33412105 DOI: 10.1016/j.devcel.2020.12.009] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2020] [Revised: 10/12/2020] [Accepted: 12/07/2020] [Indexed: 12/13/2022]
Abstract
In mammals, perivascular cell-derived scarring after spinal cord injury impedes axonal regrowth. In contrast, the extracellular matrix (ECM) in the spinal lesion site of zebrafish is permissive and required for axon regeneration. However, the cellular mechanisms underlying this interspecies difference have not been investigated. Here, we show that an injury to the zebrafish spinal cord triggers recruitment of pdgfrb+ myoseptal and perivascular cells in a PDGFR signaling-dependent manner. Interference with pdgfrb+ cell recruitment or depletion of pdgfrb+ cells inhibits axonal regrowth and recovery of locomotor function. Transcriptional profiling and functional experiments reveal that pdgfrb+ cells upregulate expression of axon growth-promoting ECM genes (cthrc1a and col12a1a/b) and concomitantly reduce synthesis of matrix molecules that are detrimental to regeneration (lum and mfap2). Our data demonstrate that a switch in ECM composition is critical for axon regeneration after spinal cord injury and identify the cellular source and components of the growth-promoting lesion ECM.
Collapse
Affiliation(s)
- Vasiliki Tsata
- Center for Regenerative Therapies TU Dresden, Technische Universität Dresden, 01307 Dresden, Germany; Developmental Biology, Clinical, Experimental Surgery and Translational Research Center, Biomedical Research Foundation Academy of Athens, 11527 Athens, Greece
| | - Stephanie Möllmert
- Max Planck Institute for the Science of Light, 91058 Erlangen, Germany; Max-Planck-Zentrum für Physik und Medizin, 91058 Erlangen, Germany
| | - Christine Schweitzer
- Max Planck Institute for the Science of Light, 91058 Erlangen, Germany; Max-Planck-Zentrum für Physik und Medizin, 91058 Erlangen, Germany
| | - Julia Kolb
- Max Planck Institute for the Science of Light, 91058 Erlangen, Germany; Max-Planck-Zentrum für Physik und Medizin, 91058 Erlangen, Germany
| | - Conrad Möckel
- Max Planck Institute for the Science of Light, 91058 Erlangen, Germany; Max-Planck-Zentrum für Physik und Medizin, 91058 Erlangen, Germany
| | - Benjamin Böhm
- Max Planck Institute for the Science of Light, 91058 Erlangen, Germany; Max-Planck-Zentrum für Physik und Medizin, 91058 Erlangen, Germany
| | - Gonzalo Rosso
- Max Planck Institute for the Science of Light, 91058 Erlangen, Germany; Max-Planck-Zentrum für Physik und Medizin, 91058 Erlangen, Germany; Institute of Physiology II, University of Münster, 48149 Münster, Germany
| | - Christian Lange
- Center for Regenerative Therapies TU Dresden, Technische Universität Dresden, 01307 Dresden, Germany
| | - Mathias Lesche
- DRESDEN-concept Genome Center c/o Center for Molecular and Cellular Bioengineering (CMCB), Technische Universität Dresden, 01307 Dresden, Germany
| | - Juliane Hammer
- Center for Regenerative Therapies TU Dresden, Technische Universität Dresden, 01307 Dresden, Germany
| | - Gokul Kesavan
- Center for Regenerative Therapies TU Dresden, Technische Universität Dresden, 01307 Dresden, Germany
| | - Dimitris Beis
- Developmental Biology, Clinical, Experimental Surgery and Translational Research Center, Biomedical Research Foundation Academy of Athens, 11527 Athens, Greece
| | - Jochen Guck
- Max Planck Institute for the Science of Light, 91058 Erlangen, Germany; Max-Planck-Zentrum für Physik und Medizin, 91058 Erlangen, Germany; Biotechnology Center, Center for Molecular and Cellular Bioengineering, Technische Universität Dresden, 01307 Dresden, Germany
| | - Michael Brand
- Center for Regenerative Therapies TU Dresden, Technische Universität Dresden, 01307 Dresden, Germany; Biotechnology Center, Center for Molecular and Cellular Bioengineering, Technische Universität Dresden, 01307 Dresden, Germany
| | - Daniel Wehner
- Center for Regenerative Therapies TU Dresden, Technische Universität Dresden, 01307 Dresden, Germany; Max Planck Institute for the Science of Light, 91058 Erlangen, Germany; Max-Planck-Zentrum für Physik und Medizin, 91058 Erlangen, Germany.
| |
Collapse
|
7
|
Demirci Y, Cucun G, Poyraz YK, Mohammed S, Heger G, Papatheodorou I, Ozhan G. Comparative Transcriptome Analysis of the Regenerating Zebrafish Telencephalon Unravels a Resource With Key Pathways During Two Early Stages and Activation of Wnt/β-Catenin Signaling at the Early Wound Healing Stage. Front Cell Dev Biol 2020; 8:584604. [PMID: 33163496 PMCID: PMC7581945 DOI: 10.3389/fcell.2020.584604] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Accepted: 09/11/2020] [Indexed: 01/22/2023] Open
Abstract
Owing to its pronounced regenerative capacity in many tissues and organs, the zebrafish brain represents an ideal platform to understand the endogenous regeneration mechanisms that restore tissue integrity and function upon injury or disease. Although radial glial and neuronal cell populations have been characterized with respect to specific marker genes, comprehensive transcriptomic profiling of the regenerating telencephalon has not been conducted so far. Here, by processing the lesioned and unlesioned hemispheres of the telencephalon separately, we reveal the differentially expressed genes (DEGs) at the early wound healing and early proliferative stages of regeneration, i.e., 20 h post-lesion (hpl) and 3 days post-lesion (dpl), respectively. At 20 hpl, we detect a far higher number of DEGs in the lesioned hemisphere than in the unlesioned half and only 7% of all DEGs in both halves. However, this difference disappears at 3 dpl, where the lesioned and unlesioned hemispheres share 40% of all DEGs. By performing an extensive comparison of the gene expression profiles in these stages, we unravel that the lesioned hemispheres at 20 hpl and 3 dpl exhibit distinct transcriptional profiles. We further unveil a prominent activation of Wnt/β-catenin signaling at 20 hpl, returning to control level in the lesioned site at 3 dpl. Wnt/β-catenin signaling indeed appears to control a large number of genes associated primarily with the p53, apoptosis, forkhead box O (FoxO), mitogen-activated protein kinase (MAPK), and mammalian target of rapamycin (mTOR) signaling pathways specifically at 20 hpl. Based on these results, we propose that the lesioned and unlesioned hemispheres react to injury dynamically during telencephalon regeneration and that the activation of Wnt/β-catenin signaling at the early wound healing stage plays a key role in the regulation of cellular and molecular events.
Collapse
Affiliation(s)
- Yeliz Demirci
- İzmir Biomedicine and Genome Center (IBG), Dokuz Eylül University Health Campus, İzmir, Turkey.,İzmir International Biomedicine and Genome Institute (IBG-İzmir), Dokuz Eylül University, İzmir, Turkey.,European Molecular Biology Laboratory - European Bioinformatics Institute (EMBL-EBI), Cambridge, United Kingdom
| | - Gokhan Cucun
- İzmir Biomedicine and Genome Center (IBG), Dokuz Eylül University Health Campus, İzmir, Turkey.,İzmir International Biomedicine and Genome Institute (IBG-İzmir), Dokuz Eylül University, İzmir, Turkey
| | - Yusuf Kaan Poyraz
- İzmir Biomedicine and Genome Center (IBG), Dokuz Eylül University Health Campus, İzmir, Turkey.,İzmir International Biomedicine and Genome Institute (IBG-İzmir), Dokuz Eylül University, İzmir, Turkey
| | - Suhaib Mohammed
- European Molecular Biology Laboratory - European Bioinformatics Institute (EMBL-EBI), Cambridge, United Kingdom
| | | | - Irene Papatheodorou
- European Molecular Biology Laboratory - European Bioinformatics Institute (EMBL-EBI), Cambridge, United Kingdom
| | - Gunes Ozhan
- İzmir Biomedicine and Genome Center (IBG), Dokuz Eylül University Health Campus, İzmir, Turkey.,İzmir International Biomedicine and Genome Institute (IBG-İzmir), Dokuz Eylül University, İzmir, Turkey
| |
Collapse
|
8
|
Oprişoreanu AM, Smith HL, Arya S, Webster R, Zhong Z, Eaton-Hart C, Wehner D, Cardozo MJ, Becker T, Talbot K, Becker CG. Interaction of Axonal Chondrolectin with Collagen XIXa1 Is Necessary for Precise Neuromuscular Junction Formation. Cell Rep 2020; 29:1082-1098.e10. [PMID: 31665626 DOI: 10.1016/j.celrep.2019.09.033] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Revised: 08/13/2019] [Accepted: 09/12/2019] [Indexed: 01/05/2023] Open
Abstract
Chondrolectin (Chodl) is needed for motor axon extension in zebrafish and is dysregulated in mouse models of spinal muscular atrophy (SMA). However, the mechanistic basis of Chodl function is not known. Here, we use Chodl-deficient zebrafish and mouse mutants to show that the absence of Chodl leads to anatomical and functional defects of the neuromuscular synapse. In zebrafish, the growth of an identified motor axon beyond an "en passant" synapse and later axon branching from synaptic points are impaired, leading to functional deficits. Mechanistically, motor-neuron-autonomous Chodl function depends on its intracellular domain and on binding muscle-derived collagen XIXa1 by its extracellular C-type lectin domain. Our data support evolutionarily conserved roles of Chodl in synaptogenesis and provide evidence for a "synapse-first" scenario of motor axon growth in zebrafish.
Collapse
Affiliation(s)
- Ana-Maria Oprişoreanu
- Centre for Discovery Brain Sciences, University of Edinburgh, The Chancellor's Building, 49 Little France Crescent, Edinburgh EH16 4SB, UK
| | - Hannah L Smith
- Centre for Discovery Brain Sciences, University of Edinburgh, The Chancellor's Building, 49 Little France Crescent, Edinburgh EH16 4SB, UK
| | - Sukrat Arya
- Nuffield Department of Clinical Neurosciences, West Wing, John Radcliffe Hospital, University of Oxford, Oxford OX3 9DU, UK
| | - Richard Webster
- Nuffield Department of Clinical Neurosciences, West Wing, John Radcliffe Hospital, University of Oxford, Oxford OX3 9DU, UK
| | - Zhen Zhong
- Centre for Discovery Brain Sciences, University of Edinburgh, The Chancellor's Building, 49 Little France Crescent, Edinburgh EH16 4SB, UK
| | - Charlotte Eaton-Hart
- Nuffield Department of Clinical Neurosciences, West Wing, John Radcliffe Hospital, University of Oxford, Oxford OX3 9DU, UK
| | - Daniel Wehner
- Centre for Discovery Brain Sciences, University of Edinburgh, The Chancellor's Building, 49 Little France Crescent, Edinburgh EH16 4SB, UK
| | - Marcos J Cardozo
- Centre for Discovery Brain Sciences, University of Edinburgh, The Chancellor's Building, 49 Little France Crescent, Edinburgh EH16 4SB, UK
| | - Thomas Becker
- Centre for Discovery Brain Sciences, University of Edinburgh, The Chancellor's Building, 49 Little France Crescent, Edinburgh EH16 4SB, UK; Euan MacDonald Centre for Motor Neurone Disease Research, University of Edinburgh, Edinburgh, UK.
| | - Kevin Talbot
- Nuffield Department of Clinical Neurosciences, West Wing, John Radcliffe Hospital, University of Oxford, Oxford OX3 9DU, UK.
| | - Catherina G Becker
- Centre for Discovery Brain Sciences, University of Edinburgh, The Chancellor's Building, 49 Little France Crescent, Edinburgh EH16 4SB, UK; Euan MacDonald Centre for Motor Neurone Disease Research, University of Edinburgh, Edinburgh, UK.
| |
Collapse
|
9
|
Miller AE, Hu P, Barker TH. Feeling Things Out: Bidirectional Signaling of the Cell-ECM Interface, Implications in the Mechanobiology of Cell Spreading, Migration, Proliferation, and Differentiation. Adv Healthc Mater 2020; 9:e1901445. [PMID: 32037719 PMCID: PMC7274903 DOI: 10.1002/adhm.201901445] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Revised: 01/10/2020] [Indexed: 12/16/2022]
Abstract
Biophysical cues stemming from the extracellular environment are rapidly transduced into discernible chemical messages (mechanotransduction) that direct cellular activities-placing the extracellular matrix (ECM) as a potent regulator of cell behavior. Dynamic reciprocity between the cell and its associated matrix is essential to the maintenance of tissue homeostasis and dysregulation of both ECM mechanical signaling, via pathological ECM turnover, and internal mechanotransduction pathways contribute to disease progression. This review covers the current understandings of the key modes of signaling used by both the cell and ECM to coregulate one another. By taking an outside-in approach, the inherent complexities and regulatory processes at each level of signaling (ECM, plasma membrane, focal adhesion, and cytoplasm) are captured to give a comprehensive picture of the internal and external mechanoregulatory environment. Specific emphasis is placed on the focal adhesion complex which acts as a central hub of mechanical signaling, regulating cell spreading, migration, proliferation, and differentiation. In addition, a wealth of available knowledge on mechanotransduction is curated to generate an integrated signaling network encompassing the central components of the focal adhesion, cytoplasm and nucleus that act in concert to promote durotaxis, proliferation, and differentiation in a stiffness-dependent manner.
Collapse
Affiliation(s)
- Andrew E Miller
- Department of Biomedical Engineering, University of Virginia, 415 Lane Rd. MR5 1225, Charlottesville, VA, 22903, USA
| | - Ping Hu
- Department of Biomedical Engineering, University of Virginia, 415 Lane Rd. MR5 1225, Charlottesville, VA, 22903, USA
| | - Thomas H Barker
- Department of Biomedical Engineering, University of Virginia, 415 Lane Rd. MR5 1225, Charlottesville, VA, 22903, USA
| |
Collapse
|
10
|
Rescan PY. Development of myofibres and associated connective tissues in fish axial muscle: Recent insights and future perspectives. Differentiation 2019; 106:35-41. [PMID: 30852471 DOI: 10.1016/j.diff.2019.02.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Revised: 02/27/2019] [Accepted: 02/28/2019] [Indexed: 01/18/2023]
Abstract
Fish axial muscle consists of a series of W-shaped muscle blocks, called myomeres, that are composed primarily of multinucleated contractile muscle cells (myofibres) gathered together by an intricate network of connective tissue that transmits forces generated by myofibre contraction to the axial skeleton. This review summarises current knowledge on the successive and overlapping myogenic waves contributing to axial musculature formation and growth in fish. Additionally, this review presents recent insights into muscle connective tissue development in fish, focusing on the early formation of collagenous myosepta separating adjacent myomeres and the late formation of intramuscular connective sheaths (i.e. endomysium and perimysium) that is completed only at the fry stage when connective fibroblasts expressing collagens arise inside myomeres. Finally, this review considers the possibility that somites produce not only myogenic, chondrogenic and myoseptal progenitor cells as previously reported, but also mesenchymal cells giving rise to muscle resident fibroblasts.
Collapse
Affiliation(s)
- Pierre-Yves Rescan
- Inra, UR1037 - Laboratoire de Physiologie et Génomique des Poissons, Campus de Beaulieu - Bât 16A, 35042 Rennes Cedex, France.
| |
Collapse
|
11
|
Sever M, Gunay G, Guler MO, Tekinay AB. Tenascin-C derived signaling induces neuronal differentiation in a three-dimensional peptide nanofiber gel. Biomater Sci 2018; 6:1859-1868. [PMID: 29799029 DOI: 10.1039/c7bm00850c] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
The development of new biomaterials mimicking the neuronal extracellular matrix (ECM) requires signals for the induction of neuronal differentiation and regeneration. In addition to the biological and chemical cues, the physical properties of the ECM should also be considered while designing regenerative materials for nervous tissue. In this study, we investigated the influence of the microenvironment on tenascin-C signaling using 2D surfaces and 3D scaffolds generated by a peptide amphiphile nanofiber gel with a tenascin-C derived peptide epitope (VFDNFVLK). While tenascin-C mimetic PA nanofibers significantly increased the length and number of neurites produced by PC12 cells on 2D cell culture, more extensive neurite outgrowth was observed in the 3D gel environment. PC12 cells encapsulated within the 3D tenascin-C mimetic peptide nanofiber gel also exhibited significantly increased expression of neural markers compared to the cells on 2D surfaces. Our results emphasize the synergistic effects of the 3D conformation of peptide nanofibers along with the tenascin-C signaling and growth factors on the neuronal differentiation of PC12 cells, which may further provide more tissue-like morphology for therapeutic applications.
Collapse
Affiliation(s)
- Melike Sever
- Institute of Materials Science and Nanotechnology, National Nanotechnology Research Center (UNAM), Bilkent University, Ankara 06800, Turkey.
| | | | | | | |
Collapse
|
12
|
Abstract
The extracellular matrix (ECM) has central roles in tissue integrity and remodeling throughout the life span of animals. While collagens are the most abundant structural components of ECM in most tissues, tissue-specific molecular complexity is contributed by ECM glycoproteins. The matricellular glycoproteins are categorized primarily according to functional criteria and represented predominantly by the thrombospondin, tenascin, SPARC/osteonectin, and CCN families. These proteins do not self-assemble into ECM fibrils; nevertheless, they shape ECM properties through interactions with structural ECM proteins, growth factors, and cells. Matricellular proteins also promote cell migration or morphological changes through adhesion-modulating or counter-adhesive actions on cell-ECM adhesions, intracellular signaling, and the actin cytoskeleton. Typically, matricellular proteins are most highly expressed during embryonic development. In adult tissues, expression is more limited unless activated by cues for dynamic tissue remodeling and cell motility, such as occur during inflammatory response and wound repair. Many insights in the complex roles of matricellular proteins have been obtained from studies of gene knockout mice. However, with the exception of chordate-specific tenascins, these are highly conserved proteins that are encoded in many animal phyla. This review will consider the increasing body of research on matricellular proteins in nonmammalian animal models. These models provide better access to the very earliest stages of embryonic development and opportunities to study biological processes such as limb and organ regeneration. In aggregate, this research is expanding concepts of the functions and mechanisms of action of matricellular proteins.
Collapse
Affiliation(s)
- Josephine C Adams
- School of Biochemistry, University of Bristol, Bristol, United Kingdom.
| |
Collapse
|
13
|
Learning to swim, again: Axon regeneration in fish. Exp Neurol 2017; 287:318-330. [DOI: 10.1016/j.expneurol.2016.02.022] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2016] [Revised: 02/25/2016] [Accepted: 02/27/2016] [Indexed: 01/10/2023]
|
14
|
Abstract
Tendons are important components of our musculoskeletal system. Injuries to these tissues are very common, resulting from occupational-related injuries, sports-related trauma, and age-related degeneration. Unfortunately, there are few treatment options, and current therapies rarely restore injured tendons to their original function. An improved understanding of the pathways regulating their development and repair would have significant impact in stimulating the formulation of regenerative-based approaches for tendon injury. The zebrafish provides an ideal system in which to perform genetic and chemical screens to identify new pathways involved in tendon biology. Until recently, there had been few descriptions of tendons and ligaments in the zebrafish and their similarity to mammalian tendon tissues. In this chapter, we describe the development of the zebrafish tendon and ligament tissues in the context of their gene expression, structure, and interactions with neighboring musculoskeletal tissues. We highlight the similarities with tendon development in higher vertebrates, showing that the craniofacial tendons and ligaments in zebrafish morphologically, molecularly, and structurally resemble mammalian tendons and ligaments from embryonic to adult stages. We detail methods for fluorescent in situ hybridization and immunohistochemistry as an assay to examine morphological changes in the zebrafish musculoskeleton. Staining assays such as these could provide the foundation for screen-based approaches to identify new regulators of tendon development, morphogenesis, and repair. These discoveries would provide new targets and pathways to study in the context of regenerative medicine-based approaches to improve tendon healing.
Collapse
Affiliation(s)
- J W Chen
- Massachusetts General Hospital, Boston, MA, United States; Harvard Medical School, Boston, MA, United States
| | - J L Galloway
- Massachusetts General Hospital, Boston, MA, United States; Harvard Medical School, Boston, MA, United States
| |
Collapse
|
15
|
Harris GM, Madigan NN, Lancaster KZ, Enquist LW, Windebank AJ, Schwartz J, Schwarzbauer JE. Nerve Guidance by a Decellularized Fibroblast Extracellular Matrix. Matrix Biol 2016; 60-61:176-189. [PMID: 27641621 DOI: 10.1016/j.matbio.2016.08.011] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2016] [Revised: 08/25/2016] [Accepted: 08/25/2016] [Indexed: 12/22/2022]
Abstract
Spinal cord and peripheral nerve injuries require the regeneration of nerve fibers across the lesion site for successful recovery. Providing guidance cues and soluble factors to promote neurite outgrowth and cell survival can enhance repair. The extracellular matrix (ECM) plays a key role in tissue repair by controlling cell adhesion, motility, and growth. In this study, we explored the ability of a mesenchymal ECM to support neurite outgrowth from neurons in the superior cervical ganglia (SCG). Length and morphology of neurites extended on a decellularized fibroblast ECM were compared to those on substrates coated with laminin, a major ECM protein in neural tissue, or fibronectin, the main component of a mesenchymal ECM. Average radial neurite extension was equivalent on laminin and on the decellularized ECM, but contrasted with the shorter, curved neurites observed on the fibronectin substrate. Differences between neurites on fibronectin and on other substrates were confirmed by fast Fourier transform analyses. To control the direction of neurite outgrowth, we developed an ECM with linearly aligned fibril organization by orienting the fibroblasts that deposit the matrix on a polymeric surface micropatterned with a striped chemical interface. Neurites projected from SCGs appeared to reorient in the direction of the pattern. These results highlight the ability of a mesenchymal ECM to enhance neurite extension and to control the directional outgrowth of neurites. This micropatterned decellularized ECM architecture has potential as a regenerative microenvironment for nerve repair.
Collapse
Affiliation(s)
- Greg M Harris
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544
| | | | - Karen Z Lancaster
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544
| | - Lynn W Enquist
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544
| | | | - Jeffrey Schwartz
- Department of Chemistry, Princeton University, Princeton, NJ 08544
| | | |
Collapse
|
16
|
Slow Muscle Precursors Lay Down a Collagen XV Matrix Fingerprint to Guide Motor Axon Navigation. J Neurosci 2016; 36:2663-76. [PMID: 26937007 DOI: 10.1523/jneurosci.2847-15.2016] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
UNLABELLED The extracellular matrix (ECM) provides local positional information to guide motoneuron axons toward their muscle target. Collagen XV is a basement membrane component mainly expressed in skeletal muscle. We have identified two zebrafish paralogs of the human COL15A1 gene, col15a1a and col15a1b, which display distinct expression patterns. Here we show that col15a1b is expressed and deposited in the motor path ECM by slow muscle precursors also called adaxial cells. We further demonstrate that collagen XV-B deposition is both temporally and spatially regulated before motor axon extension from the spinal cord in such a way that it remains in this region after the adaxial cells have migrated toward the periphery of the myotome. Loss- and gain-of-function experiments in zebrafish embryos demonstrate that col15a1b expression and subsequent collagen XV-B deposition and organization in the motor path ECM depend on a previously undescribed two-step mechanism involving Hedgehog/Gli and unplugged/MuSK signaling pathways. In silico analysis predicts a putative Gli binding site in the col15a1b proximal promoter. Using col15a1b promoter-reporter constructs, we demonstrate that col15a1b participates in the slow muscle genetic program as a direct target of Hedgehog/Gli signaling. Loss and gain of col15a1b function provoke pathfinding errors in primary and secondary motoneuron axons both at and beyond the choice point where axon pathway selection takes place. These defects result in muscle atrophy and compromised swimming behavior, a phenotype partially rescued by injection of a smyhc1:col15a1b construct. These reveal an unexpected and novel role for collagen XV in motor axon pathfinding and neuromuscular development. SIGNIFICANCE STATEMENT In addition to the archetypal axon guidance cues, the extracellular matrix provides local information that guides motor axons from the spinal cord to their muscle targets. Many of the proteins involved are unknown. Using the zebrafish model, we identified an unexpected role of the extracellular matrix collagen XV in motor axon pathfinding. We show that the synthesis of collagen XV-B by slow muscle precursors and its deposition in the common motor path are dependent on a novel two-step mechanism that determines axon decisions at a choice point during motor axonogenesis. Zebrafish and humans use common molecular cues and regulatory mechanisms for the neuromuscular system development. And as such, our study reveals COL15A1 as a candidate gene for orphan neuromuscular disorders.
Collapse
|
17
|
Tintignac LA, Brenner HR, Rüegg MA. Mechanisms Regulating Neuromuscular Junction Development and Function and Causes of Muscle Wasting. Physiol Rev 2015; 95:809-52. [DOI: 10.1152/physrev.00033.2014] [Citation(s) in RCA: 224] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
The neuromuscular junction is the chemical synapse between motor neurons and skeletal muscle fibers. It is designed to reliably convert the action potential from the presynaptic motor neuron into the contraction of the postsynaptic muscle fiber. Diseases that affect the neuromuscular junction may cause failure of this conversion and result in loss of ambulation and respiration. The loss of motor input also causes muscle wasting as muscle mass is constantly adapted to contractile needs by the balancing of protein synthesis and protein degradation. Finally, neuromuscular activity and muscle mass have a major impact on metabolic properties of the organisms. This review discusses the mechanisms involved in the development and maintenance of the neuromuscular junction, the consequences of and the mechanisms involved in its dysfunction, and its role in maintaining muscle mass during aging. As life expectancy is increasing, loss of muscle mass during aging, called sarcopenia, has emerged as a field of high medical need. Interestingly, aging is also accompanied by structural changes at the neuromuscular junction, suggesting that the mechanisms involved in neuromuscular junction maintenance might be disturbed during aging. In addition, there is now evidence that behavioral paradigms and signaling pathways that are involved in longevity also affect neuromuscular junction stability and sarcopenia.
Collapse
Affiliation(s)
- Lionel A. Tintignac
- Biozentrum, University of Basel, Basel, Switzerland; Department of Biomedicine, University of Basel, Basel, Switzerland; and INRA, UMR866 Dynamique Musculaire et Métabolisme, Montpellier, France
| | - Hans-Rudolf Brenner
- Biozentrum, University of Basel, Basel, Switzerland; Department of Biomedicine, University of Basel, Basel, Switzerland; and INRA, UMR866 Dynamique Musculaire et Métabolisme, Montpellier, France
| | - Markus A. Rüegg
- Biozentrum, University of Basel, Basel, Switzerland; Department of Biomedicine, University of Basel, Basel, Switzerland; and INRA, UMR866 Dynamique Musculaire et Métabolisme, Montpellier, France
| |
Collapse
|
18
|
Menelaou E, Paul LT, Perera SN, Svoboda KR. Motoneuron axon pathfinding errors in zebrafish: differential effects related to concentration and timing of nicotine exposure. Toxicol Appl Pharmacol 2015; 284:65-78. [PMID: 25668718 PMCID: PMC4567840 DOI: 10.1016/j.taap.2015.01.022] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2014] [Revised: 01/27/2015] [Accepted: 01/28/2015] [Indexed: 12/12/2022]
Abstract
Nicotine exposure during embryonic stages of development can affect many neurodevelopmental processes. In the developing zebrafish, exposure to nicotine was reported to cause axonal pathfinding errors in the later born secondary motoneurons (SMNs). These alterations in SMN axon morphology coincided with muscle degeneration at high nicotine concentrations (15-30 μM). Previous work showed that the paralytic mutant zebrafish known as sofa potato exhibited nicotine-induced effects onto SMN axons at these high concentrations but in the absence of any muscle deficits, indicating that pathfinding errors could occur independent of muscle effects. In this study, we used varying concentrations of nicotine at different developmental windows of exposure to specifically isolate its effects onto subpopulations of motoneuron axons. We found that nicotine exposure can affect SMN axon morphology in a dose-dependent manner. At low concentrations of nicotine, SMN axons exhibited pathfinding errors, in the absence of any nicotine-induced muscle abnormalities. Moreover, the nicotine exposure paradigms used affected the 3 subpopulations of SMN axons differently, but the dorsal projecting SMN axons were primarily affected. We then identified morphologically distinct pathfinding errors that best described the nicotine-induced effects on dorsal projecting SMN axons. To test whether SMN pathfinding was potentially influenced by alterations in the early born primary motoneuron (PMN), we performed dual labeling studies, where both PMN and SMN axons were simultaneously labeled with antibodies. We show that only a subset of the SMN axon pathfinding errors coincided with abnormal PMN axonal targeting in nicotine-exposed zebrafish. We conclude that nicotine exposure can exert differential effects depending on the levels of nicotine and developmental exposure window.
Collapse
Affiliation(s)
- Evdokia Menelaou
- Department of Biological Sciences, Louisiana State University, Baton Rouge, LA 70803, USA
| | - Latoya T Paul
- Department of Biological Sciences, Louisiana State University, Baton Rouge, LA 70803, USA
| | - Surangi N Perera
- Joseph J. Zilber School of Public Health, University of Wisconsin - Milwaukee, Milwaukee, WI 53205, USA
| | - Kurt R Svoboda
- Department of Biological Sciences, Louisiana State University, Baton Rouge, LA 70803, USA; Joseph J. Zilber School of Public Health, University of Wisconsin - Milwaukee, Milwaukee, WI 53205, USA.
| |
Collapse
|
19
|
Abstract
Despite the importance of tendons and ligaments for transmitting movement and providing stability to the musculoskeletal system, their development is considerably less well understood than that of the tissues they serve to connect. Zebrafish have been widely used to address questions in muscle and skeletal development, yet few studies describe their tendon and ligament tissues. We have analyzed in zebrafish the expression of several genes known to be enriched in mammalian tendons and ligaments, including scleraxis (scx), collagen 1a2 (col1a2) and tenomodulin (tnmd), or in the tendon-like myosepta of the zebrafish (xirp2a). Co-expression studies with muscle and cartilage markers demonstrate the presence of scxa, col1a2 and tnmd at sites between the developing muscle and cartilage, and xirp2a at the myotendinous junctions. We determined that the zebrafish craniofacial tendon and ligament progenitors are neural crest derived, as in mammals. Cranial and fin tendon progenitors can be induced in the absence of differentiated muscle or cartilage, although neighboring muscle and cartilage are required for tendon cell maintenance and organization, respectively. By contrast, myoseptal scxa expression requires muscle for its initiation. Together, these data suggest a conserved role for muscle in tendon development. Based on the similarities in gene expression, morphology, collagen ultrastructural arrangement and developmental regulation with that of mammalian tendons, we conclude that the zebrafish tendon populations are homologous to their force-transmitting counterparts in higher vertebrates. Within this context, the zebrafish model can be used to provide new avenues for studying tendon biology in a vertebrate genetic system.
Collapse
Affiliation(s)
- Jessica W Chen
- Center for Regenerative Medicine, Harvard Stem Cell Institute, Department of Orthopaedic Surgery, Massachusetts General Hospital, 185 Cambridge Street, Boston, MA 02114, USA
| | | |
Collapse
|
20
|
Babin PJ, Goizet C, Raldúa D. Zebrafish models of human motor neuron diseases: advantages and limitations. Prog Neurobiol 2014; 118:36-58. [PMID: 24705136 DOI: 10.1016/j.pneurobio.2014.03.001] [Citation(s) in RCA: 154] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2013] [Revised: 03/11/2014] [Accepted: 03/14/2014] [Indexed: 01/08/2023]
Abstract
Motor neuron diseases (MNDs) are an etiologically heterogeneous group of disorders of neurodegenerative origin, which result in degeneration of lower (LMNs) and/or upper motor neurons (UMNs). Neurodegenerative MNDs include pure hereditary spastic paraplegia (HSP), which involves specific degeneration of UMNs, leading to progressive spasticity of the lower limbs. In contrast, spinal muscular atrophy (SMA) involves the specific degeneration of LMNs, with symmetrical muscle weakness and atrophy. Amyotrophic lateral sclerosis (ALS), the most common adult-onset MND, is characterized by the degeneration of both UMNs and LMNs, leading to progressive muscle weakness, atrophy, and spasticity. A review of the comparative neuroanatomy of the human and zebrafish motor systems showed that, while the zebrafish was a homologous model for LMN disorders, such as SMA, it was only partially relevant in the case of UMN disorders, due to the absence of corticospinal and rubrospinal tracts in its central nervous system. Even considering the limitation of this model to fully reproduce the human UMN disorders, zebrafish offer an excellent alternative vertebrate model for the molecular and genetic dissection of MND mechanisms. Its advantages include the conservation of genome and physiological processes and applicable in vivo tools, including easy imaging, loss or gain of function methods, behavioral tests to examine changes in motor activity, and the ease of simultaneous chemical/drug testing on large numbers of animals. This facilitates the assessment of the environmental origin of MNDs, alone or in combination with genetic traits and putative modifier genes. Positive hits obtained by phenotype-based small-molecule screening using zebrafish may potentially be effective drugs for treatment of human MNDs.
Collapse
Affiliation(s)
- Patrick J Babin
- Univ. Bordeaux, Maladies Rares: Génétique et Métabolisme (MRGM), EA 4576, Talence, France.
| | - Cyril Goizet
- Univ. Bordeaux, Maladies Rares: Génétique et Métabolisme (MRGM), EA 4576, Talence, France; CHU Bordeaux, Hôpital Pellegrin, Service de Génétique Médicale, Bordeaux, France
| | | |
Collapse
|
21
|
Lin CY, Huang CC, Wang WD, Hsiao CD, Cheng CF, Wu YT, Lu YF, Hwang SPL. Low temperature mitigates cardia bifida in zebrafish embryos. PLoS One 2013; 8:e69788. [PMID: 23922799 PMCID: PMC3724881 DOI: 10.1371/journal.pone.0069788] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2013] [Accepted: 06/12/2013] [Indexed: 11/18/2022] Open
Abstract
The coordinated migration of bilateral cardiomyocytes and the formation of the cardiac cone are essential for heart tube formation. We investigated gene regulatory mechanisms involved in myocardial migration, and regulation of the timing of cardiac cone formation in zebrafish embryos. Through screening of zebrafish treated with ethylnitrosourea, we isolated a mutant with a hypomorphic allele of mil (s1pr2)/edg5, called s1pr2as10 (as10). Mutant embryos with this allele expressed less mil/edg5 mRNA and exhibited cardia bifida prior to 28 hours post-fertilization. Although the bilateral hearts of the mutants gradually fused together, the resulting formation of two atria and one tightly-packed ventricle failed to support normal blood circulation. Interestingly, cardia bifida of s1pr2as10 embryos could be rescued and normal circulation could be restored by incubating the embryos at low temperature (22.5°C). Rescue was also observed in gata5 and bon cardia bifida morphants raised at 22.5°C. The use of DNA microarrays, digital gene expression analyses, loss-of-function, as well as mRNA and protein rescue experiments, revealed that low temperature mitigates cardia bifida by regulating the expression of genes encoding components of the extracellular matrix (fibronectin 1, tenascin-c, tenascin-w). Furthermore, the addition of N-acetyl cysteine (NAC), a reactive oxygen species (ROS) scavenger, significantly decreased the effect of low temperature on mitigating cardia bifida in s1pr2as10 embryos. Our study reveals that temperature coordinates the development of the heart tube and somitogenesis, and that extracellular matrix genes (fibronectin 1, tenascin-c and tenascin-w) are involved.
Collapse
Affiliation(s)
- Che-Yi Lin
- Institute of Bioscience and Biotechnology, National Taiwan Ocean University, Keelung, Taiwan
| | | | | | | | | | | | | | | |
Collapse
|
22
|
Liu C, Ma W, Su W, Zhang J. Prdm14 acts upstream of islet2 transcription to regulate axon growth of primary motoneurons in zebrafish. Development 2012; 139:4591-600. [PMID: 23136389 DOI: 10.1242/dev.083055] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The precise formation of three-dimensional motor circuits is essential for movement control. Within these circuits, motoneurons (MNs) are specified from spinal progenitors by dorsoventral signals and distinct transcriptional programs. Different MN subpopulations have stereotypic cell body positions and show specific spatial axon trajectories. Our knowledge of MN axon outgrowth remains incomplete. Here, we report a zebrafish gene-trap mutant, short lightning (slg), in which prdm14 expression is disrupted. slg mutant embryos show shortened axons in caudal primary (CaP) MNs resulting in defective embryonic movement. Both the CaP neuronal defects and behavior abnormality of the mutants can be phenocopied by injection of a prdm14 morpholino into wild-type embryos. By removing a copy of the inserted transposon from homozygous mutants, prdm14 expression and normal embryonic movement were restored, confirming that loss of prdm14 expression accounts for the observed defects. Mechanistically, Prdm14 protein binds to the promoter region of islet2, a known transcription factor required for CaP development. Notably, disruption of islet2 function caused similar CaP axon outgrowth defects as observed in slg mutant embryos. Furthermore, overexpression of islet2 in slg mutant embryos rescued the shortened CaP axon phenotypes. Together, these data reveal that prdm14 regulates CaP axon outgrowth through activation of islet2 expression.
Collapse
Affiliation(s)
- Chao Liu
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | | | | | | |
Collapse
|
23
|
Becker TS, Rinkwitz S. Zebrafish as a genomics model for human neurological and polygenic disorders. Dev Neurobiol 2012; 72:415-28. [PMID: 21465670 DOI: 10.1002/dneu.20888] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Whole exome sequencing and, to a lesser extent, genome-wide association studies, have provided unprecedented advances in identifying genes and candidate genomic regions involved in the development of human disease. Further progress will come from sequencing the entire genome of multiple patients and normal controls to evaluate overall mutational burden and disease risk. A major challenge will be the interpretation of the resulting data and distinguishing true pathogenic mutations from rare benign variants.While in model organisms such as the zebrafish,mutants are sought that disrupt the function of individual genes, human mutations that cause, or are associated with, the development of disease, are often not acting in a Mendelian fashion, are frequently of small effect size, are late onset, and may reside in noncoding parts of the genome. The zebrafish model is uniquely poised for understanding human coding- and noncoding variants because of its sequenced genome, a large body of knowledge on gene expression and function, rapid generation time, and easy access to embryos. A critical advantage is the ease of zebrafish transgenesis, both for the testing of human regulatory DNA driving expression of fluorescent reporter proteins, and the expression of mutated disease-associated human proteins in specific neurons to rapidly model aspects of neurological disorders. The zebrafish affords progress both through its model genome and it is rapidly developing transparent model vertebrate embryo.
Collapse
Affiliation(s)
- Thomas S Becker
- Sydney Medical School, University of Sydney, Camperdown, Australia.
| | | |
Collapse
|
24
|
Chondrolectin mediates growth cone interactions of motor axons with an intermediate target. J Neurosci 2012; 32:4426-39. [PMID: 22457492 DOI: 10.1523/jneurosci.5179-11.2012] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
The C-type lectin chondrolectin (chodl) represents one of the major gene products dysregulated in spinal muscular atrophy models in mice. However, to date, no function has been determined for the gene. We have identified chodl and other novel genes potentially involved in motor axon differentiation, by expression profiling of transgenically labeled motor neurons in embryonic zebrafish. To enrich the profile for genes involved in differentiation of peripheral motor axons, we inhibited the function of LIM-HDs (LIM homeodomain factors) by overexpression of a dominant-negative cofactor, thereby rendering labeled axons unable to grow out of the spinal cord. Importantly, labeled cells still exhibited axon growth and most cells retained markers of motor neuron identity. Functional tests of chodl, by overexpression and knockdown, confirm crucial functions of this gene for motor axon growth in vivo. Indeed, knockdown of chodl induces arrest or stalling of motor axon growth at the horizontal myoseptum, an intermediate target and navigational choice point, and reduced muscle innervation at later developmental stages. This phenotype is rescued by chodl overexpression, suggesting that correct expression levels of chodl are important for interactions of growth cones of motor axons with the horizontal myoseptum. Combined, these results identify upstream regulators and downstream functions of chodl during motor axon growth.
Collapse
|
25
|
Role of zebrafish lbx2 in embryonic lateral line development. PLoS One 2011; 6:e29515. [PMID: 22216300 PMCID: PMC3245281 DOI: 10.1371/journal.pone.0029515] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2011] [Accepted: 11/29/2011] [Indexed: 11/19/2022] Open
Abstract
Background The zebrafish ladybird homeobox homologous gene 2 (lbx2) has been suggested to play a key role in the regulation of hypaxial myogenic precursor cell migration. Unlike their lbx counterparts in mammals, the function of teleost lbx genes beyond myogenesis during embryonic development remains unexplored. Principal Findings Abrogation of lbx2 function using a specific independent morpholino oligonucleotide (MO) or truncated lbx2 mRNA with an engrailed domain deletion (lbx2eh-) resulted in defective formation of the zebrafish posterior lateral line (PLL). Migration of the PLL primordium was altered and accompanied by increased cell death in the primordium of lbx2-MO-injected embryos. A decreased number of muscle pioneer cells and impaired expression pattern of sdf1a in the horizontal myoseptum was observed in lbx2 morphants. Significance Injection of lbx2 MO or lbx2eh- mRNA resulted in defective PPL formation and altered sdf1a expression, confirming an important function for lbx2 in sdf1a-dependent migration. In addition, the disassociation of PPL nerve extension with PLL primordial migration in some lbx2 morphants suggests that pathfinding of the PLL primordium and the lateral line nerve may be regulated independently.
Collapse
|
26
|
Pandey R, Blanco J, Udolph G. The glucuronyltransferase GlcAT-P is required for stretch growth of peripheral nerves in Drosophila. PLoS One 2011; 6:e28106. [PMID: 22132223 PMCID: PMC3223219 DOI: 10.1371/journal.pone.0028106] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2011] [Accepted: 11/01/2011] [Indexed: 12/29/2022] Open
Abstract
During development, the growth of the animal body is accompanied by a concomitant elongation of the peripheral nerves, which requires the elongation of integrated nerve fibers and the axons projecting therein. Although this process is of fundamental importance to almost all organisms of the animal kingdom, very little is known about the mechanisms regulating this process. Here, we describe the identification and characterization of novel mutant alleles of GlcAT-P, the Drosophila ortholog of the mammalian glucuronyltransferase b3gat1. GlcAT-P mutants reveal shorter larval peripheral nerves and an elongated ventral nerve cord (VNC). We show that GlcAT-P is expressed in a subset of neurons in the central brain hemispheres, in some motoneurons of the ventral nerve cord as well as in central and peripheral nerve glia. We demonstrate that in GlcAT-P mutants the VNC is under tension of shorter peripheral nerves suggesting that the VNC elongates as a consequence of tension imparted by retarded peripheral nerve growth during larval development. We also provide evidence that for growth of peripheral nerve fibers GlcAT-P is critically required in hemocytes; however, glial cells are also important in this process. The glial specific repo gene acts as a modifier of GlcAT-P and loss or reduction of repo function in a GlcAT-P mutant background enhances VNC elongation. We propose a model in which hemocytes are required for aspects of glial cell biology which in turn affects the elongation of peripheral nerves during larval development. Our data also identifies GlcAT-P as a first candidate gene involved in growth of integrated peripheral nerves and therefore establishes Drosophila as an amenable in-vivo model system to study this process at the cellular and molecular level in more detail.
Collapse
Affiliation(s)
- Rahul Pandey
- Neural Development and Repair, Institute of Medical Biology, Singapore, Singapore
| | - Jorge Blanco
- Neural Development and Repair, Institute of Medical Biology, Singapore, Singapore
| | - Gerald Udolph
- Neural Development and Repair, Institute of Medical Biology, Singapore, Singapore
- * E-mail:
| |
Collapse
|
27
|
Hale LA, Fowler DK, Eisen JS. Netrin signaling breaks the equivalence between two identified zebrafish motoneurons revealing a new role of intermediate targets. PLoS One 2011; 6:e25841. [PMID: 22003409 PMCID: PMC3189217 DOI: 10.1371/journal.pone.0025841] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2011] [Accepted: 09/12/2011] [Indexed: 12/11/2022] Open
Abstract
Background We previously showed that equivalence between two identified zebrafish motoneurons is broken by interactions with identified muscle fibers that act as an intermediate target for the axons of these motoneurons. Here we investigate the molecular basis of the signaling interaction between the intermediate target and the motoneurons. Principal Findings We provide evidence that Netrin 1a is an intermediate target-derived signal that causes two equivalent motoneurons to adopt distinct fates. We show that although these two motoneurons express the same Netrin receptors, their axons respond differently to Netrin 1a encountered at the intermediate target. Furthermore, we demonstrate that when Netrin 1a is knocked down, more distal intermediate targets that express other Netrins can also function to break equivalence between these motoneurons. Significance Our results suggest a new role for intermediate targets in breaking neuronal equivalence. The data we present reveal that signals encountered during axon pathfinding can cause equivalent neurons to adopt distinct fates. Such signals may be key in diversifying a neuronal population and leading to correct circuit formation.
Collapse
Affiliation(s)
- Laura A. Hale
- Institute of Neuroscience, University of Oregon, Eugene, Oregon, United States of America
| | - Daniel K. Fowler
- Institute of Neuroscience, University of Oregon, Eugene, Oregon, United States of America
| | - Judith S. Eisen
- Institute of Neuroscience, University of Oregon, Eugene, Oregon, United States of America
- * E-mail:
| |
Collapse
|
28
|
Banerjee S, Gordon L, Donn TM, Berti C, Moens CB, Burden SJ, Granato M. A novel role for MuSK and non-canonical Wnt signaling during segmental neural crest cell migration. Development 2011; 138:3287-96. [PMID: 21750038 DOI: 10.1242/dev.067306] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Trunk neural crest cells delaminate from the dorsal neural tube as an uninterrupted sheet; however, they convert into segmentally organized streams before migrating through the somitic territory. These neural crest cell streams join the segmental trajectories of pathfinding spinal motor axons, suggesting that interactions between these two cell types might be important for neural crest cell migration. Here, we show that in the zebrafish embryo migration of both neural crest cells and motor axons is temporally synchronized and spatially restricted to the center of the somite, but that motor axons are dispensable for segmental neural crest cell migration. Instead, we find that muscle-specific receptor kinase (MuSK) and its putative ligand Wnt11r are crucial for restricting neural crest cell migration to the center of each somite. Moreover, we find that blocking planar cell polarity (PCP) signaling in somitic muscle cells also results in non-segmental neural crest cell migration. Using an F-actin biosensor we show that in the absence of MuSK neural crest cells fail to retract non-productive leading edges, resulting in non-segmental migration. Finally, we show that MuSK knockout mice display similar neural crest cell migration defects, suggesting a novel, evolutionarily conserved role for MuSK in neural crest migration. We propose that a Wnt11r-MuSK dependent, PCP-like pathway restricts neural crest cells to their segmental path.
Collapse
Affiliation(s)
- Santanu Banerjee
- Department of Cell and Developmental Biology, University of Pennsylvania School of Medicine, Philadelphia, PA 19104-6058, USA
| | | | | | | | | | | | | |
Collapse
|
29
|
Yu YM, Cristofanilli M, Valiveti A, Ma L, Yoo M, Morellini F, Schachner M. The extracellular matrix glycoprotein tenascin-C promotes locomotor recovery after spinal cord injury in adult zebrafish. Neuroscience 2011; 183:238-50. [PMID: 21443931 DOI: 10.1016/j.neuroscience.2011.03.043] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2011] [Revised: 03/19/2011] [Accepted: 03/21/2011] [Indexed: 12/31/2022]
Abstract
Adult zebrafish, by virtue of exhibiting spontaneous recovery after spinal lesion, have evolved into a paradigmatic vertebrate model system to identify novel genes vital for successful regeneration after spinal cord injury. Due to a remarkable level of conservation between zebrafish and human genomes, such genes, once identified, could point to possibilities for addressing the multiple issues on how to deal with functional recovery after spinal cord injury in humans. In the current study, the extracellular matrix glycoprotein tenascin-C was studied in the zebrafish spinal cord injury model to assess the often disparate functions of this multidomain molecule under in vivo conditions. This in vivo study was deemed necessary since in vitro studies had shown discrepant functional effects on neurite outgrowth: tenascin-C inhibits neurite outgrowth when presented as a molecular barrier adjacent to a conducive substrate, but enhances neurite outgrowth when presented as a uniform substrate. Thus, our current study addresses the question as to which of these features prevails in vivo: whether tenascin-C reduces or enhances axonal regrowth after injury in a well accepted vertebrate model of spinal cord injury. We show upregulation of tenascin-C expression in regenerating neurons of the nucleus of median longitudinal fascicle (NMLF) in the brainstem and spinal motoneurons. Inhibition of tenascin-C expression by antisense oligonucleotide (morpholino) resulted in impaired locomotor recovery, reduced regrowth of axons from brainstem neurons and reduced synapse formation by the regrowing brainstem axons on spinal motoneurons, all vital indicators of regeneration. Our results thus point to an advantageous role of tenascin-C in promoting spinal cord regeneration, by promoting axonal regrowth and synapse formation in the spinal cord caudal to the lesion site after injury.
Collapse
Affiliation(s)
- Y-M Yu
- W. M. Keck Center for Collaborative Neuroscience and Department of Cell Biology and Neuroscience, Rutgers, The State University of New Jersey, Piscataway, 604 Allison Road, NJ 08854, USA
| | | | | | | | | | | | | |
Collapse
|
30
|
Hilario JD, Wang C, Beattie CE. Collagen XIXa1 is crucial for motor axon navigation at intermediate targets. Development 2010; 137:4261-9. [PMID: 21098567 DOI: 10.1242/dev.051730] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
During development, motor axons navigate from the spinal cord to their muscle targets in the periphery using stereotyped pathways. These pathways are broken down into shorter segments by intermediate targets where axon growth cones are believed to coordinate guidance cues. In zebrafish stumpy mutants, embryonic development proceeds normally; however, as trunk motor axons stall at their intermediate targets, suggesting that Stumpy is needed specifically for motor axon growth cones to proceed past intermediate targets. Fine mapping and positional cloning revealed that stumpy was the zebrafish homolog of the atypical FACIT collagen collagenXIXa1 (colXIX). colXIX expression was observed in a temporal and spatial pattern, consistent with a role in motor axon guidance at intermediate targets. Knocking down zebrafish ColXIX phenocopied the stumpy phenotype and this morpholino phenotype could be rescued by adding back either mouse or zebrafish colXIX RNA. The stumpy phenotype was also partially rescued in mutants by first knocking down zebrafish ColXIX and adding back colXIX RNA, suggesting that the mutation is acting as a dominant negative. Together, these results demonstrate a novel function for a FACIT collagen in guiding vertebrate motor axons through intermediate targets.
Collapse
Affiliation(s)
- Jona D Hilario
- The Ohio State University Center for Molecular Neurobiology and Department of Neuroscience, Columbus, OH 43210, USA
| | | | | |
Collapse
|
31
|
Klausmeyer A, Conrad R, Faissner A, Wiese S. Influence of glial-derived matrix molecules, especially chondroitin sulfates, on neurite growth and survival of cultured mouse embryonic motoneurons. J Neurosci Res 2010; 89:127-41. [PMID: 21162121 DOI: 10.1002/jnr.22531] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2010] [Revised: 09/02/2010] [Accepted: 09/08/2010] [Indexed: 12/24/2022]
Abstract
Mechanisms controlling neuronal survival and regeneration play an important role during development, after birth, and under lesion conditions. Isolated embryonic mouse motoneurons have been a useful tool for studying such basic mechanisms. These cultured motoneurons depend on extracellular matrix (ECM) molecules, which are potent mediators of survival and axonal growth and guidance in the CNS and in vitro, exhibiting either attractive or repellent guidance cues. Additionally, ECM proteoglycans and glycoproteins are components of the glial scar acting as a growth barrier for regenerating axons. Compared with CNS axon outgrowth, less is known about the cues that guide motoneurons toward their peripheral targets. Because we are interested in the effects of glial-derived chondroitin sulfate proteoglycans (CSPGs), we have worked out a model system for investigating the influences of glial-derived matrix molecules on motoneuron outgrowth and survival. We used cultured embryonic mouse motoneurons to investigate axon growth effects of matrix molecules produced by the glial-derived cell lines A7, Neu7, and Oli-neu primary astrocytes as well as the immortalized Schwann cell line IMS32. The results indicate that molecules of the ECM, especially chondroitin sulfates, play an important role as axon growth-promoting cues. We could demonstrate a modifying effect of the matrix components on motoneuron survival and caspase3-induced apoptosis.
Collapse
Affiliation(s)
- Alice Klausmeyer
- Department of Cellmorphology and Molecular Neurobiology, Laboratory of Molecular Cellbiology, Faculty of Biology and Biotechnology, Ruhr-University-Bochum, Bochum, Germany
| | | | | | | |
Collapse
|
32
|
Jing L, Gordon LR, Shtibin E, Granato M. Temporal and spatial requirements of unplugged/MuSK function during zebrafish neuromuscular development. PLoS One 2010; 5:e8843. [PMID: 20107509 PMCID: PMC2809748 DOI: 10.1371/journal.pone.0008843] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2009] [Accepted: 12/15/2009] [Indexed: 01/22/2023] Open
Abstract
One of the earliest events in neuromuscular junction (NMJ) development is the accumulation of acetylcholine receptor (AChR) at the center of muscle cells. The unplugged/MuSK (muscle specific tyrosine kinase) gene is essential to initiate AChR clustering but also to restrict approaching growth cones to the muscle center, thereby coordinating pre- and postsynaptic development. To determine how unplugged/MuSK signaling coordinates these two processes, we examined the temporal and spatial requirements of unplugged/MuSK in zebrafish embryos using heat-shock inducible transgenes. Here, we show that despite its expression in muscle cells from the time they differentiate, unplugged/MuSK activity is first required just prior to the appearance of AChR clusters to simultaneously induce AChR accumulation and to guide motor axons. Furthermore, we demonstrate that ectopic expression of unplugged/MuSK throughout the muscle membrane results in wildtype-like AChR prepattern and neuromuscular synapses in the central region of muscle cells. We propose that AChR prepatterning and axonal guidance are spatio-temporally coordinated through common unplugged/MuSK signals, and that additional factor(s) restrict unplugged/MuSK signaling to a central muscle zone critical for establishing mid-muscle synaptogenesis.
Collapse
Affiliation(s)
- Lili Jing
- Department of Cell and Developmental Biology, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
- Department of Biology, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Laura R. Gordon
- Department of Cell and Developmental Biology, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Elena Shtibin
- Department of Cell and Developmental Biology, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Michael Granato
- Department of Cell and Developmental Biology, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
- * E-mail:
| |
Collapse
|
33
|
Abstract
Multiple molecular cues guide neuronal axons to their targets during development. Previous studies in vitro have shown that mechanical stimulation also can affect axon growth; however, whether mechanical force contributes to axon guidance in vivo is unknown. We investigated the role of muscle contractions in the guidance of zebrafish peripheral Rohon-Beard (RB) sensory axons in vivo. We analyzed several mutants that affect muscle contraction through different molecular pathways, including a new mutant allele of the titin a (pik) gene, mutants that affect the hedgehog signaling pathway, and a nicotinic acetylcholine receptor mutant. We found RB axon defects in these mutants, the severity of which appeared to correlate with the extent of muscle contraction loss. These axons extend between the muscle and skin and normally have ventral trajectories and repel each other on contact. RB peripheral axons in muscle mutants extend longitudinally instead of ventrally, and the axons fail to repel one another on contact. In addition, we showed that limiting muscle movements by embedding embryos in agarose caused similar defects in peripheral RB axon guidance. This work suggests that the mechanical forces generated by muscle contractions are necessary for proper sensory axon pathfinding in vivo.
Collapse
|
34
|
Kii I, Nishiyama T, Li M, Matsumoto KI, Saito M, Amizuka N, Kudo A. Incorporation of tenascin-C into the extracellular matrix by periostin underlies an extracellular meshwork architecture. J Biol Chem 2009; 285:2028-39. [PMID: 19887451 DOI: 10.1074/jbc.m109.051961] [Citation(s) in RCA: 213] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Extracellular matrix (ECM) underlies a complicated multicellular architecture that is subjected to significant forces from mechanical environment. Although various components of the ECM have been enumerated, mechanisms that evolve the sophisticated ECM architecture remain to be addressed. Here we show that periostin, a matricellular protein, promotes incorporation of tenascin-C into the ECM and organizes a meshwork architecture of the ECM. We found that both periostin null mice and tenascin-C null mice exhibited a similar phenotype, confined tibial periostitis, which possibly corresponds to medial tibial stress syndrome in human sports injuries. Periostin possessed adjacent domains that bind to tenascin-C and the other ECM protein: fibronectin and type I collagen, respectively. These adjacent domains functioned as a bridge between tenascin-C and the ECM, which increased deposition of tenascin-C on the ECM. The deposition of hexabrachions of tenascin-C may stabilize bifurcations of the ECM fibrils, which is integrated into the extracellular meshwork architecture. This study suggests a role for periostin in adaptation of the ECM architecture in the mechanical environment.
Collapse
Affiliation(s)
- Isao Kii
- Department of Biological Information, Tokyo Institute of Technology, 4259 Midori-ku, Nagatsuta, Yokohama 226-8501
| | | | | | | | | | | | | |
Collapse
|
35
|
Wnt signals organize synaptic prepattern and axon guidance through the zebrafish unplugged/MuSK receptor. Neuron 2009; 61:721-33. [PMID: 19285469 DOI: 10.1016/j.neuron.2008.12.025] [Citation(s) in RCA: 148] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2008] [Revised: 11/13/2008] [Accepted: 12/24/2008] [Indexed: 11/24/2022]
Abstract
Early during neuromuscular development, acetylcholine receptors (AChRs) accumulate at the center of muscle fibers, precisely where motor growth cones navigate and synapses eventually form. Here, we show that Wnt11r binds to the zebrafish unplugged/MuSK ectodomain to organize this central muscle zone. In the absence of such a zone, prepatterned AChRs fail to aggregate and, as visualized by live-cell imaging, growth cones stray from their central path. Using inducible unplugged/MuSK transgenes, we show that organization of the central muscle zone is dispensable for the formation of neural synapses, but essential for AChR prepattern and motor growth cone guidance. Finally, we show that blocking noncanonical dishevelled signaling in muscle fibers disrupts AChR prepatterning and growth cone guidance. We propose that Wnt ligands activate unplugged/MuSK signaling in muscle fibers to restrict growth cone guidance and AChR prepatterns to the muscle center through a mechanism reminiscent of the planar cell polarity pathway.
Collapse
|
36
|
Hilario JD, Rodino-Klapac LR, Wang C, Beattie CE. Semaphorin 5A is a bifunctional axon guidance cue for axial motoneurons in vivo. Dev Biol 2008; 326:190-200. [PMID: 19059233 DOI: 10.1016/j.ydbio.2008.11.007] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2008] [Revised: 11/07/2008] [Accepted: 11/10/2008] [Indexed: 10/21/2022]
Abstract
Semaphorins are a large class of proteins that function throughout the nervous system to guide axons. It had previously been shown that Semaphorin 5A (Sema5A) was a bifunctional axon guidance cue for mammalian midbrain neurons. We found that zebrafish sema5A was expressed in myotomes during the period of motor axon outgrowth. To determine whether Sema5A functioned in motor axon guidance, we knocked down Sema5A, which resulted in two phenotypes: a delay in motor axon extension into the ventral myotome and aberrant branching of these motor axons. Both phenotypes were rescued by injection of full-length rat Sema5A mRNA. However, adding back RNA encoding the sema domain alone significantly rescued the branching phenotype in sema5A morphants. Conversely, adding back RNA encoding the thrombospondin repeat (TSR) domain alone into sema5A morphants exclusively rescued delay in ventral motor axon extension. Together, these data show that Sema5A is a bifunctional axon guidance cue for vertebrate motor axons in vivo. The TSR domain promotes growth of developing motor axons into the ventral myotome whereas the sema domain mediates repulsion and keeps these motor axons from branching into surrounding myotome regions.
Collapse
Affiliation(s)
- Jona D Hilario
- Center for Molecular Neurobiology and Department of Neuroscience, The Ohio State University, 190 Rightmire Hall, 1060 Carmack Rd, Columbus, OH 43210, USA
| | | | | | | |
Collapse
|
37
|
Sato-Maeda M, Obinata M, Shoji W. Position fine-tuning of caudal primary motoneurons in the zebrafish spinal cord. Development 2008; 135:323-32. [DOI: 10.1242/dev.007559] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
In zebrafish embryos, each myotome is typically innervated by three primary motoneurons (PMNs): the caudal primary (CaP), middle primary (MiP) and rostral primary (RoP). PMN axons first exit the spinal cord through a single exit point located at the midpoint of the overlying somite, which is formed beneath the CaP cell body and is pioneered by the CaP axon. However, the placement of CaP cell bodies with respect to corresponding somites is poorly understood. Here, we determined the early events in CaP cell positioning using neuropilin 1a (nrp1a):gfp transgenic embryos in which CaPs were specifically labeled with GFP. CaP cell bodies first exhibit an irregular pattern in presence of newly formed corresponding somites and then migrate to achieve their proper positions by axonogenesis stages. CaPs are generated in excess compared with the number of somites, and two CaPs often overlap at the same position through this process. Next, we showed that CaP cell bodies remain in the initial irregular positions after knockdown of Neuropilin1a, a component of the class III semaphorin receptor. Irregular CaP position frequently results in aberrant double exit points of motor axons, and secondary motor axons form aberrant exit points following CaP axons. Its expression pattern suggests that sema3ab regulates the CaP position. Indeed, irregular CaP positions and exit points are induced by Sema3ab knockdown, whose ectopic expression can alter the position of CaP cell bodies. Results suggest that Semaphorin-Neuropilin signaling plays an important role in position fine-tuning of CaP cell bodies to ensure proper exit points of motor axons.
Collapse
Affiliation(s)
- Mika Sato-Maeda
- Department of Cell Biology, Institute of Development, Aging and Cancer,Tohoku University, Sendai 980-8575, Japan
| | - Masuo Obinata
- Department of Cell Biology, Institute of Development, Aging and Cancer,Tohoku University, Sendai 980-8575, Japan
| | - Wataru Shoji
- Department of Cell Biology, Institute of Development, Aging and Cancer,Tohoku University, Sendai 980-8575, Japan
| |
Collapse
|
38
|
Feldner J, Reimer MM, Schweitzer J, Wendik B, Meyer D, Becker T, Becker CG. PlexinA3 restricts spinal exit points and branching of trunk motor nerves in embryonic zebrafish. J Neurosci 2007; 27:4978-83. [PMID: 17475806 PMCID: PMC6672091 DOI: 10.1523/jneurosci.1132-07.2007] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
The pioneering primary motor axons in the zebrafish trunk are guided by multiple cues along their pathways. Plexins are receptor components for semaphorins that influence motor axon growth and path finding. We cloned plexinA3 in zebrafish and localized plexinA3 mRNA in primary motor neurons during axon outgrowth. Antisense morpholino knock-down led to substantial errors in motor axon growth. Errors comprised aberrant branching of primary motor nerves as well as additional exit points of axons from the spinal cord. Excessively branched and supernumerary nerves were found in both ventral and dorsal pathways of motor axons. The trunk environment and several other types of axons, including trigeminal axons, were not detectably affected by plexinA3 knock-down. RNA overexpression rescued all morpholino effects. Synergistic effects of combined morpholino injections indicate interactions of plexinA3 with semaphorin3A homologs. Thus, plexinA3 is a crucial receptor for axon guidance cues in primary motor neurons.
Collapse
Affiliation(s)
- Julia Feldner
- Institute for Molecular Bioscience, University of Queensland, St Lucia QLD 4072, Australia
- Zentrum für Molekulare Neurobiologie, University of Hamburg, D-20246 Hamburg, Germany
| | - Michell M. Reimer
- Centre for Neuroscience Research, Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh EH9 1QH, United Kingdom
| | - Jörn Schweitzer
- Institut für Biologie 1, Universität Freiburg, Freiburg, D-79104, Germany
| | - Björn Wendik
- Institut für Biologie 1, Universität Freiburg, Freiburg, D-79104, Germany
| | - Dirk Meyer
- Institut für Molekularbiologie, Leopold-Franzens-Universität Innsbruck, A-6020 Innsbruck, Austria, and
| | - Thomas Becker
- Centre for Neuroscience Research, Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh EH9 1QH, United Kingdom
- Zentrum für Molekulare Neurobiologie, University of Hamburg, D-20246 Hamburg, Germany
| | - Catherina G. Becker
- Centre for Neuroscience Research, Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh EH9 1QH, United Kingdom
- Zentrum für Molekulare Neurobiologie, University of Hamburg, D-20246 Hamburg, Germany
| |
Collapse
|
39
|
Tucker RP, Drabikowski K, Hess JF, Ferralli J, Chiquet-Ehrismann R, Adams JC. Phylogenetic analysis of the tenascin gene family: evidence of origin early in the chordate lineage. BMC Evol Biol 2006; 6:60. [PMID: 16893461 PMCID: PMC1578592 DOI: 10.1186/1471-2148-6-60] [Citation(s) in RCA: 81] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2006] [Accepted: 08/07/2006] [Indexed: 11/25/2022] Open
Abstract
BACKGROUND Tenascins are a family of glycoproteins found primarily in the extracellular matrix of embryos where they help to regulate cell proliferation, adhesion and migration. In order to learn more about their origins and relationships to each other, as well as to clarify the nomenclature used to describe them, the tenascin genes of the urochordate Ciona intestinalis, the pufferfish Tetraodon nigroviridis and Takifugu rubripes and the frog Xenopus tropicalis were identified and their gene organization and predicted protein products compared with the previously characterized tenascins of amniotes. RESULTS A single tenascin gene was identified in the genome of C. intestinalis that encodes a polypeptide with domain features common to all vertebrate tenascins. Both pufferfish genomes encode five tenascin genes: two tenascin-C paralogs, a tenascin-R with domain organization identical to mammalian and avian tenascin-R, a small tenascin-X with previously undescribed GK repeats, and a tenascin-W. Four tenascin genes corresponding to tenascin-C, tenascin-R, tenascin-X and tenascin-W were also identified in the X. tropicalis genome. Multiple sequence alignment reveals that differences in the size of tenascin-W from various vertebrate classes can be explained by duplications of specific fibronectin type III domains. The duplicated domains are encoded on single exons and contain putative integrin-binding motifs. A phylogenetic tree based on the predicted amino acid sequences of the fibrinogen-related domains demonstrates that tenascin-C and tenascin-R are the most closely related vertebrate tenascins, with the most conserved repeat and domain organization. Taking all lines of evidence together, the data show that the tenascins referred to as tenascin-Y and tenascin-N are actually members of the tenascin-X and tenascin-W gene families, respectively. CONCLUSION The presence of a tenascin gene in urochordates but not other invertebrate phyla suggests that tenascins may be specific to chordates. Later genomic duplication events led to the appearance of four family members in vertebrates: tenascin-C, tenascin-R, tenascin-W and tenascin-X.
Collapse
Affiliation(s)
- RP Tucker
- Department of Cell Biology and Human Anatomy, University of California at Davis, Davis, CA 95616, USA
| | - K Drabikowski
- Friedrich Miescher Institute, Novartis Research Foundation, Basel, Switzerland
- Institute of Biology 3, University of Freiburg, Freiburg, Germany
| | - JF Hess
- Department of Cell Biology and Human Anatomy, University of California at Davis, Davis, CA 95616, USA
| | - J Ferralli
- Friedrich Miescher Institute, Novartis Research Foundation, Basel, Switzerland
| | - R Chiquet-Ehrismann
- Friedrich Miescher Institute, Novartis Research Foundation, Basel, Switzerland
| | - JC Adams
- Dept. of Cell Biology, Lerner Research Institute and Dept. of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Cleveland Clinic Foundation, Cleveland, OH 44118, USA
| |
Collapse
|