1
|
Awad M, Sayed RKA, Mohammadin D, Hussein MM, Mokhtar DM. Structural characteristics and regenerative potential: Insights from the molly fish spinal cord. Microsc Res Tech 2024; 87:2643-2653. [PMID: 38923674 DOI: 10.1002/jemt.24633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 05/16/2024] [Accepted: 05/31/2024] [Indexed: 06/28/2024]
Abstract
Unlike mammals, species such as fish and amphibians can regenerate damaged spinal cords, offering insights into potential therapeutic targets. This study investigates the structural features of the molly fish spinal cord through light and electron microscopy. The most notable characteristic was the presence of Mauthner cells (M-cells), which exhibited large cell bodies and processes, as well as synaptic connections with astrocytes. These astrocytic connections contained synaptic vesicles, suggesting electrical transmission at the M-cell endings. Astrocytes, which were labeled with glial fibrillary acidic protein (GFAP), contained cytoplasmic glycogen granules, potentially serving as an emergency fuel source. Two types of oligodendrocytes were identified: a small, dark cell and a larger, lighter cell, both of which reacted strongly with oligodendrocyte transcription factor 2 (Olig2). The dark oligodendrocyte resembled human oligodendrocyte precursors, while the light oligodendrocyte was similar to mature human oligodendrocytes. Additionally, proliferative neurons in the substantia grisea centralis expressed myostatin, Nrf2, and Sox9. Collectively, these findings suggest that the molly fish spinal cord has advanced structural features conducive to spinal cord regeneration and could serve as an excellent model for studying central nervous system regeneration. Further studies on the functional aspects of the molly fish spinal cord are recommended. RESEARCH HIGHLIGHTS: Mauthner cells (M-cell), with their typical large cell body and processes, were the most characteristic feature in Molly fish spinal cord, where it presented synaptic connections with astrocytes and their ends contained synaptic vesicles indicating an electrical transmission in the M-cells endings. Two types of oligodendrocytes could be recognized; both reacted intensely with Oligodendrocyte transcription factor 2 (Olig2). The proliferative neurons of the substantia grisea centralis expressed myostatin, Nrf2, and Sox9. The findings of this study suggest that molly fish possess highly developed structural features conducive to spinal cord regeneration. Consequently, they could be deemed an exemplary model for investigating central nervous system regeneration.
Collapse
Affiliation(s)
- Mahmoud Awad
- Department of Histology, Faculty of Veterinary Medicine, South Valley University, Qena, Egypt
| | - Ramy K A Sayed
- Department of Histology, Faculty of Veterinary Medicine, Sohag University, Sohag, Egypt
| | - Dalia Mohammadin
- Department of Histology, Faculty of Veterinary Medicine, South Valley University, Qena, Egypt
| | - Marwa M Hussein
- Department of Cell and Tissues, Faculty of Veterinary Medicine, Assiut University, Assiut, Egypt
| | - Doaa M Mokhtar
- Department of Cell and Tissues, Faculty of Veterinary Medicine, Assiut University, Assiut, Egypt
- Department of Histology and Anatomy, School of Veterinary Medicine, Badr University in Assiut, Assiut, Egypt
| |
Collapse
|
2
|
Karuppasamy M, English KG, Henry CA, Manzini MC, Parant JM, Wright MA, Ruparelia AA, Currie PD, Gupta VA, Dowling JJ, Maves L, Alexander MS. Standardization of zebrafish drug testing parameters for muscle diseases. Dis Model Mech 2024; 17:dmm050339. [PMID: 38235578 PMCID: PMC10820820 DOI: 10.1242/dmm.050339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2023] [Accepted: 12/06/2023] [Indexed: 01/19/2024] Open
Abstract
Skeletal muscular diseases predominantly affect skeletal and cardiac muscle, resulting in muscle weakness, impaired respiratory function and decreased lifespan. These harmful outcomes lead to poor health-related quality of life and carry a high healthcare economic burden. The absence of promising treatments and new therapies for muscular disorders requires new methods for candidate drug identification and advancement in animal models. Consequently, the rapid screening of drug compounds in an animal model that mimics features of human muscle disease is warranted. Zebrafish are a versatile model in preclinical studies that support developmental biology and drug discovery programs for novel chemical entities and repurposing of established drugs. Due to several advantages, there is an increasing number of applications of the zebrafish model for high-throughput drug screening for human disorders and developmental studies. Consequently, standardization of key drug screening parameters, such as animal husbandry protocols, drug compound administration and outcome measures, is paramount for the continued advancement of the model and field. Here, we seek to summarize and explore critical drug treatment and drug screening parameters in the zebrafish-based modeling of human muscle diseases. Through improved standardization and harmonization of drug screening parameters and protocols, we aim to promote more effective drug discovery programs.
Collapse
Affiliation(s)
- Muthukumar Karuppasamy
- Division of Neurology, Department of Pediatrics, University of Alabama at Birmingham and Children's of Alabama, Birmingham, AL 35294, USA
| | - Katherine G. English
- Division of Neurology, Department of Pediatrics, University of Alabama at Birmingham and Children's of Alabama, Birmingham, AL 35294, USA
| | - Clarissa A. Henry
- Graduate School of Biomedical Science and Engineering, University of Maine, Orono, ME 04469, USA
- School of Biology and Ecology, University of Maine, Orono, ME 04469, USA
| | - M. Chiara Manzini
- Child Health Institute of New Jersey and Department of Neuroscience and Cell Biology, Rutgers, Robert Wood Johnson Medical School, New Brunswick, NJ 08901, USA
| | - John M. Parant
- Department of Pharmacology and Toxicology, University of Alabama at Birmingham Heersink School of Medicine, Birmingham, AL 35294, USA
| | - Melissa A. Wright
- Department of Pediatrics, Section of Child Neurology, University of Colorado at Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Avnika A. Ruparelia
- Department of Anatomy and Physiology, School of Biomedical Sciences, Faculty of Medicine Dentistry and Health Sciences, University of Melbourne, Melbourne, Victoria 3010, Australia
- Centre for Muscle Research, Department of Anatomy and Physiology, University of Melbourne, Melbourne, Victoria 3010, Australia
- Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria 3800, Australia
| | - Peter D. Currie
- Centre for Muscle Research, Department of Anatomy and Physiology, University of Melbourne, Melbourne, Victoria 3010, Australia
- Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria 3800, Australia
- EMBL Australia, Victorian Node, Monash University, Clayton, Victoria 3800, Australia
| | - Vandana A. Gupta
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - James J. Dowling
- Division of Neurology, The Hospital for Sick Children, Toronto, Ontario M5G 1X8, Canada
- Department of Paediatrics, University of Toronto, Toronto, Ontario M5G 1X8, Canada
- Program for Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Ontario M5G 0A4, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario M5G 0A4, Canada
| | - Lisa Maves
- Center for Developmental Biology and Regenerative Medicine, Seattle Children's Research Institute, Seattle, WA 98101, USA
- Department of Pediatrics, University of Washington, Seattle, WA 98195, USA
| | - Matthew S. Alexander
- Division of Neurology, Department of Pediatrics, University of Alabama at Birmingham and Children's of Alabama, Birmingham, AL 35294, USA
- UAB Center for Exercise Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
- Department of Genetics, University of Alabama at Birmingham, Birmingham, AL 35294, USA
- Civitan International Research Center, University of Alabama at Birmingham, Birmingham, AL 35294, USA
- UAB Center for Neurodegeneration and Experimental Therapeutics (CNET), Birmingham, AL 35294, USA
| |
Collapse
|
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
|
Cigliola V, Shoffner A, Lee N, Ou J, Gonzalez TJ, Hoque J, Becker CJ, Han Y, Shen G, Faw TD, Abd-El-Barr MM, Varghese S, Asokan A, Poss KD. Spinal cord repair is modulated by the neurogenic factor Hb-egf under direction of a regeneration-associated enhancer. Nat Commun 2023; 14:4857. [PMID: 37567873 PMCID: PMC10421883 DOI: 10.1038/s41467-023-40486-5] [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: 06/27/2023] [Accepted: 07/26/2023] [Indexed: 08/13/2023] Open
Abstract
Unlike adult mammals, zebrafish regenerate spinal cord tissue and recover locomotor ability after a paralyzing injury. Here, we find that ependymal cells in zebrafish spinal cords produce the neurogenic factor Hb-egfa upon transection injury. Animals with hb-egfa mutations display defective swim capacity, axon crossing, and tissue bridging after spinal cord transection, associated with disrupted indicators of neuron production. Local recombinant human HB-EGF delivery alters ependymal cell cycling and tissue bridging, enhancing functional regeneration. Epigenetic profiling reveals a tissue regeneration enhancer element (TREE) linked to hb-egfa that directs gene expression in spinal cord injuries. Systemically delivered recombinant AAVs containing this zebrafish TREE target gene expression to crush injuries of neonatal, but not adult, murine spinal cords. Moreover, enhancer-based HB-EGF delivery by AAV administration improves axon densities after crush injury in neonatal cords. Our results identify Hb-egf as a neurogenic factor necessary for innate spinal cord regeneration and suggest strategies to improve spinal cord repair in mammals.
Collapse
Affiliation(s)
- Valentina Cigliola
- Duke Regeneration Center, Duke University, Durham, NC, USA
- Department of Cell Biology, Duke University Medical Center, Durham, NC, USA
- Université Côte d'Azur, Inserm, CNRS, Institut de Biologie Valrose, Nice, France
| | - Adam Shoffner
- Duke Regeneration Center, Duke University, Durham, NC, USA
- Department of Cell Biology, Duke University Medical Center, Durham, NC, USA
- Department of Surgery, Duke University Medical Center, Durham, NC, USA
| | - Nutishia Lee
- Duke Regeneration Center, Duke University, Durham, NC, USA
- Department of Cell Biology, Duke University Medical Center, Durham, NC, USA
| | - Jianhong Ou
- Duke Regeneration Center, Duke University, Durham, NC, USA
- Department of Cell Biology, Duke University Medical Center, Durham, NC, USA
| | - Trevor J Gonzalez
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC, USA
| | - Jiaul Hoque
- Department of Orthopedic Surgery, Duke University School of Medicine, Durham, NC, USA
| | - Clayton J Becker
- Duke Regeneration Center, Duke University, Durham, NC, USA
- Department of Cell Biology, Duke University Medical Center, Durham, NC, USA
| | - Yanchao Han
- Department of Cardiovascular Surgery of the First Affiliated Hospital and Institute for Cardiovascular Science, Soochow University, Suzhou, Jiangsu, China
| | - Grace Shen
- Duke Regeneration Center, Duke University, Durham, NC, USA
- Department of Cell Biology, Duke University Medical Center, Durham, NC, USA
| | - Timothy D Faw
- Duke Regeneration Center, Duke University, Durham, NC, USA
- Department of Orthopedic Surgery, Duke University School of Medicine, Durham, NC, USA
- Duke Institute for Brain Sciences, Duke University, Durham, NC, USA
| | | | - Shyni Varghese
- Department of Orthopedic Surgery, Duke University School of Medicine, Durham, NC, USA
- Department of Mechanical Engineering and Materials Science, Duke University, Durham, NC, USA
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Aravind Asokan
- Duke Regeneration Center, Duke University, Durham, NC, USA
- Department of Surgery, Duke University Medical Center, Durham, NC, USA
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC, USA
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Kenneth D Poss
- Duke Regeneration Center, Duke University, Durham, NC, USA.
- Department of Cell Biology, Duke University Medical Center, Durham, NC, USA.
| |
Collapse
|
5
|
Adler J. The Axon Initial Segment Plays a Dynamic Role in Peripheral Motor Neuron Synapse Regeneration following Injury. J Neurosci 2023; 43:3199-3201. [PMID: 37137706 PMCID: PMC10162451 DOI: 10.1523/jneurosci.2326-22.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 03/16/2023] [Accepted: 03/21/2023] [Indexed: 05/05/2023] Open
Affiliation(s)
- Joy Adler
- Anschutz Medical Neuroscience Graduate Training Program, University of Colorado, Aurora, Colorado 80045
| |
Collapse
|
6
|
Oprişoreanu AM, Ryan F, Richmond C, Dzekhtsiarova Y, Carragher NO, Becker T, David S, Becker CG. Drug screening in zebrafish larvae reveals inflammation-related modulators of secondary damage after spinal cord injury in mice. Theranostics 2023; 13:2531-2551. [PMID: 37215570 PMCID: PMC10196818 DOI: 10.7150/thno.81332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Accepted: 04/11/2023] [Indexed: 05/24/2023] Open
Abstract
Prolonged inflammation after spinal cord injury is detrimental to recovery. To find pharmacological modulators of the inflammation response, we designed a rapid drug screening paradigm in larval zebrafish followed by testing of hit compounds in a mouse spinal cord injury model. Methods: We used reduced il-1β linked green fluorescent protein (GFP) reporter gene expression as a read-out for reduced inflammation in a screen of 1081 compounds in larval zebrafish. Hit drugs were tested in a moderate contusion model in mice for cytokine regulation, and improved tissue preservation and locomotor recovery. Results: Three compounds robustly reduced il-1β expression in zebrafish. Cimetidine, an over-the-counter H2 receptor antagonist, also reduced the number of pro-inflammatory neutrophils and rescued recovery after injury in a zebrafish mutant with prolonged inflammation. Cimetidine action on il-1β expression levels was abolished by somatic mutation of H2 receptor hrh2b, suggesting specific action. In mice, systemic treatment with Cimetidine led to significantly improved recovery of locomotor behavior as compared to controls, accompanied by decreased neuronal tissue loss and a shift towards a pro-regenerative profile of cytokine gene expression. Conclusion: Our screen revealed H2 receptor signaling as a promising target for future therapeutic interventions in spinal cord injury. This work highlights the usefulness of the zebrafish model for rapid screening of drug libraries to identify therapeutics to treat mammalian spinal cord injury.
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
- Center for Regenerative Therapies Dresden, TU Dresden, Fetscherstraße 105, 01307 Dresden, Germany
| | - Fari Ryan
- Centre for Research in Neuroscience, Research Institute of the McGill University Health Centre, 1650 Cedar Ave., Montreal, Quebec, H3G 1A4
| | - Claire Richmond
- Centre for Discovery Brain Sciences, University of Edinburgh, The Chancellor's Building, 49 Little France Crescent, Edinburgh, EH16 4SB, UK
| | - Yuliya Dzekhtsiarova
- Center for Regenerative Therapies Dresden, TU Dresden, Fetscherstraße 105, 01307 Dresden, Germany
| | - Neil O. Carragher
- Cancer Research UK Edinburgh Centre, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, EH4 2XR, UK
| | - Thomas Becker
- Centre for Discovery Brain Sciences, University of Edinburgh, The Chancellor's Building, 49 Little France Crescent, Edinburgh, EH16 4SB, UK
- Center for Regenerative Therapies Dresden, TU Dresden, Fetscherstraße 105, 01307 Dresden, Germany
| | - Samuel David
- Centre for Research in Neuroscience, Research Institute of the McGill University Health Centre, 1650 Cedar Ave., Montreal, Quebec, H3G 1A4
| | - Catherina G. Becker
- Centre for Discovery Brain Sciences, University of Edinburgh, The Chancellor's Building, 49 Little France Crescent, Edinburgh, EH16 4SB, UK
- Center for Regenerative Therapies Dresden, TU Dresden, Fetscherstraße 105, 01307 Dresden, Germany
| |
Collapse
|
7
|
Zhu P, Zheng P, Kong X, Wang S, Cao M, Zhao C. Rassf7a promotes spinal cord regeneration and controls spindle orientation in neural progenitor cells. EMBO Rep 2023; 24:e54984. [PMID: 36408859 PMCID: PMC9827555 DOI: 10.15252/embr.202254984] [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: 03/06/2022] [Revised: 10/24/2022] [Accepted: 10/27/2022] [Indexed: 11/22/2022] Open
Abstract
Spinal cord injury (SCI) can cause long-lasting disability in mammals due to the lack of axonal regrowth together with the inability to reinitiate spinal neurogenesis at the injury site. Deciphering the mechanisms that regulate the proliferation and differentiation of neural progenitor cells is critical for understanding spinal neurogenesis after injury. Compared with mammals, zebrafish show a remarkable capability of spinal cord regeneration. Here, we show that Rassf7a, a member of the Ras-association domain family, promotes spinal cord regeneration after injury. Zebrafish larvae harboring a rassf7a mutation show spinal cord regeneration and spinal neurogenesis defects. Live imaging shows abnormal asymmetric neurogenic divisions and spindle orientation defects in mutant neural progenitor cells. In line with this, the expression of rassf7a is enriched in neural progenitor cells. Subcellular analysis shows that Rassf7a localizes to the centrosome and is essential for cell cycle progression. Our data indicate a role for Rassf7a in modulating spindle orientation and the proliferation of neural progenitor cells after spinal cord injury.
Collapse
Affiliation(s)
- Panpan Zhu
- Institute of Evolution and Marine BiodiversityOcean University of ChinaQingdaoChina
- Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and TechnologyQingdaoChina
- Sars‐Fang Centre, Ministry of Education Key Laboratory of Marine Genetics and Breeding, College of Marine Life SciencesOcean University of ChinaQingdaoChina
| | - Pengfei Zheng
- Institute of Evolution and Marine BiodiversityOcean University of ChinaQingdaoChina
| | - Xinlong Kong
- Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Department of PathophysiologyShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Shuo Wang
- Institute of Evolution and Marine BiodiversityOcean University of ChinaQingdaoChina
| | - Muqing Cao
- Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Department of PathophysiologyShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Chengtian Zhao
- Institute of Evolution and Marine BiodiversityOcean University of ChinaQingdaoChina
- Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and TechnologyQingdaoChina
- Sars‐Fang Centre, Ministry of Education Key Laboratory of Marine Genetics and Breeding, College of Marine Life SciencesOcean University of ChinaQingdaoChina
| |
Collapse
|
8
|
Assaying Optic Nerve Regeneration in Larval Zebrafish. Methods Mol Biol 2023; 2636:191-203. [PMID: 36881301 DOI: 10.1007/978-1-0716-3012-9_10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/08/2023]
Abstract
Zebrafish have a remarkable capacity for spontaneously regenerating their central nervous system. Larval zebrafish are optically transparent and therefore are widely used to dynamically visualize cellular processes in vivo, such as nerve regeneration. Regeneration of retinal ganglion cell (RGC) axons within the optic nerve has been previously studied in adult zebrafish. In contrast, assays of optic nerve regeneration have previously not been established in larval zebrafish. In order to take advantage of the imaging capabilities in the larval zebrafish model, we recently developed an assay to physically transect RGC axons and monitor optic nerve regeneration in larval zebrafish. We found that RGC axons rapidly and robustly regrow to the optic tectum. Here, we describe the methods for performing the optic nerve transections, as well as methods for visualizing RGC regeneration in larval zebrafish.
Collapse
|
9
|
Ren DL, Hu B, Shao GJ, Wang XL, Wei ML. DUSP2 deletion with CRISPR/Cas9 promotes Mauthner cell axonal regeneration at the early stage of zebrafish. Neural Regen Res 2023; 18:577-581. [DOI: 10.4103/1673-5374.350208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
10
|
Saettele AL, Wong HTC, Kindt KS, Warchol ME, Sheets L. Prolonged Dexamethasone Exposure Enhances Zebrafish Lateral-Line Regeneration But Disrupts Mitochondrial Homeostasis and Hair Cell Function. J Assoc Res Otolaryngol 2022; 23:683-700. [PMID: 36261670 PMCID: PMC9789251 DOI: 10.1007/s10162-022-00875-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Accepted: 09/20/2022] [Indexed: 01/06/2023] Open
Abstract
The synthetic glucocorticoid dexamethasone is commonly used to treat inner ear disorders. Previous work in larval zebrafish has shown that dexamethasone treatment enhances hair cell regeneration, yet dexamethasone has also been shown to inhibit regeneration of peripheral nerves after lesion. We therefore used the zebrafish model to determine the impact of dexamethasone treatment on lateral-line hair cells and primary afferents. To explore dexamethasone in the context of regeneration, we used copper sulfate (CuSO4) to induce hair cell loss and retraction of nerve terminals, and then allowed animals to recover in dexamethasone for 48 h. Consistent with previous work, we observed significantly more regenerated hair cells in dexamethasone-treated larvae. Importantly, we found that the afferent processes beneath neuromasts also regenerated in the presence of dexamethasone and formed an appropriate number of synapses, indicating that innervation of hair cells was not inhibited by dexamethasone. In addition to regeneration, we also explored the effects of prolonged dexamethasone exposure on lateral-line homeostasis and function. Following dexamethasone treatment, we observed hyperpolarized mitochondrial membrane potentials (ΔΨm) in neuromast hair cells and supporting cells. Hair cells exposed to dexamethasone were also more vulnerable to neomycin-induced cell death. In response to a fluid-jet delivered saturating stimulus, calcium influx through hair cell mechanotransduction channels was significantly reduced, yet presynaptic calcium influx was unchanged. Cumulatively, these observations indicate that dexamethasone enhances hair cell regeneration in lateral-line neuromasts, yet also disrupts mitochondrial homeostasis, making hair cells more vulnerable to ototoxic insults and possibly impacting hair cell function.
Collapse
Affiliation(s)
- Allison L Saettele
- Department of Otolaryngology, Washington University School of Medicine, St. Louis, MO, USA
| | - Hiu-Tung C Wong
- Section On Sensory Cell Development and Function, National Institutes On Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, USA
| | - Katie S Kindt
- Section On Sensory Cell Development and Function, National Institutes On Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, USA
| | - Mark E Warchol
- Department of Otolaryngology, Washington University School of Medicine, St. Louis, MO, USA
- Department of Neuroscience, Washington University School of Medicine, St. Louis, MO, USA
| | - Lavinia Sheets
- Department of Otolaryngology, Washington University School of Medicine, St. Louis, MO, USA.
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO, USA.
| |
Collapse
|
11
|
Hossainian D, Shao E, Jiao B, Ilin VA, Parris RS, Zhou Y, Bai Q, Burton EA. Quantification of functional recovery in a larval zebrafish model of spinal cord injury. J Neurosci Res 2022; 100:2044-2054. [PMID: 35986577 PMCID: PMC10695274 DOI: 10.1002/jnr.25118] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 07/19/2022] [Accepted: 08/01/2022] [Indexed: 11/12/2023]
Abstract
Human spinal cord injury (SCI) is characterized by permanent loss of damaged axons, resulting in chronic disability. In contrast, zebrafish can regenerate axonal projections following central nervous system injury and re-establish synaptic contacts with distant targets; elucidation of the underlying molecular events is an important goal with translational potential for improving outcomes in SCI patients. We generated transgenic zebrafish with GFP-labeled axons and transected their spinal cords at 10 days post-fertilization. Intravital confocal microscopy revealed robust axonal regeneration following the procedure, with abundant axons bridging the transection site by 48 h post-injury. In order to analyze neurological function in this model, we developed and validated new open-source software to measure zebrafish lateral trunk curvature during propulsive and turning movements at high temporal resolution. Immediately following spinal cord transection, axial movements were dramatically decreased caudal to the lesion site, but preserved rostral to the injury, suggesting the induction of motor paralysis below the transection level. Over the subsequent 96 h, the magnitude of movements caudal to the lesion recovered to baseline, but the rate of change of truncal curvature did not fully recover, suggesting incomplete restoration of caudal strength over this time course. Quantification of both morphological and functional recovery following SCI will be important for the analysis of axonal regeneration and downstream events necessary for restoration of motor function. An extensive array of genetic and pharmacological interventions can be deployed in the larval zebrafish model to investigate the underlying molecular mechanisms.
Collapse
Affiliation(s)
- Darius Hossainian
- Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA
- Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA
| | - Enhua Shao
- Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA
- Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA
- Tsinghua University Medical School, Beijing, China
| | - Binxuan Jiao
- Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA
- Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA
- Tsinghua University Medical School, Beijing, China
| | - Vladimir A. Ilin
- Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA
- Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA
| | - Ritika S. Parris
- Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA
- Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA
| | - Yangzhong Zhou
- Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA
- Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA
- Tsinghua University Medical School, Beijing, China
| | - Qing Bai
- Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA
- Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA
| | - Edward A. Burton
- Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA
- Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA
- Geriatric Research, Education and Clinical Center, Pittsburgh VA Healthcare System, Pittsburgh, PA, 15213, USA
| |
Collapse
|
12
|
Alper SR, Dorsky RI. Unique advantages of zebrafish larvae as a model for spinal cord regeneration. Front Mol Neurosci 2022; 15:983336. [PMID: 36157068 PMCID: PMC9489991 DOI: 10.3389/fnmol.2022.983336] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Accepted: 08/18/2022] [Indexed: 11/30/2022] Open
Abstract
The regenerative capacity of the spinal cord in mammals ends at birth. In contrast, teleost fish and amphibians retain this capacity throughout life, leading to the use of the powerful zebrafish model system to identify novel mechanisms that promote spinal cord regeneration. While adult zebrafish offer an effective comparison with non-regenerating mammals, they lack the complete array of experimental approaches that have made this animal model so successful. In contrast, the optical transparency, simple anatomy and complex behavior of zebrafish larvae, combined with the known conservation of pro-regenerative signals and cell types between larval and adult stages, suggest that they may hold even more promise as a system for investigating spinal cord regeneration. In this review, we highlight characteristics and advantages of the larval model that underlie its potential to provide future therapeutic approaches for treating human spinal cord injury.
Collapse
|
13
|
Iribarne M, Hyde DR. Different inflammation responses modulate Müller glia proliferation in the acute or chronically damaged zebrafish retina. Front Cell Dev Biol 2022; 10:892271. [PMID: 36120571 PMCID: PMC9472244 DOI: 10.3389/fcell.2022.892271] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Accepted: 07/20/2022] [Indexed: 11/13/2022] Open
Abstract
Unlike mammals, zebrafish regenerate in response to retinal damage. Because microglia are activated by retinal damage, we investigated their role during regeneration following either acute or chronic damage. At three weeks post-fertilization (wpf), both wild-type fish exhibiting NMDA-induced acute ganglion and amacrine cell death and gold rush (gosh) mutant fish possessing chronic cone photoreceptor degeneration displayed reactive microglia/macrophages and Müller glia proliferation. Dexamethasone-treated retinas, to inhibit the immune response, lacked reactive microglia/macrophages and possessed fewer PCNA-positive cells, while LPS treatment increased microglia/macrophages and PCNA-labeled cells. NMDA-injured retinas upregulated expression of il-1β and tnfα pro-inflammatory cytokine genes, followed by increased expression of il-10 and arg1 anti-inflammatory/remodeling cytokine genes. A transient early TNFα pro-inflammatory microglia/macrophage population was visualized in NMDA-damaged retinas. In contrast, gosh mutant retinas exhibited a slight increase of pro-inflammatory cytokine gene expression concurrently with a greater increased anti-inflammatory/remodeling cytokine gene expression. Few TNFα pro-inflammatory microglia/macrophages were observed in the gosh retina. Understanding why acute and chronic damage results in different inflammation profiles and their effects on regulating zebrafish retinal regeneration would provide important clues toward improving therapeutic strategies for repairing injured mammalian tissues.
Collapse
Affiliation(s)
- Maria Iribarne
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN, United States
- Center for Zebrafish Research, University of Notre Dame, Notre Dame, IN, United States
- Center for Stem Cells and Regenerative Medicine, University of Notre Dame, Notre Dame, IN, United States
| | - David R. Hyde
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN, United States
- Center for Zebrafish Research, University of Notre Dame, Notre Dame, IN, United States
- Center for Stem Cells and Regenerative Medicine, University of Notre Dame, Notre Dame, IN, United States
- *Correspondence: David R. Hyde,
| |
Collapse
|
14
|
Green LA, O'Dea MR, Hoover CA, DeSantis DF, Smith CJ. The embryonic zebrafish brain is seeded by a lymphatic-dependent population of mrc1 + microglia precursors. Nat Neurosci 2022; 25:849-864. [PMID: 35710983 PMCID: PMC10680068 DOI: 10.1038/s41593-022-01091-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Accepted: 05/06/2022] [Indexed: 02/02/2023]
Abstract
Microglia are the resident macrophages of the CNS that serve critical roles in brain construction. Although human brains contain microglia by 4 weeks gestation, an understanding of the earliest microglia that seed the brain during its development remains unresolved. Using time-lapse imaging in zebrafish, we discovered a mrc1a+ microglia precursor population that seeds the brain before traditionally described microglia. These early microglia precursors are dependent on lymphatic vasculature that surrounds the brain and are independent of pu1+ yolk sac-derived microglia. Single-cell RNA-sequencing datasets reveal Mrc1+ microglia in the embryonic brains of mice and humans. We then show in zebrafish that these early mrc1a+ microglia precursors preferentially expand during pathophysiological states in development. Taken together, our results identify a critical role of lymphatics in the microglia precursors that seed the early embryonic brain.
Collapse
Affiliation(s)
- Lauren A Green
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN, USA
- The Center for Stem Cells and Regenerative Medicine, University of Notre Dame, Notre Dame, IN, USA
| | - Michael R O'Dea
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN, USA
| | - Camden A Hoover
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN, USA
- The Center for Stem Cells and Regenerative Medicine, University of Notre Dame, Notre Dame, IN, USA
| | - Dana F DeSantis
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN, USA
- The Center for Stem Cells and Regenerative Medicine, University of Notre Dame, Notre Dame, IN, USA
| | - Cody J Smith
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN, USA.
- The Center for Stem Cells and Regenerative Medicine, University of Notre Dame, Notre Dame, IN, USA.
| |
Collapse
|
15
|
Könemann S, von Wyl M, Vom Berg C. Zebrafish Larvae Rapidly Recover from Locomotor Effects and Neuromuscular Alterations Induced by Cholinergic Insecticides. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2022; 56:8449-8462. [PMID: 35575681 DOI: 10.1021/acs.est.2c00161] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
Owing to the importance of acetylcholine as a neurotransmitter, many insecticides target the cholinergic system. Across phyla, cholinergic signaling is essential for many neuro-developmental processes including axonal pathfinding and synaptogenesis. Consequently, early-life exposure to such insecticides can disturb these processes, resulting in an impaired nervous system. One test frequently used to assess developmental neurotoxicity is the zebrafish light-dark transition test, which measures larval locomotion as a response to light changes. However, it is only poorly understood which structural alterations cause insecticide-induced locomotion defects and how persistent these alterations are. Therefore, this study aimed to link locomotion defects with effects on neuromuscular structures, including motorneurons, synapses, and muscles, and to investigate the longevity of the effects. The cholinergic insecticides diazinon and dimethoate (organophosphates), methomyl and pirimicarb (carbamates), and imidacloprid and thiacloprid (neonicotinoids) were used to induce hypoactivity. Our analyses revealed that some insecticides did not alter any of the structures assessed, while others affected axon branching (methomyl, imidacloprid) or muscle integrity (methomyl, thiacloprid). The majority of effects, even structural, were reversible within 24 to 72 h. Overall, we find that both neurodevelopmental and non-neurodevelopmental effects of different longevity can account for the reduced locomotion. These findings provide unprecedented insights into the underpinnings of insecticide-induced hypoactivity.
Collapse
Affiliation(s)
- Sarah Könemann
- Department of Environmental Toxicology, Eawag, Überlandstrasse 133, 8600 Dübendorf, Switzerland
- École Polytechnique Fédéral de Lausanne, EPFL, Route Cantonale, 1015 Lausanne, Switzerland
| | - Melissa von Wyl
- Department of Environmental Toxicology, Eawag, Überlandstrasse 133, 8600 Dübendorf, Switzerland
- University of Zurich, UZH, Rämistrassse 71, 8006 Zurich, Switzerland
| | - Colette Vom Berg
- Department of Environmental Toxicology, Eawag, Überlandstrasse 133, 8600 Dübendorf, Switzerland
| |
Collapse
|
16
|
Becker T, Becker CG. Regenerative neurogenesis: the integration of developmental, physiological and immune signals. Development 2022; 149:275248. [PMID: 35502778 PMCID: PMC9124576 DOI: 10.1242/dev.199907] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
In fishes and salamanders, but not mammals, neural stem cells switch back to neurogenesis after injury. The signalling environment of neural stem cells is strongly altered by the presence of damaged cells and an influx of immune, as well as other, cells. Here, we summarise our recently expanded knowledge of developmental, physiological and immune signals that act on neural stem cells in the zebrafish central nervous system to directly, or indirectly, influence their neurogenic state. These signals act on several intracellular pathways, which leads to changes in chromatin accessibility and gene expression, ultimately resulting in regenerative neurogenesis. Translational approaches in non-regenerating mammals indicate that central nervous system stem cells can be reprogrammed for neurogenesis. Understanding signalling mechanisms in naturally regenerating species show the path to experimentally promoting neurogenesis in mammals.
Collapse
Affiliation(s)
- Thomas Becker
- Center for Regenerative Therapies at the TU Dresden, Technische Universität Dresden, 01307 Dresden, Germany.,Centre for Discovery Brain Sciences, University of Edinburgh Medical School, Biomedical Science, Edinburgh, EH16 4SB, Scotland
| | - Catherina G Becker
- Center for Regenerative Therapies at the TU Dresden, Technische Universität Dresden, 01307 Dresden, Germany.,Centre for Discovery Brain Sciences, University of Edinburgh Medical School, Biomedical Science, Edinburgh, EH16 4SB, Scotland
| |
Collapse
|
17
|
Geurtzen K, López-Delgado AC, Duseja A, Kurzyukova A, Knopf F. Laser-mediated osteoblast ablation triggers a pro-osteogenic inflammatory response regulated by reactive oxygen species and glucocorticoid signaling in zebrafish. Development 2022; 149:275194. [DOI: 10.1242/dev.199803] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Accepted: 02/22/2022] [Indexed: 12/31/2022]
Abstract
ABSTRACT
In zebrafish, transgenic labeling approaches, robust regenerative responses and excellent in vivo imaging conditions enable precise characterization of immune cell behavior in response to injury. Here, we monitored osteoblast-immune cell interactions in bone, a tissue which is particularly difficult to in vivo image in tetrapod species. Ablation of individual osteoblasts leads to recruitment of neutrophils and macrophages in varying numbers, depending on the extent of the initial insult, and initiates generation of cathepsin K+ osteoclasts from macrophages. Osteoblast ablation triggers the production of pro-inflammatory cytokines and reactive oxygen species, which are needed for successful macrophage recruitment. Excess glucocorticoid signaling as it occurs during the stress response inhibits macrophage recruitment, maximum speed and changes the macrophage phenotype. Although osteoblast loss is compensated for within a day by contribution of committed osteoblasts, macrophages continue to populate the region. Their presence is required for osteoblasts to fill the lesion site. Our model enables visualization of bone repair after microlesions at single-cell resolution and demonstrates a pro-osteogenic function of tissue-resident macrophages in non-mammalian vertebrates.
Collapse
Affiliation(s)
- Karina Geurtzen
- Center for Regenerative Therapies TU Dresden (CRTD), Center for Molecular and Cellular Bioengineering (CMCB), TU Dresden, 01307 Dresden, Germany
- Center for Healthy Aging, Medical Faculty Carl Gustav Carus, TU Dresden, 01307 Dresden, Germany
- Laboratory of Clinical and Experimental Endocrinology, Department of Chronic Diseases, Metabolism and Ageing, KU Leuven, 3000 Leuven, Belgium
| | - Alejandra Cristina López-Delgado
- Center for Regenerative Therapies TU Dresden (CRTD), Center for Molecular and Cellular Bioengineering (CMCB), TU Dresden, 01307 Dresden, Germany
- Center for Healthy Aging, Medical Faculty Carl Gustav Carus, TU Dresden, 01307 Dresden, Germany
| | - Ankita Duseja
- Center for Regenerative Therapies TU Dresden (CRTD), Center for Molecular and Cellular Bioengineering (CMCB), TU Dresden, 01307 Dresden, Germany
- Center for Healthy Aging, Medical Faculty Carl Gustav Carus, TU Dresden, 01307 Dresden, Germany
- Department of Oncology and Metabolism, Metabolic Bone Centre, Sorby Wing, Northern General Hospital, Sheffield S5 7AU, UK
| | - Anastasia Kurzyukova
- Center for Regenerative Therapies TU Dresden (CRTD), Center for Molecular and Cellular Bioengineering (CMCB), TU Dresden, 01307 Dresden, Germany
- Center for Healthy Aging, Medical Faculty Carl Gustav Carus, TU Dresden, 01307 Dresden, Germany
- Faculty of Health and Medical Sciences, Biotech Research & Innovation Centre (BRIC), University of Copenhagen, 2200 Copenhagen, Denmark
| | - Franziska Knopf
- Center for Regenerative Therapies TU Dresden (CRTD), Center for Molecular and Cellular Bioengineering (CMCB), TU Dresden, 01307 Dresden, Germany
- Center for Healthy Aging, Medical Faculty Carl Gustav Carus, TU Dresden, 01307 Dresden, Germany
| |
Collapse
|
18
|
Riley SE, Feng Y, Hansen CG. Hippo-Yap/Taz signalling in zebrafish regeneration. NPJ Regen Med 2022; 7:9. [PMID: 35087046 PMCID: PMC8795407 DOI: 10.1038/s41536-022-00209-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Accepted: 12/14/2021] [Indexed: 12/29/2022] Open
Abstract
The extent of tissue regeneration varies widely between species. Mammals have a limited regenerative capacity whilst lower vertebrates such as the zebrafish (Danio rerio), a freshwater teleost, can robustly regenerate a range of tissues, including the spinal cord, heart, and fin. The molecular and cellular basis of this altered response is one of intense investigation. In this review, we summarise the current understanding of the association between zebrafish regeneration and Hippo pathway function, a phosphorylation cascade that regulates cell proliferation, mechanotransduction, stem cell fate, and tumorigenesis, amongst others. We also compare this function to Hippo pathway activity in the regenerative response of other species. We find that the Hippo pathway effectors Yap/Taz facilitate zebrafish regeneration and that this appears to be latent in mammals, suggesting that therapeutically promoting precise and temporal YAP/TAZ signalling in humans may enhance regeneration and hence reduce morbidity.
Collapse
Affiliation(s)
- Susanna E Riley
- University of Edinburgh Centre for Inflammation Research, Institute for Regeneration and Repair, Queen's Medical Research Institute, Edinburgh bioQuarter, 47 Little France Crescent, Edinburgh, EH16 4TJ, UK
| | - Yi Feng
- University of Edinburgh Centre for Inflammation Research, Institute for Regeneration and Repair, Queen's Medical Research Institute, Edinburgh bioQuarter, 47 Little France Crescent, Edinburgh, EH16 4TJ, UK
| | - Carsten Gram Hansen
- University of Edinburgh Centre for Inflammation Research, Institute for Regeneration and Repair, Queen's Medical Research Institute, Edinburgh bioQuarter, 47 Little France Crescent, Edinburgh, EH16 4TJ, UK.
| |
Collapse
|
19
|
Perez JC, Gerber YN, Perrin FE. Dynamic Diversity of Glial Response Among Species in Spinal Cord Injury. Front Aging Neurosci 2021; 13:769548. [PMID: 34899275 PMCID: PMC8662749 DOI: 10.3389/fnagi.2021.769548] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Accepted: 10/29/2021] [Indexed: 12/11/2022] Open
Abstract
The glial scar that forms after traumatic spinal cord injury (SCI) is mostly composed of microglia, NG2 glia, and astrocytes and plays dual roles in pathophysiological processes induced by the injury. On one hand, the glial scar acts as a chemical and physical obstacle to spontaneous axonal regeneration, thus preventing functional recovery, and, on the other hand, it partly limits lesion extension. The complex activation pattern of glial cells is associated with cellular and molecular crosstalk and interactions with immune cells. Interestingly, response to SCI is diverse among species: from amphibians and fishes that display rather limited (if any) glial scarring to mammals that exhibit a well-identifiable scar. Additionally, kinetics of glial activation varies among species. In rodents, microglia become activated before astrocytes, and both glial cell populations undergo activation processes reflected amongst others by proliferation and migration toward the injury site. In primates, glial cell activation is delayed as compared to rodents. Here, we compare the spatial and temporal diversity of the glial response, following SCI amongst species. A better understanding of mechanisms underlying glial activation and scar formation is a prerequisite to develop timely glial cell-specific therapeutic strategies that aim to increase functional recovery.
Collapse
Affiliation(s)
| | - Yannick N Gerber
- MMDN, Université de Montpellier, EPHE, INSERM, Montpellier, France
| | - Florence E Perrin
- MMDN, Université de Montpellier, EPHE, INSERM, Montpellier, France.,Institut Universitaire de France (IUF), Paris, France
| |
Collapse
|
20
|
Shen WY, Fu XH, Cai J, Li WC, Fan BY, Pang YL, Zhao CX, Abula M, Kong XH, Yao X, Feng SQ. Identification of key genes involved in recovery from spinal cord injury in adult zebrafish. Neural Regen Res 2021; 17:1334-1342. [PMID: 34782579 PMCID: PMC8643032 DOI: 10.4103/1673-5374.327360] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
Zebrafish are an effective vertebrate model to study the mechanisms underlying recovery after spinal cord injury. The subacute phase after spinal cord injury is critical to the recovery of neurological function, which involves tissue bridging and axon regeneration. In this study, we found that zebrafish spontaneously recovered 44% of their swimming ability within the subacute phase (2 weeks) after spinal cord injury. During this period, we identified 7762 differentially expressed genes in spinal cord tissue: 2950 were up-regulated and 4812 were down-regulated. These differentially expressed genes were primarily concentrated in the biological processes of the respiratory chain, axon regeneration, and cell-component morphogenesis. The genes were also mostly involved in the regulation of metabolic pathways, the cell cycle, and gene-regulation pathways. We verified the gene expression of two differentially expressed genes, clasp2 up-regulation and h1m down-regulation, in zebrafish spinal cord tissue in vitro. Pathway enrichment analysis revealed that up-regulated clasp2 functions similarly to microtubule-associated protein, which is responsible for axon extension regulated by microtubules. Down-regulated h1m controls endogenous stem cell differentiation after spinal cord injury. This study provides new candidate genes, clasp2 and h1m, as potential therapeutic intervention targets for spinal cord injury repair by neuroregeneration. All experimental procedures and protocols were approved by the Animal Ethics Committee of Tianjin Institute of Medical & Pharmaceutical Sciences (approval No. IMPS-EAEP-Q-2019-02) on September 24, 2019.
Collapse
Affiliation(s)
- Wen-Yuan Shen
- International Science and Technology Cooperation Base of Spinal Cord Injury, Tianjin Key Laboratory of Spine and Spinal Cord, Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, China
| | - Xuan-Hao Fu
- International Science and Technology Cooperation Base of Spinal Cord Injury, Tianjin Key Laboratory of Spine and Spinal Cord, Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, China
| | - Jun Cai
- Tianjin Medicine and Health Research Center, Tianjin Institute of Medical & Pharmaceutical Sciences, Tianjin, China
| | - Wen-Chang Li
- Tianjin Medicine and Health Research Center, Tianjin Institute of Medical & Pharmaceutical Sciences, Tianjin, China
| | - Bao-You Fan
- International Science and Technology Cooperation Base of Spinal Cord Injury, Tianjin Key Laboratory of Spine and Spinal Cord, Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, China
| | - Yi-Lin Pang
- International Science and Technology Cooperation Base of Spinal Cord Injury, Tianjin Key Laboratory of Spine and Spinal Cord, Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, China
| | - Chen-Xi Zhao
- International Science and Technology Cooperation Base of Spinal Cord Injury, Tianjin Key Laboratory of Spine and Spinal Cord, Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, China
| | - Muhtidir Abula
- International Science and Technology Cooperation Base of Spinal Cord Injury, Tianjin Key Laboratory of Spine and Spinal Cord, Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, China
| | | | - Xue Yao
- International Science and Technology Cooperation Base of Spinal Cord Injury, Tianjin Key Laboratory of Spine and Spinal Cord, Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, China
| | - Shi-Qing Feng
- International Science and Technology Cooperation Base of Spinal Cord Injury, Tianjin Key Laboratory of Spine and Spinal Cord, Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, China
| |
Collapse
|
21
|
Choe CP, Choi SY, Kee Y, Kim MJ, Kim SH, Lee Y, Park HC, Ro H. Transgenic fluorescent zebrafish lines that have revolutionized biomedical research. Lab Anim Res 2021; 37:26. [PMID: 34496973 PMCID: PMC8424172 DOI: 10.1186/s42826-021-00103-2] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Accepted: 08/26/2021] [Indexed: 12/22/2022] Open
Abstract
Since its debut in the biomedical research fields in 1981, zebrafish have been used as a vertebrate model organism in more than 40,000 biomedical research studies. Especially useful are zebrafish lines expressing fluorescent proteins in a molecule, intracellular organelle, cell or tissue specific manner because they allow the visualization and tracking of molecules, intracellular organelles, cells or tissues of interest in real time and in vivo. In this review, we summarize representative transgenic fluorescent zebrafish lines that have revolutionized biomedical research on signal transduction, the craniofacial skeletal system, the hematopoietic system, the nervous system, the urogenital system, the digestive system and intracellular organelles.
Collapse
Affiliation(s)
- Chong Pyo Choe
- Division of Life Science, Gyeongsang National University, Jinju, 52828, Republic of Korea.,Division of Applied Life Science, Plant Molecular Biology and Biotechnology Research Center, Gyeongsang National University, Jinju, 52828, Republic of Korea
| | - Seok-Yong Choi
- Department of Biomedical Sciences, Chonnam National University Medical School, Hwasun, 58128, Republic of Korea
| | - Yun Kee
- Division of Biomedical Convergence, College of Biomedical Science, Kangwon National University, Chuncheon, 24341, Republic of Korea.
| | - Min Jung Kim
- Department of Biological Sciences, Sookmyung Women's University, Seoul, 04310, Republic of Korea
| | - Seok-Hyung Kim
- Department of Marine Life Sciences and Fish Vaccine Research Center, Jeju National University, Jeju, 63243, Republic of Korea
| | - Yoonsung Lee
- Center for Genomic Integrity, Institute for Basic Science (IBS), Ulsan, 44919, Republic of Korea
| | - Hae-Chul Park
- Department of Biomedical Sciences, College of Medicine, Korea University, Ansan, 15355, Republic of Korea
| | - Hyunju Ro
- Department of Biological Sciences, College of Bioscience and Biotechnology, Chungnam National University, Daejeon, 34134, Republic of Korea
| |
Collapse
|
22
|
Haspel G, Severi KE, Fauci LJ, Cohen N, Tytell ED, Morgan JR. Resilience of neural networks for locomotion. J Physiol 2021; 599:3825-3840. [PMID: 34187088 DOI: 10.1113/jp279214] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Accepted: 06/22/2021] [Indexed: 01/15/2023] Open
Abstract
Locomotion is an essential behaviour for the survival of all animals. The neural circuitry underlying locomotion is therefore highly robust to a wide variety of perturbations, including injury and abrupt changes in the environment. In the short term, fault tolerance in neural networks allows locomotion to persist immediately after mild to moderate injury. In the longer term, in many invertebrates and vertebrates, neural reorganization including anatomical regeneration can restore locomotion after severe perturbations that initially caused paralysis. Despite decades of research, very little is known about the mechanisms underlying locomotor resilience at the level of the underlying neural circuits and coordination of central pattern generators (CPGs). Undulatory locomotion is an ideal behaviour for exploring principles of circuit organization, neural control and resilience of locomotion, offering a number of unique advantages including experimental accessibility and modelling tractability. In comparing three well-characterized undulatory swimmers, lampreys, larval zebrafish and Caenorhabditis elegans, we find similarities in the manifestation of locomotor resilience. To advance our understanding, we propose a comparative approach, integrating experimental and modelling studies, that will allow the field to begin identifying shared and distinct solutions for overcoming perturbations to persist in orchestrating this essential behaviour.
Collapse
Affiliation(s)
- Gal Haspel
- Federated Department of Biological Sciences, New Jersey Institute of Technology, Newark, NJ, 07102, USA
| | - Kristen E Severi
- Federated Department of Biological Sciences, New Jersey Institute of Technology, Newark, NJ, 07102, USA
| | - Lisa J Fauci
- Department of Mathematics, Tulane University, New Orleans, LA, 70118, USA
| | - Netta Cohen
- School of Computing, University of Leeds, Leeds, LS2 9JT, UK
| | - Eric D Tytell
- Department of Biology, Tufts University, Medford, MA, 02155, USA
| | - Jennifer R Morgan
- The Eugene Bell Center for Regenerative Biology and Tissue Engineering, Marine Biological Laboratory, Woods Hole, MA, 02543, USA
| |
Collapse
|
23
|
Tsata V, Wehner D. Know How to Regrow-Axon Regeneration in the Zebrafish Spinal Cord. Cells 2021; 10:cells10061404. [PMID: 34204045 PMCID: PMC8228677 DOI: 10.3390/cells10061404] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 05/25/2021] [Accepted: 06/04/2021] [Indexed: 12/14/2022] Open
Abstract
The capacity for long-distance axon regeneration and functional recovery after spinal cord injury is poor in mammals but remarkable in some vertebrates, including fish and salamanders. The cellular and molecular basis of this interspecies difference is beginning to emerge. This includes the identification of target cells that react to the injury and the cues directing their pro-regenerative responses. Among existing models of successful spinal cord regeneration, the zebrafish is arguably the most understood at a mechanistic level to date. Here, we review the spinal cord injury paradigms used in zebrafish, and summarize the breadth of neuron-intrinsic and -extrinsic factors that have been identified to play pivotal roles in the ability of zebrafish to regenerate central nervous system axons and recover function.
Collapse
Affiliation(s)
- Vasiliki Tsata
- Experimental Surgery, Clinical and Translational Research Center, Biomedical Research Foundation Academy of Athens, 11527 Athens, Greece
- Correspondence: (V.T.); (D.W.)
| | - Daniel Wehner
- Max Planck Institute for the Science of Light, 91058 Erlangen, Germany
- Max-Planck-Zentrum für Physik und Medizin, 91058 Erlangen, Germany
- Correspondence: (V.T.); (D.W.)
| |
Collapse
|
24
|
Li X, Gao Y, Tian F, Du R, Yuan Y, Li P, Liu F, Wang C. miR-31 promotes neural stem cell proliferation and restores motor function after spinal cord injury. Exp Biol Med (Maywood) 2021; 246:1274-1286. [PMID: 33715531 PMCID: PMC8371310 DOI: 10.1177/1535370221997071] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Accepted: 02/01/2021] [Indexed: 01/17/2023] Open
Abstract
This study aims to examine whether miR-31 promotes endogenous NSC proliferation and be used for spinal cord injury management. In the present study, the morpholino knockdown of miR-31 induced abnormal neuronal apoptosis in zebrafish, resulting in impaired development of the tail. miR-31 agomir transfection in NSCs increased Nestin expression and decreased ChAT and GFAP expression levels. miR-31 induced the proliferation of mouse NSCs by upregulating the Notch signaling pathway, and more NSCs entered G1; Notch was inhibited by miR-31 inactivation. Injection of a miR-31 agomir into mouse models of spinal cord injury could effectively restore motor functions after spinal cord injury, which was achieved by promoting the proliferation of endogenous NSCs. After the injection of a miR-31 agomir in spinal cord injury mice, the expression of Nestin and GFAP increased, while GFAP expression decreased. In conclusion, the zebrafish experiments prove that a lack of miR-31 will block nervous system development. In spinal cord injury mouse models, miR-31 overexpression might promote spinal cord injury repair.
Collapse
Affiliation(s)
- Xiao Li
- Laboratory Animal Research Center of Shanxi Medical University, Shanxi Key Laboratory of Animal and Animal Model of Human Diseases, Taiyuan 030001, China
| | - Yuantao Gao
- Queen Mary School, Nanchang University, Nanchang 330000, China
| | - Feng Tian
- Laboratory Animal Research Center of Shanxi Medical University, Shanxi Key Laboratory of Animal and Animal Model of Human Diseases, Taiyuan 030001, China
| | - Ruochen Du
- Laboratory Animal Research Center of Shanxi Medical University, Shanxi Key Laboratory of Animal and Animal Model of Human Diseases, Taiyuan 030001, China
| | - Yitong Yuan
- Laboratory Animal Research Center of Shanxi Medical University, Shanxi Key Laboratory of Animal and Animal Model of Human Diseases, Taiyuan 030001, China
| | - Pengfei Li
- Laboratory Animal Research Center of Shanxi Medical University, Shanxi Key Laboratory of Animal and Animal Model of Human Diseases, Taiyuan 030001, China
| | - Fang Liu
- Laboratory Animal Research Center of Shanxi Medical University, Shanxi Key Laboratory of Animal and Animal Model of Human Diseases, Taiyuan 030001, China
| | - Chunfang Wang
- Laboratory Animal Research Center of Shanxi Medical University, Shanxi Key Laboratory of Animal and Animal Model of Human Diseases, Taiyuan 030001, China
| |
Collapse
|
25
|
Keatinge M, Tsarouchas TM, Munir T, Porter NJ, Larraz J, Gianni D, Tsai HH, Becker CG, Lyons DA, Becker T. CRISPR gRNA phenotypic screening in zebrafish reveals pro-regenerative genes in spinal cord injury. PLoS Genet 2021; 17:e1009515. [PMID: 33914736 PMCID: PMC8084196 DOI: 10.1371/journal.pgen.1009515] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Accepted: 03/28/2021] [Indexed: 12/30/2022] Open
Abstract
Zebrafish exhibit robust regeneration following spinal cord injury, promoted by macrophages that control post-injury inflammation. However, the mechanistic basis of how macrophages regulate regeneration is poorly understood. To address this gap in understanding, we conducted a rapid in vivo phenotypic screen for macrophage-related genes that promote regeneration after spinal injury. We used acute injection of synthetic RNA Oligo CRISPR guide RNAs (sCrRNAs) that were pre-screened for high activity in vivo. Pre-screening of over 350 sCrRNAs allowed us to rapidly identify highly active sCrRNAs (up to half, abbreviated as haCRs) and to effectively target 30 potentially macrophage-related genes. Disruption of 10 of these genes impaired axonal regeneration following spinal cord injury. We selected 5 genes for further analysis and generated stable mutants using haCRs. Four of these mutants (tgfb1a, tgfb3, tnfa, sparc) retained the acute haCR phenotype, validating the approach. Mechanistically, tgfb1a haCR-injected and stable mutant zebrafish fail to resolve post-injury inflammation, indicated by prolonged presence of neutrophils and increased levels of il1b expression. Inhibition of Il-1β rescues the impaired axon regeneration in the tgfb1a mutant. Hence, our rapid and scalable screening approach has identified functional regulators of spinal cord regeneration, but can be applied to any biological function of interest.
Collapse
Affiliation(s)
- Marcus Keatinge
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | | | - Tahimina Munir
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Nicola J. Porter
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Juan Larraz
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Davide Gianni
- Biogen, Cambridge, Massachusetts, United States of America
| | - Hui-Hsin Tsai
- Biogen, Cambridge, Massachusetts, United States of America
| | - Catherina G. Becker
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - David A. Lyons
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Thomas Becker
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
26
|
Asakawa K, Handa H, Kawakami K. Illuminating ALS Motor Neurons With Optogenetics in Zebrafish. Front Cell Dev Biol 2021; 9:640414. [PMID: 33816488 PMCID: PMC8012537 DOI: 10.3389/fcell.2021.640414] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Accepted: 02/22/2021] [Indexed: 11/13/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal neurological disorder characterized by progressive degeneration of motor neurons in the brain and spinal cord. Spinal motor neurons align along the spinal cord length within the vertebral column, and extend long axons to connect with skeletal muscles covering the body surface. Due to this anatomy, spinal motor neurons are among the most difficult cells to observe in vivo. Larval zebrafish have transparent bodies that allow non-invasive visualization of whole cells of single spinal motor neurons, from somas to the neuromuscular synapses. This unique feature, combined with its amenability to genome editing, pharmacology, and optogenetics, enables functional analyses of ALS-associated proteins in the spinal motor neurons in vivo with subcellular resolution. Here, we review the zebrafish skeletal neuromuscular system and the optical methods used to study it. We then introduce a recently developed optogenetic zebrafish ALS model that uses light illumination to control oligomerization, phase transition and aggregation of the ALS-associated DNA/RNA-binding protein called TDP-43. Finally, we will discuss how this disease-in-a-fish ALS model can help solve key questions about ALS pathogenesis and lead to new ALS therapeutics.
Collapse
Affiliation(s)
- Kazuhide Asakawa
- Department of Chemical Biology, Tokyo Medical University, Tokyo, Japan
| | - Hiroshi Handa
- Department of Chemical Biology, Tokyo Medical University, Tokyo, Japan
| | - Koichi Kawakami
- Division of Molecular and Developmental Biology, National Institute of Genetics, Mishima, Japan.,Department of Genetics, Graduate University for Advanced Studies (SOKENDAI), Mishima, Japan
| |
Collapse
|
27
|
Zeng CW, Kamei Y, Shigenobu S, Sheu JC, Tsai HJ. Injury-induced Cavl-expressing cells at lesion rostral side play major roles in spinal cord regeneration. Open Biol 2021; 11:200304. [PMID: 33622104 PMCID: PMC8061693 DOI: 10.1098/rsob.200304] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
The extent of cellular heterogeneity involved in neuronal regeneration after spinal cord injury (SCI) remains unclear. Therefore, we established stress-responsive transgenic zebrafish embryos with SCI. As a result, we found an SCI-induced cell population, termed SCI stress-responsive regenerating cells (SrRCs), essential for neuronal regeneration post-SCI. SrRCs were mostly composed of subtypes of radial glia (RGs-SrRCs) and neuron stem/progenitor cells (NSPCs-SrRCs) that are able to differentiate into neurons, and they formed a bridge across the lesion and connected with neighbouring undamaged motor neurons post-SCI. Compared to SrRCs at the caudal side of the SCI site (caudal-SrRCs), rostral-SrRCs participated more actively in neuronal regeneration. After RNA-seq analysis, we discovered that caveolin 1 (cav1) was significantly upregulated in rostral-SrRCs and that cav1 was responsible for the axonal regrowth and regenerative capability of rostral-SrRCs. Collectively, we define a specific SCI-induced cell population, SrRCs, involved in neuronal regeneration, demonstrate that rostral-SrRCs exhibit higher neuronal differentiation capability and prove that cav1 is predominantly expressed in rostral-SrRCs, playing a major role in neuronal regeneration after SCI.
Collapse
Affiliation(s)
- Chih-Wei Zeng
- Institute of Molecular and Cellular Biology, College of Life Science, National Taiwan University, Taipei 10617, Taiwan.,Liver Disease Prevention and Treatment Research Foundation, Taipei 10008, Taiwan
| | - Yasuhiro Kamei
- Spectrography and Bioimaging Facility, National Institute for Basic Biology (NIBB), National Institutes of Natural Sciences (NINS), Okazaki 444-8585, Japan.,Department of Basic Biology, The Graduate University for Advanced Studies (SOKENDAI), Okazaki 444-8585, Japan
| | - Shuji Shigenobu
- Department of Basic Biology, The Graduate University for Advanced Studies (SOKENDAI), Okazaki 444-8585, Japan.,Functional Genomics Facility, NIBB, NINS, Okazaki 444-8585, Japan
| | - Jin-Chuan Sheu
- Liver Disease Prevention and Treatment Research Foundation, Taipei 10008, Taiwan
| | - Huai-Jen Tsai
- Institute of Biomedical Sciences, Mackay Medical College, New Taipei City 25245, Taiwan.,Department of Life Science, Fu Jen Catholic University, New Taipei City 242062, Taiwan
| |
Collapse
|
28
|
Xie Y, Meijer AH, Schaaf MJM. Modeling Inflammation in Zebrafish for the Development of Anti-inflammatory Drugs. Front Cell Dev Biol 2021; 8:620984. [PMID: 33520995 PMCID: PMC7843790 DOI: 10.3389/fcell.2020.620984] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2020] [Accepted: 12/18/2020] [Indexed: 12/16/2022] Open
Abstract
Dysregulation of the inflammatory response in humans can lead to various inflammatory diseases, like asthma and rheumatoid arthritis. The innate branch of the immune system, including macrophage and neutrophil functions, plays a critical role in all inflammatory diseases. This part of the immune system is well-conserved between humans and the zebrafish, which has emerged as a powerful animal model for inflammation, because it offers the possibility to image and study inflammatory responses in vivo at the early life stages. This review focuses on different inflammation models established in zebrafish, and how they are being used for the development of novel anti-inflammatory drugs. The most commonly used model is the tail fin amputation model, in which part of the tail fin of a zebrafish larva is clipped. This model has been used to study fundamental aspects of the inflammatory response, like the role of specific signaling pathways, the migration of leukocytes, and the interaction between different immune cells, and has also been used to screen libraries of natural compounds, approved drugs, and well-characterized pathway inhibitors. In other models the inflammation is induced by chemical treatment, such as lipopolysaccharide (LPS), leukotriene B4 (LTB4), and copper, and some chemical-induced models, such as treatment with trinitrobenzene sulfonic acid (TNBS), specifically model inflammation in the gastro-intestinal tract. Two mutant zebrafish lines, carrying a mutation in the hepatocyte growth factor activator inhibitor 1a gene (hai1a) and the cdp-diacylglycerolinositol 3-phosphatidyltransferase (cdipt) gene, show an inflammatory phenotype, and they provide interesting model systems for studying inflammation. These zebrafish inflammation models are often used to study the anti-inflammatory effects of glucocorticoids, to increase our understanding of the mechanism of action of this class of drugs and to develop novel glucocorticoid drugs. In this review, an overview is provided of the available inflammation models in zebrafish, and how they are used to unravel molecular mechanisms underlying the inflammatory response and to screen for novel anti-inflammatory drugs.
Collapse
|
29
|
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
|
30
|
Abstract
Tissue or organ regeneration is a complex process with successful outcomes depending on the type of tissue and organism. Upon damage, mammals can only efficiently restore a few tissues including the liver, skin, epithelia of the lung, kidney, and gut. In contrast, lower vertebrates such as zebrafish possess an extraordinary regeneration ability, which restores the normal function of a broad spectrum of tissues including heart, fin, brain, spinal cord, and retina. This regeneration process is either mediated by the proliferation of resident stem cells, or cells that dedifferentiate into a stem cell-like. In recent years, evidence has suggested that the innate immune system can modulate stem cell activity to initiate the regenerative response to damage. This review will explore some of the newer concepts of inflammation in zebrafish regeneration in different tissues. Understanding how inflammation regulates regeneration in zebrafish would provide important clues to improve the therapeutic strategies for repairing injured mammalian tissues that do not have an inherent regenerative capacity.
Collapse
Affiliation(s)
- Maria Iribarne
- Center for Zebrafish Research, Department of Biological Sciences; Center for Stem Cells and Regenerative Medicine, University of Notre Dame, Notre Dame, IN, USA
| |
Collapse
|
31
|
Tsata V, Kroehne V, Wehner D, Rost F, Lange C, Hoppe C, Kurth T, Reinhardt S, Petzold A, Dahl A, Loeffler M, Reimer MM, Brand M. Reactive oligodendrocyte progenitor cells (re-)myelinate the regenerating zebrafish spinal cord. Development 2020; 147:dev193946. [PMID: 33158923 DOI: 10.1242/dev.193946] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2020] [Accepted: 10/28/2020] [Indexed: 12/11/2022]
Abstract
Spinal cord injury (SCI) results in loss of neurons, oligodendrocytes and myelin sheaths, all of which are not efficiently restored. The scarcity of oligodendrocytes in the lesion site impairs re-myelination of spared fibres, which leaves axons denuded, impedes signal transduction and contributes to permanent functional deficits. In contrast to mammals, zebrafish can functionally regenerate the spinal cord. Yet, little is known about oligodendroglial lineage biology and re-myelination capacity after SCI in a regeneration-permissive context. Here, we report that, in adult zebrafish, SCI results in axonal, oligodendrocyte and myelin sheath loss. We find that OPCs, the oligodendrocyte progenitor cells, survive the injury, enter a reactive state, proliferate and differentiate into oligodendrocytes. Concomitantly, the oligodendrocyte population is re-established to pre-injury levels within 2 weeks. Transcriptional profiling revealed that reactive OPCs upregulate the expression of several myelination-related genes. Interestingly, global reduction of axonal tracts and partial re-myelination, relative to pre-injury levels, persist at later stages of regeneration, yet are sufficient for functional recovery. Taken together, these findings imply that, in the zebrafish spinal cord, OPCs replace lost oligodendrocytes and, thus, re-establish myelination during regeneration.
Collapse
Affiliation(s)
- Vasiliki Tsata
- Center for Regenerative Therapies Dresden TU Dresden (CRTD) and Cluster of Excellence, Physics of Life (PoL), TU Dresden, Dresden 01307, Germany
| | - Volker Kroehne
- Center for Regenerative Therapies Dresden TU Dresden (CRTD) and Cluster of Excellence, Physics of Life (PoL), TU Dresden, Dresden 01307, Germany
| | - Daniel Wehner
- Center for Regenerative Therapies Dresden TU Dresden (CRTD) and Cluster of Excellence, Physics of Life (PoL), TU Dresden, Dresden 01307, Germany
- Max Planck Institute for the Science of Light, Erlangen 91058, Germany
- Max-Planck-Zentrum für Physik und Medizin, Erlangen 91058, Germany
| | - Fabian Rost
- Center for Regenerative Therapies Dresden TU Dresden (CRTD) and Cluster of Excellence, Physics of Life (PoL), TU Dresden, Dresden 01307, Germany
- Center for Information Services and High Performance Computing, TU Dresden, Dresden 01062, Germany
| | - Christian Lange
- Center for Regenerative Therapies Dresden TU Dresden (CRTD) and Cluster of Excellence, Physics of Life (PoL), TU Dresden, Dresden 01307, Germany
| | - Cornelia Hoppe
- Center for Regenerative Therapies Dresden TU Dresden (CRTD) and Cluster of Excellence, Physics of Life (PoL), TU Dresden, Dresden 01307, Germany
| | - Thomas Kurth
- Center for Molecular and Cellular Bioengineering (CMCB), TU Dresden, Technology Platform, Dresden 01307, Germany
| | - Susanne Reinhardt
- Dresden Genome Center c/o Center for Regenerative Therapies TU Dresden (CRTD), TU Dresden, Dresden 01307, Germany
| | - Andreas Petzold
- Dresden Genome Center c/o Center for Regenerative Therapies TU Dresden (CRTD), TU Dresden, Dresden 01307, Germany
| | - Andreas Dahl
- Dresden Genome Center c/o Center for Regenerative Therapies TU Dresden (CRTD), TU Dresden, Dresden 01307, Germany
| | - Markus Loeffler
- Center for Advancing Electronics Dresden (cfaed)/Dresden Center for Nanoanalysis (DCN), TU Dresden, Dresden 01062, Germany
| | - Michell M Reimer
- Center for Regenerative Therapies Dresden TU Dresden (CRTD) and Cluster of Excellence, Physics of Life (PoL), TU Dresden, Dresden 01307, Germany
| | - Michael Brand
- Center for Regenerative Therapies Dresden TU Dresden (CRTD) and Cluster of Excellence, Physics of Life (PoL), TU Dresden, Dresden 01307, Germany
| |
Collapse
|
32
|
Var SR, Byrd-Jacobs CA. Role of Macrophages and Microglia in Zebrafish Regeneration. Int J Mol Sci 2020; 21:E4768. [PMID: 32635596 PMCID: PMC7369716 DOI: 10.3390/ijms21134768] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 07/02/2020] [Accepted: 07/03/2020] [Indexed: 12/11/2022] Open
Abstract
Currently, there is no treatment for recovery of human nerve function after damage to the central nervous system (CNS), and there are limited regenerative capabilities in the peripheral nervous system. Since fish are known for their regenerative abilities, understanding how these species modulate inflammatory processes following injury has potential translational importance for recovery from damage and disease. Many diseases and injuries involve the activation of innate immune cells to clear damaged cells. The resident immune cells of the CNS are microglia, the primary cells that respond to infection and injury, and their peripheral counterparts, macrophages. These cells serve as key modulators of development and plasticity and have been shown to be important in the repair and regeneration of structure and function after injury. Zebrafish are an emerging model for studying macrophages in regeneration after injury and microglia in neurodegenerative disorders such as Parkinson's disease and Alzheimer's disease. These fish possess a high degree of neuroanatomical, neurochemical, and emotional/social behavioral resemblance with humans, serving as an ideal simulator for many pathologies. This review explores literature on macrophage and microglial involvement in facilitating regeneration. Understanding innate immune cell behavior following damage may help to develop novel methods for treating toxic and chronic inflammatory processes that are seen in trauma and disease.
Collapse
|
33
|
Mruk K, Ciepla P, Piza PA, Alnaqib MA, Chen JK. Targeted cell ablation in zebrafish using optogenetic transcriptional control. Development 2020; 147:dev183640. [PMID: 32414936 PMCID: PMC7328002 DOI: 10.1242/dev.183640] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Accepted: 04/22/2020] [Indexed: 12/11/2022]
Abstract
Cell ablation is a powerful method for elucidating the contributions of individual cell populations to embryonic development and tissue regeneration. Targeted cell loss in whole organisms has been typically achieved through expression of a cytotoxic or prodrug-activating gene product in the cell type of interest. This approach depends on the availability of tissue-specific promoters, and it does not allow further spatial selectivity within the promoter-defined region(s). To address this limitation, we have used the light-inducible GAVPO transactivator in combination with two genetically encoded cell-ablation technologies: the nitroreductase/nitrofuran system and a cytotoxic variant of the M2 ion channel. Our studies establish ablative methods that provide the tissue specificity afforded by cis-regulatory elements and the conditionality of optogenetics. Our studies also demonstrate differences between the nitroreductase and M2 systems that influence their efficacies for specific applications. Using this integrative approach, we have ablated cells in zebrafish embryos with both spatial and temporal control.
Collapse
Affiliation(s)
- Karen Mruk
- Department of Chemical and Systems Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
- School of Pharmacy, University of Wyoming, Laramie, WY 82071, USA
| | - Paulina Ciepla
- Department of Chemical and Systems Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Patrick A Piza
- Department of Chemical and Systems Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Mohammad A Alnaqib
- Department of Chemical and Systems Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - James K Chen
- Department of Chemical and Systems Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
- Department of Chemistry, Stanford University, Stanford, CA 94305, USA
| |
Collapse
|
34
|
Flaive A, Cabelguen JM, Ryczko D. The serotonin reuptake blocker citalopram destabilizes fictive locomotor activity in salamander axial circuits through 5-HT 1A receptors. J Neurophysiol 2020; 123:2326-2342. [PMID: 32401145 DOI: 10.1152/jn.00179.2020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Serotoninergic (5-HT) neurons are powerful modulators of spinal locomotor circuits. Most studies on 5-HT modulation focused on the effect of exogenous 5-HT and these studies provided key information about the cellular mechanisms involved. Less is known about the effects of increased release of endogenous 5-HT with selective serotonin reuptake inhibitors. In mammals, such molecules were shown to destabilize the fictive locomotor output of spinal limb networks through 5-HT1A receptors. However, in tetrapods little is known about the effects of increased 5-HT release on the locomotor output of axial networks, which are coordinated with limb circuits during locomotion from basal vertebrates to mammals. Here, we examined the effect of citalopram on fictive locomotion generated in axial segments of isolated spinal cords in salamanders, a tetrapod where raphe 5-HT reticulospinal neurons and intraspinal 5-HT neurons are present as in other vertebrates. Using electrophysiological recordings of ventral roots, we show that fictive locomotion generated by bath-applied glutamatergic agonists is destabilized by citalopram. Citalopram-induced destabilization was prevented by a 5-HT1A receptor antagonist, whereas a 5-HT1A receptor agonist destabilized fictive locomotion. Using immunofluorescence experiments, we found 5-HT-positive fibers and varicosities in proximity with motoneurons and glutamatergic interneurons that are likely involved in rhythmogenesis. Our results show that increasing 5-HT release has a deleterious effect on axial locomotor activity through 5-HT1A receptors. This is consistent with studies in limb networks of turtle and mouse, suggesting that this part of the complex 5-HT modulation of spinal locomotor circuits is common to limb and axial networks in limbed vertebrates.NEW & NOTEWORTHY Little is known about the modulation exerted by endogenous serotonin on axial locomotor circuits in tetrapods. Using axial ventral root recordings in salamanders, we found that a serotonin reuptake blocker destabilized fictive locomotor activity through 5-HT1A receptors. Our anatomical results suggest that serotonin is released on motoneurons and glutamatergic interneurons possibly involved in rhythmogenesis. Our study suggests that common serotoninergic mechanisms modulate axial motor circuits in amphibians and limb motor circuits in reptiles and mammals.
Collapse
Affiliation(s)
- Aurélie Flaive
- Département de Pharmacologie-Physiologie, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Sherbrooke, Quebec, Canada
| | - Jean-Marie Cabelguen
- Neurocentre Magendie, INSERM U 862, Université de Bordeaux, Bordeaux Cedex, France
| | - Dimitri Ryczko
- Département de Pharmacologie-Physiologie, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Sherbrooke, Quebec, Canada.,Centre de recherche du Centre Hospitalier Universitaire de Sherbrooke, Sherbrooke, Quebec, Canada.,Institut de Pharmacologie de Sherbrooke, Sherbrooke, Quebec, Canada.,Centre des neurosciences de Sherbrooke, Sherbrooke, Quebec, Canada
| |
Collapse
|
35
|
Cigliola V, Becker CJ, Poss KD. Building bridges, not walls: spinal cord regeneration in zebrafish. Dis Model Mech 2020; 13:13/5/dmm044131. [PMID: 32461216 PMCID: PMC7272344 DOI: 10.1242/dmm.044131] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Spinal cord injury is a devastating condition in which massive cell death and disruption of neural circuitry lead to long-term chronic functional impairment and paralysis. In mammals, spinal cord tissue has minimal capacity to regenerate after injury. In stark contrast, the regeneration of a completely transected spinal cord and accompanying reversal of paralysis in adult zebrafish is arguably one of the most spectacular biological phenomena in nature. Here, we review reports from the last decade that dissect the mechanisms of spinal cord regeneration in zebrafish. We highlight recent progress as well as areas requiring emphasis in a line of study that has great potential to uncover strategies for human spinal cord repair. Summary: Unlike mammals, teleost fish are capable of efficient, spontaneous recovery after a paralyzing spinal cord injury. Here, we highlight the major events through which laboratory model zebrafish regenerate spinal cord tissue.
Collapse
Affiliation(s)
- Valentina Cigliola
- Department of Cell Biology, Duke University Medical Center, Durham, NC 27710, USA.,Regeneration Next, Duke University, Durham, NC 27710, USA
| | - Clayton J Becker
- Department of Cell Biology, Duke University Medical Center, Durham, NC 27710, USA.,Regeneration Next, Duke University, Durham, NC 27710, USA
| | - Kenneth D Poss
- Department of Cell Biology, Duke University Medical Center, Durham, NC 27710, USA .,Regeneration Next, Duke University, Durham, NC 27710, USA
| |
Collapse
|
36
|
Herzog C, Greenald D, Larraz J, Keatinge M, Herrgen L. RNA-seq analysis and compound screening highlight multiple signalling pathways regulating secondary cell death after acute CNS injury in vivo. Biol Open 2020; 9:9/5/bio050260. [PMID: 32366533 PMCID: PMC7225090 DOI: 10.1242/bio.050260] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Understanding the molecular mechanisms that regulate secondary cell death after acute central nervous system (CNS) injury is critical for the development of effective neuroprotective drugs. Previous research has shown that neurotoxic processes including excitotoxicity, oxidative stress and neuroinflammation can cause secondary cell death. Nevertheless, clinical trials targeting these processes have been largely unsuccessful, suggesting that the signalling pathways underlying secondary cell death remain incompletely understood. Due to their suitability for live imaging and their amenability to genetic and pharmacological manipulation, larval zebrafish provide an ideal platform for studying the regulation of secondary cell death in vivo Here, we use RNA-seq gene expression profiling and compound screening to identify signalling pathways that regulate secondary cell death after acute neural injury in larval zebrafish. RNA-seq analysis of genes upregulated in cephalic mpeg1+ macrophage-lineage cells isolated from mpeg1:GFP transgenic larvae after neural injury suggested an involvement of cytokine and polyamine signalling in secondary cell death. Furthermore, screening a library of FDA approved compounds indicated roles for GABA, serotonin and dopamine signalling. Overall, our results highlight multiple signalling pathways that regulate secondary cell death in vivo, and thus provide a starting point for the development of novel neuroprotective treatments for patients with CNS injury.This article has an associated First Person interview with the two first authors of the paper.
Collapse
Affiliation(s)
- Chiara Herzog
- Centre for Discovery Brain Sciences, Deanery of Biomedical Sciences, The University of Edinburgh, 49 Little France Crescent, Edinburgh EH16 4SB, UK
| | - David Greenald
- Centre for Discovery Brain Sciences, Deanery of Biomedical Sciences, The University of Edinburgh, 49 Little France Crescent, Edinburgh EH16 4SB, UK
| | - Juan Larraz
- Centre for Discovery Brain Sciences, Deanery of Biomedical Sciences, The University of Edinburgh, 49 Little France Crescent, Edinburgh EH16 4SB, UK
| | - Marcus Keatinge
- Centre for Discovery Brain Sciences, Deanery of Biomedical Sciences, The University of Edinburgh, 49 Little France Crescent, Edinburgh EH16 4SB, UK
| | - Leah Herrgen
- Centre for Discovery Brain Sciences, Deanery of Biomedical Sciences, The University of Edinburgh, 49 Little France Crescent, Edinburgh EH16 4SB, UK
| |
Collapse
|
37
|
|
38
|
Park S, Lee JY, You S, Song G, Lim W. Neurotoxic effects of aflatoxin B1 on human astrocytes in vitro and on glial cell development in zebrafish in vivo. JOURNAL OF HAZARDOUS MATERIALS 2020; 386:121639. [PMID: 31787402 DOI: 10.1016/j.jhazmat.2019.121639] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Revised: 11/02/2019] [Accepted: 11/07/2019] [Indexed: 06/10/2023]
Abstract
Aflatoxin B1 is one of the well-known mycotoxins and mainly found in contaminated animal feed and various agricultural products inducing acute and chronic toxicology, tumor, and abnormal neural development. However, the effects of aflatoxin B1 on the human brain, especially on astrocytes, have not been studied in depth. In the present study, we studied the neurotoxic effects of aflatoxin B1, in vitro and in vivo. Aflatoxin B1 decreased the proliferation and stopped cell cycle progression at the sub G0/G1 stage with an increase in BAX, BAK, and cytochrome c proteins in human astrocytes. In addition, it increased the mitochondrial depolarization, oxidative stress, and calcium influx in both the cytosol and mitochondria. Surprisingly, inhibition of calcium overload in the cytosol and mitochondria, using calcium chelators and an inhibitor, partially rescued the proliferation of aflatoxin B1-treated astrocytes. Based on the toxicity assays using zebrafish models, aflatoxin B1 decreased the embryo survival rate with physiological changes and an increase in the caspase and tp53 genes. It also decreased the expression of gfap, mbp, and olig2 in the transgenic zebrafish embryo's brain and axon. Our results revealed the specific mechanism of the neurotoxic effects of aflatoxin B1 on human astrocytes and zebrafish glial cells.
Collapse
Affiliation(s)
- Sunwoo Park
- Institute of Animal Molecular Biotechnology and Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul, 02841, Republic of Korea
| | - Jin-Young Lee
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, Wisconsin 53226, USA
| | - Seungkwon You
- Institute of Animal Molecular Biotechnology and Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul, 02841, Republic of Korea.
| | - Gwonhwa Song
- Institute of Animal Molecular Biotechnology and Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul, 02841, Republic of Korea.
| | - Whasun Lim
- Department of Food and Nutrition, Kookmin University, Seoul, 02707, Republic of Korea.
| |
Collapse
|
39
|
Mazzolini J, Le Clerc S, Morisse G, Coulonges C, Kuil LE, van Ham TJ, Zagury J, Sieger D. Gene expression profiling reveals a conserved microglia signature in larval zebrafish. Glia 2020; 68:298-315. [PMID: 31508850 PMCID: PMC6916425 DOI: 10.1002/glia.23717] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Revised: 08/21/2019] [Accepted: 08/22/2019] [Indexed: 12/23/2022]
Abstract
Microglia are the resident macrophages of the brain. Over the past decade, our understanding of the function of these cells has significantly improved. Microglia do not only play important roles in the healthy brain but are involved in almost every brain pathology. Gene expression profiling allowed to distinguish microglia from other macrophages and revealed that the full microglia signature can only be observed in vivo. Thus, animal models are irreplaceable to understand the function of these cells. One of the popular models to study microglia is the zebrafish larva. Due to their optical transparency and genetic accessibility, zebrafish larvae have been employed to understand a variety of microglia functions in the living brain. Here, we performed RNA sequencing of larval zebrafish microglia at different developmental time points: 3, 5, and 7 days post fertilization (dpf). Our analysis reveals that larval zebrafish microglia rapidly acquire the core microglia signature and many typical microglia genes are expressed from 3 dpf onwards. The majority of changes in gene expression happened between 3 and 5 dpf, suggesting that differentiation mainly takes place during these days. Furthermore, we compared the larval microglia transcriptome to published data sets of adult zebrafish microglia, mouse microglia, and human microglia. Larval microglia shared a significant number of expressed genes with their adult counterparts in zebrafish as well as with mouse and human microglia. In conclusion, our results show that larval zebrafish microglia mature rapidly and express the core microglia gene signature that seems to be conserved across species.
Collapse
Affiliation(s)
- Julie Mazzolini
- Centre for Discovery Brain SciencesUniversity of EdinburghEdinburghUK
| | - Sigrid Le Clerc
- Laboratoire GBCM, EA7528, Conservatoire National des Arts et MétiersHESAM UniversitéParisFrance
| | - Gregoire Morisse
- Centre for Discovery Brain SciencesUniversity of EdinburghEdinburghUK
| | - Cédric Coulonges
- Laboratoire GBCM, EA7528, Conservatoire National des Arts et MétiersHESAM UniversitéParisFrance
| | - Laura E. Kuil
- Department of Clinical Genetics, Erasmus MCUniversity Medical Center RotterdamRotterdamThe Netherlands
| | - Tjakko J. van Ham
- Department of Clinical Genetics, Erasmus MCUniversity Medical Center RotterdamRotterdamThe Netherlands
| | - Jean‐François Zagury
- Laboratoire GBCM, EA7528, Conservatoire National des Arts et MétiersHESAM UniversitéParisFrance
| | - Dirk Sieger
- Centre for Discovery Brain SciencesUniversity of EdinburghEdinburghUK
| |
Collapse
|
40
|
Sabin KZ, Echeverri K. The role of the immune system during regeneration of the central nervous system. ACTA ACUST UNITED AC 2019; 7. [PMID: 32864529 DOI: 10.1016/j.regen.2019.100023] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Central nervous system damage in mammals leads to neuronal cell death, axonal degeneration, and formation of a glial scar resulting in functional and behavioral defects. Other vertebrates, like fish and salamanders, have retained the ability to functionally regenerate after central nervous system injury. To date research from many research organisms has led to a more concise understanding of the response of local neural cells to injury. However, it has become clear that non-neural cells of the immune system play an important role in determining the tissue response to injury. In this review we briefly consider the mammalian response to injury compared to organisms with the natural ability to regenerate. We then discuss similarities and differences in how cells of the innate and adaptive immune system respond and contribute to tissue repair in various species.
Collapse
Affiliation(s)
- K Z Sabin
- Eugene Bell Center for Regenerative Biology & Tissue Engineering, Marine Biological Laboratory, Woods Hole, MA 02543 USA
| | - K Echeverri
- Eugene Bell Center for Regenerative Biology & Tissue Engineering, Marine Biological Laboratory, Woods Hole, MA 02543 USA
| |
Collapse
|
41
|
Gao T, Li J, Li N, Gao Y, Yu L, Zhuang S, Zhao Y, Dong X. lncrps25 play an essential role in motor neuron development through controlling the expression of olig2 in zebrafish. J Cell Physiol 2019; 235:3485-3496. [PMID: 31549395 DOI: 10.1002/jcp.29237] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Accepted: 09/03/2019] [Indexed: 12/17/2022]
Abstract
lncrps25 is an intergenic long noncoding RNA (lncRNA), which is location close to rps25 (ribosomal protein S25) gene, is reported share high conserved sequence with NREP (neuronal regeneration-related protein) 3'-untranslated region. The function and mechanism of most of the lncRNA in embryo development remain largely unknown. In zebrafish, lncrps25 is widely expressed in the early embryonic stage and spinal cord during development. Morpholino (MO) knockdown of zebrafish lncrps25 exhibit locomotor behavior defects, caused by abnormal development of motor neurons. In addition, the defect of swimming ability and motor neurons could be recovery by microinject with lncrps25 RNA in lncrps25 morphants. By performing RNA sequencing and quantitative real-time polymerase chain reaction, we found that olig2 (oligodendrocyte transcription factor 2) messenger RNA (mRNA) was downregulated in lncrps25 morphants. Moreover, overexpression of olig2 mRNA in lncrps25 morphants partially rescued motor neurons development. Taken together, these results indicate that lncrps25 plays an essential role in the development of motor neurons in zebrafish.
Collapse
Affiliation(s)
- Tianheng Gao
- College of Oceanography, Hohai University, Nanjing, China
| | - Jingyun Li
- Nanjing Maternal and Child Health Medical Institute, Women's Hospital of Nanjing Medical University (Nanjing Maternity and Child Health Care Hospital), Nanjing, China
| | - Nan Li
- Ministry of Education Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Nanjing University, Nanjing, China
| | - Yan Gao
- Ministry of Education Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Nanjing University, Nanjing, China
| | - Lingling Yu
- Department of Pediatric, Jingjiang People's Hospital Affiliated to Yangzhou University, Jingjiang, China
| | - Sisi Zhuang
- Nanjing Maternal and Child Health Medical Institute, Women's Hospital of Nanjing Medical University (Nanjing Maternity and Child Health Care Hospital), Nanjing, China.,Department of Pediatrics, The First Affiliated Hospital, Nanjing Medical University, Nanjing, China
| | - Yingmin Zhao
- Department of Pediatric, Jingjiang People's Hospital Affiliated to Yangzhou University, Jingjiang, China
| | - Xiaohua Dong
- Department of Pediatric, Jingjiang People's Hospital Affiliated to Yangzhou University, Jingjiang, China
| |
Collapse
|
42
|
Marques IJ, Lupi E, Mercader N. Model systems for regeneration: zebrafish. Development 2019; 146:146/18/dev167692. [DOI: 10.1242/dev.167692] [Citation(s) in RCA: 94] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Accepted: 08/19/2019] [Indexed: 12/13/2022]
Abstract
ABSTRACT
Tissue damage can resolve completely through healing and regeneration, or can produce permanent scarring and loss of function. The response to tissue damage varies across tissues and between species. Determining the natural mechanisms behind regeneration in model organisms that regenerate well can help us develop strategies for tissue recovery in species with poor regenerative capacity (such as humans). The zebrafish (Danio rerio) is one of the most accessible vertebrate models to study regeneration. In this Primer, we highlight the tools available to study regeneration in the zebrafish, provide an overview of the mechanisms underlying regeneration in this system and discuss future perspectives for the field.
Collapse
Affiliation(s)
- Ines J. Marques
- Institute of Anatomy, University of Bern, Bern 3012, Switzerland
| | - Eleonora Lupi
- Institute of Anatomy, University of Bern, Bern 3012, Switzerland
- Acquifer, Ditabis, Digital Biomedical Imaging Systems, Pforzheim, Germany
| | - Nadia Mercader
- Institute of Anatomy, University of Bern, Bern 3012, Switzerland
- Centro Nacional de Investigaciones Cardiovasculares CNIC, Madrid 2029, Spain
| |
Collapse
|
43
|
Anguita-Salinas C, Sánchez M, Morales RA, Ceci ML, Rojas-Benítez D, Allende ML. Cellular Dynamics during Spinal Cord Regeneration in Larval Zebrafish. Dev Neurosci 2019; 41:112-122. [PMID: 31390621 DOI: 10.1159/000500185] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Accepted: 04/04/2019] [Indexed: 11/19/2022] Open
Abstract
The study of spinal cord regeneration using diverse animal models, which range from null to robust regenerative capabilities, is imperative for understanding how regeneration evolved and, eventually, to treat spinal cord injury and paralysis in humans. In this study, we used electroablation to fully transect the spinal cord of zebrafish larvae (3 days postfertilization) and examined regeneration of the tissue over time. We used transgenic lines to follow immune cells, oligodendrocytes, and neurons in vivo during the entire regenerative process. We observed that immune cells are recruited to the injury site, oligodendrocytes progenitor cells (olig2-expressing cells) invade, and axons cross the gap generated upon damage from anterior to reinnervate caudal structures. Together with the recovery of cell types and structures, a complete reversal of paralysis was observed in the lesioned larvae indicating functional regeneration. Finally, using transplantation to obtain mosaic larvae with single-labeled neurons, we show that severed spinal axons exhibited varying regenerative capabilities and plasticity depending on their original dorsoventral position in the spinal cord.
Collapse
Affiliation(s)
- Consuelo Anguita-Salinas
- FONDAP Center for Genome Regulation, Facultad de Ciencias, Universidad de Chile, Santiago, Chile
| | - Mario Sánchez
- FONDAP Center for Genome Regulation, Facultad de Ciencias, Universidad de Chile, Santiago, Chile
| | - Rodrigo A Morales
- FONDAP Center for Genome Regulation, Facultad de Ciencias, Universidad de Chile, Santiago, Chile
| | - María Laura Ceci
- FONDAP Center for Genome Regulation, Facultad de Ciencias, Universidad de Chile, Santiago, Chile
| | - Diego Rojas-Benítez
- FONDAP Center for Genome Regulation, Facultad de Ciencias, Universidad de Chile, Santiago, Chile
| | - Miguel L Allende
- FONDAP Center for Genome Regulation, Facultad de Ciencias, Universidad de Chile, Santiago, Chile,
| |
Collapse
|
44
|
Chapela D, Sousa S, Martins I, Cristóvão AM, Pinto P, Corte-Real S, Saúde L. A zebrafish drug screening platform boosts the discovery of novel therapeutics for spinal cord injury in mammals. Sci Rep 2019; 9:10475. [PMID: 31324865 PMCID: PMC6642202 DOI: 10.1038/s41598-019-47006-w] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Accepted: 07/05/2019] [Indexed: 12/14/2022] Open
Abstract
Spinal cord injury (SCI) is a complex condition, with limited therapeutic options, that results in sensory and motor disabilities. To boost discovery of novel therapeutics, we designed a simple and efficient drug screening platform. This innovative approach allows to determine locomotor rescue properties of small molecules in a zebrafish (Danio rerio) larval spinal cord transection model. We validated our screening platform by showing that Riluzole and Minocycline, two molecules that are in clinical trials for SCI, promote rescue of the locomotor function of the transected larvae. Further validation of the platform was obtained through the blind identification of D-Cycloserine, a molecule scheduled to enter phase IV clinical trials for SCI. Importantly, we identified Tranexamic acid and further showed that this molecule maintains its locomotor recovery properties in a rodent female contusion model. Our screening platform, combined with drug repurposing, promises to propel the rapid translation of novel therapeutics to improve SCI recovery in humans.
Collapse
Affiliation(s)
- Diana Chapela
- TechnoPhage, SA, Av. Prof. Egas Moniz, 1649-028, Lisboa, Portugal.,Instituto de Medicina Molecular, Faculdade de Medicina da Universidade de Lisboa, 1649-028, Lisboa, Portugal
| | - Sara Sousa
- TechnoPhage, SA, Av. Prof. Egas Moniz, 1649-028, Lisboa, Portugal
| | - Isaura Martins
- Instituto de Medicina Molecular, Faculdade de Medicina da Universidade de Lisboa, 1649-028, Lisboa, Portugal
| | - Ana Margarida Cristóvão
- Instituto de Medicina Molecular, Faculdade de Medicina da Universidade de Lisboa, 1649-028, Lisboa, Portugal
| | - Patrícia Pinto
- TechnoPhage, SA, Av. Prof. Egas Moniz, 1649-028, Lisboa, Portugal
| | - Sofia Corte-Real
- TechnoPhage, SA, Av. Prof. Egas Moniz, 1649-028, Lisboa, Portugal
| | - Leonor Saúde
- Instituto de Medicina Molecular e Instituto de Histologia e Biologia do Desenvolvimento, Faculdade de Medicina da Universidade de Lisboa, 1649-028, Lisboa, Portugal.
| |
Collapse
|
45
|
Herzog C, Pons Garcia L, Keatinge M, Greenald D, Moritz C, Peri F, Herrgen L. Rapid clearance of cellular debris by microglia limits secondary neuronal cell death after brain injury in vivo. Development 2019; 146:146/9/dev174698. [PMID: 31076485 DOI: 10.1242/dev.174698] [Citation(s) in RCA: 70] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Accepted: 04/11/2019] [Indexed: 12/25/2022]
Abstract
Moderate or severe traumatic brain injury (TBI) causes widespread neuronal cell death. Microglia, the resident macrophages of the brain, react to injury by migrating to the lesion site, where they phagocytose cellular debris. Microglial phagocytosis can have both beneficial (e.g. debris clearance) and detrimental (e.g. respiratory burst, phagoptosis) consequences. Hence, whether the overall effect of microglial phagocytosis after brain injury in vivo is neuroprotective or neurotoxic is not known. Here, we establish a system with which to carry out dynamic real-time analyses of the mechanisms regulating cell death after brain injury in vivo We show that mechanical injury to the larval zebrafish brain induces distinct phases of primary and secondary cell death. Excitotoxicity contributes to secondary cell death in zebrafish, reflecting findings from mammals. Microglia arrive at the lesion site within minutes of injury, where they rapidly engulf dead cells. Importantly, the rate of secondary cell death is increased when the rapid removal of cellular debris by microglia is reduced pharmacologically or genetically. In summary, our results provide evidence that microglial debris clearance is neuroprotective after brain injury in vivo.
Collapse
Affiliation(s)
- Chiara Herzog
- Centre for Discovery Brain Sciences, The University of Edinburgh, 49 Little France Crescent, Edinburgh EH16 4SB, UK
| | - Laura Pons Garcia
- Centre for Discovery Brain Sciences, The University of Edinburgh, 49 Little France Crescent, Edinburgh EH16 4SB, UK
| | - Marcus Keatinge
- Centre for Discovery Brain Sciences, The University of Edinburgh, 49 Little France Crescent, Edinburgh EH16 4SB, UK
| | - David Greenald
- Centre for Discovery Brain Sciences, The University of Edinburgh, 49 Little France Crescent, Edinburgh EH16 4SB, UK
| | | | - Francesca Peri
- Institute of Molecular Life Sciences, University of Zürich, Winterthurerstraße 190, 8057 Zürich, Switzerland
| | - Leah Herrgen
- Centre for Discovery Brain Sciences, The University of Edinburgh, 49 Little France Crescent, Edinburgh EH16 4SB, UK
| |
Collapse
|
46
|
Nelson CM, Lennon VA, Lee H, Krug RG, Kamalova A, Madigan NN, Clark KJ, Windebank AJ, Henley JR. Glucocorticoids Target Ependymal Glia and Inhibit Repair of the Injured Spinal Cord. Front Cell Dev Biol 2019; 7:56. [PMID: 31069223 PMCID: PMC6491705 DOI: 10.3389/fcell.2019.00056] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Accepted: 03/29/2019] [Indexed: 01/16/2023] Open
Abstract
Following injury, the mammalian spinal cord forms a glial scar and fails to regenerate. In contrast, vertebrate fish spinal cord tissue regenerates significantly to restore function. Cord transection in zebrafish (Danio rerio) initially causes paralysis and neural cell death. Subsequently, ependymal glia proliferate, bipolar glia extend across the lesion, and new neurons are born; axons from spared and nascent neurons extend along trans-lesional glial bridges to restore functional connectivity. Here we report that glucocorticoids, used in the clinical management of spinal cord injury, directly inhibit neural repair by targeting ependymal glia independently of hematogenous cells and microglia. After transecting injury, the glucocorticoid receptor in ependymal glia is regulated differentially in zebrafish (becoming inactive) vs. the rat (becoming active). Glucocorticoid blockade of neural regeneration via a direct effect on ependymal glia has important therapeutic implications for the putative benefit of corticosteroids in early management of spinal cord injury.
Collapse
Affiliation(s)
- Craig M Nelson
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN, United States.,Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, United States
| | - Vanda A Lennon
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, United States.,Department of Neurology, Mayo Clinic, Rochester, MN, United States.,Department of Immunology, Mayo Clinic, Rochester, MN, United States
| | - Han Lee
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, United States
| | - Randall G Krug
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, United States
| | - Aichurok Kamalova
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN, United States
| | | | - Karl J Clark
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, United States
| | | | - John R Henley
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN, United States.,Department of Physiology and Biomedical Engineering, Mayo Graduate School, Mayo Clinic College of Medicine and Science, Rochester, MN, United States
| |
Collapse
|
47
|
Regeneration of Dopaminergic Neurons in Adult Zebrafish Depends on Immune System Activation and Differs for Distinct Populations. J Neurosci 2019; 39:4694-4713. [PMID: 30948475 DOI: 10.1523/jneurosci.2706-18.2019] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Revised: 03/07/2019] [Accepted: 03/14/2019] [Indexed: 12/23/2022] Open
Abstract
Adult zebrafish, in contrast to mammals, regenerate neurons in their brain, but the extent and variability of this capacity is unclear. Here we ask whether the loss of various dopaminergic neuron populations is sufficient to trigger their functional regeneration. Both sexes of zebrafish were analyzed. Genetic lineage tracing shows that specific diencephalic ependymo-radial glial (ERG) progenitor cells give rise to new dopaminergic [tyrosine hydroxylase-positive (TH+)] neurons. Ablation elicits an immune response, increased proliferation of ERG progenitor cells, and increased addition of new TH+ neurons in populations that constitutively add new neurons (e.g., diencephalic population 5/6). Inhibiting the immune response attenuates neurogenesis to control levels. Boosting the immune response enhances ERG proliferation, but not addition of TH+ neurons. In contrast, in populations in which constitutive neurogenesis is undetectable (e.g., the posterior tuberculum and locus ceruleus), cell replacement and tissue integration are incomplete and transient. This is associated with a loss of spinal TH+ axons, as well as permanent deficits in shoaling and reproductive behavior. Hence, dopaminergic neuron populations in the adult zebrafish brain show vast differences in regenerative capacity that correlate with constitutive addition of neurons and depend on immune system activation.SIGNIFICANCE STATEMENT Despite the fact that zebrafish show a high propensity to regenerate neurons in the brain, this study reveals that not all types of dopaminergic neurons are functionally regenerated after specific ablation. Hence, in the same adult vertebrate brain, mechanisms of successful and incomplete regeneration can be studied. We identify progenitor cells for dopaminergic neurons and show that activating the immune system promotes the proliferation of these cells. However, in some areas of the brain this only leads to insufficient replacement of functionally important dopaminergic neurons that later disappear. Understanding the mechanisms of regeneration in zebrafish may inform interventions targeting the regeneration of functionally important neurons, such as dopaminergic neurons, from endogenous progenitor cells in nonregenerating mammals.
Collapse
|
48
|
Investigating Nrg1 Signaling in the Regenerating Axolotl Spinal Cord Using Multiplexed FISH. Dev Neurobiol 2019; 79:453-467. [DOI: 10.1002/dneu.22670] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2018] [Revised: 01/24/2019] [Accepted: 02/16/2019] [Indexed: 02/02/2023]
|
49
|
Green LA, Nebiolo JC, Smith CJ. Microglia exit the CNS in spinal root avulsion. PLoS Biol 2019; 17:e3000159. [PMID: 30794533 PMCID: PMC6402705 DOI: 10.1371/journal.pbio.3000159] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2018] [Revised: 03/06/2019] [Accepted: 02/06/2019] [Indexed: 12/31/2022] Open
Abstract
Microglia are central nervous system (CNS)-resident cells. Their ability to migrate outside of the CNS, however, is not understood. Using time-lapse imaging in an obstetrical brachial plexus injury (OBPI) model, we show that microglia squeeze through the spinal boundary and emigrate to peripheral spinal roots. Although both macrophages and microglia respond, microglia are the debris-clearing cell. Once outside the CNS, microglia re-enter the spinal cord in an altered state. These peripheral nervous system (PNS)-experienced microglia can travel to distal CNS areas from the injury site, including the brain, with debris. This emigration is balanced by two mechanisms—induced emigration via N-methyl-D-aspartate receptor (NMDA) dependence and restriction via contact-dependent cellular repulsion with macrophages. These discoveries open the possibility that microglia can migrate outside of their textbook-defined regions in disease states. Microglia are normally assumed to be confined to the central nervous system (CNS), but this study shows show that after spinal root injury, microglia can exit the CNS to clear debris. Upon re-entry, the emigrated microglia are altered and can travel to distal areas such as the brain. Cells are precisely organized in specific anatomical domains to ensure normal functioning of the nervous system. One such cell type, microglia, is usually considered to be confined to the central nervous system (CNS). Using time-lapse imaging to capture microglia as they migrate, we show that their characteristic CNS-residency can be altered after spinal root injury. After such injury, the microglia exit the spinal root to the periphery, where they clear debris at the injury site and then carry that debris back into the CNS. In addition, microglia that leave the CNS after spinal root injury become distinct from those that remain within the CNS. This emigration event of microglia after injury is driven by two mechanisms—dependence on glutamatergic signaling that induces their emigration to the injury and interactions with macrophages that prevent their ectopic exit from the spinal cord. Together, these discoveries raise the possibility that microglia could override their CNS-residency in certain disease contexts.
Collapse
Affiliation(s)
- Lauren A Green
- Department of Biological Sciences, University of Notre Dame, Notre Dame, Indiana, United States of America.,Center for Stem Cells and Regenerative Medicine, University of Notre Dame, Notre Dame, Indiana, United States of America
| | - Julia C Nebiolo
- Department of Biological Sciences, University of Notre Dame, Notre Dame, Indiana, United States of America
| | - Cody J Smith
- Department of Biological Sciences, University of Notre Dame, Notre Dame, Indiana, United States of America.,Center for Stem Cells and Regenerative Medicine, University of Notre Dame, Notre Dame, Indiana, United States of America
| |
Collapse
|
50
|
Dehnisch Ellström I, Spulber S, Hultin S, Norlin N, Ceccatelli S, Hultling C, Uhlén P. Spinal cord injury in zebrafish induced by near-infrared femtosecond laser pulses. J Neurosci Methods 2019; 311:259-266. [DOI: 10.1016/j.jneumeth.2018.10.035] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Revised: 10/28/2018] [Accepted: 10/29/2018] [Indexed: 12/09/2022]
|