1
|
Cellini BR, Edachola SV, Faw TD, Cigliola V. Blueprints for healing: central nervous system regeneration in zebrafish and neonatal mice. BMC Biol 2025; 23:115. [PMID: 40307837 PMCID: PMC12044871 DOI: 10.1186/s12915-025-02203-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Accepted: 03/31/2025] [Indexed: 05/02/2025] Open
Abstract
In adult mammals, including humans, neurons, and axons in the brain and spinal cord are inherently incapable of regenerating after injury. Studies of animals with innate capacity for regeneration are providing valuable insights into the mechanisms driving tissue healing. The aim of this review is to summarize recent data on regeneration mechanisms in the brain and spinal cord of zebrafish and neonatal mice. We infer that elucidating these mechanisms and understanding how and why they are lost in adult mammals will contribute to the development of strategies to promote central nervous system regeneration.
Collapse
Affiliation(s)
- Brianna R Cellini
- Department of Psychology and Neuroscience, Duke University, Durham, NC, 27710, USA
| | | | - Timothy D Faw
- Department of Orthopaedic Surgery, Duke University, Durham, NC, 27710, USA
- Duke Institute for Brain Sciences, Duke University, Durham, NC, 27710, USA
| | - Valentina Cigliola
- Department of Pharmacology, Vanderbilt University, Nashville, TN, 37232, USA.
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN, 37232, USA.
| |
Collapse
|
2
|
Gupta S, Hui SP. Epigenetic Cross-Talk Between Sirt1 and Dnmt1 Promotes Axonal Regeneration After Spinal Cord Injury in Zebrafish. Mol Neurobiol 2025; 62:2396-2419. [PMID: 39110393 DOI: 10.1007/s12035-024-04408-w] [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: 04/24/2024] [Accepted: 07/28/2024] [Indexed: 01/28/2025]
Abstract
Though spinal cord injury (SCI) causes irreversible sensory and motor impairments in human, adult zebrafish retain the potent regenerative capacity by injury-induced proliferation of central nervous system (CNS)-resident progenitor cells to develop new functional neurons at the lesion site. The hallmark of SCI in zebrafish lies in a series of changes in the epigenetic landscape, specifically DNA methylation and histone modifications. Decoding the post-SCI epigenetic modifications is therefore critical for the development of therapeutic remedies that boost SCI recovery process. Here, we have studied on Sirtuin1 (Sirt1), a non-classical histone deacetylase that potentially plays a critical role in neural progenitor cells (NPC) proliferation and axonal regrowth following SCI in zebrafish. We investigated the role of Sirt1 in NPC proliferation and axonal regrowth in response to injury in the regenerating spinal cord and found that Sirt1 is involved in the induction of NPC proliferation along with glial bridging during spinal cord regeneration. We also demonstrate that Sirt1 plays a pivotal role in regulating the HIPPO pathway through deacetylation-mediated inactivation of Dnmt1 and subsequent hypomethylation of yap1 promoter, leading to the induction of ctgfa expression, which drives the NPC proliferation and axonal regrowth to complete the regenerative process. In conclusion, our study reveals a novel cross-talk between two important epigenetic effectors, Sirt1 and Dnmt1, in the context of spinal cord regeneration, establishing a previously undisclosed relation between Sirt1 and Yap1 which provides a deeper understanding of the underlying mechanisms governing injury-induced NPC proliferation and axonal regrowth. Therefore, we have identified Sirt1 as a novel, major epigenetic regulator of spinal cord regeneration by modulating the HIPPO pathway in zebrafish.
Collapse
Affiliation(s)
- Samudra Gupta
- S. N. Pradhan Centre for Neurosciences, University of Calcutta, 35, Ballygunge Circular Road, Kolkata, 700019, India
| | - Subhra Prakash Hui
- S. N. Pradhan Centre for Neurosciences, University of Calcutta, 35, Ballygunge Circular Road, Kolkata, 700019, India.
| |
Collapse
|
3
|
Chen HY, Huang YC, Yeh TH, Chang CW, Shen YJ, Chen YC, Sun MQ, Cheng YC. Dtx2 Deficiency Induces Ependymo-Radial Glial Cell Proliferation and Improves Spinal Cord Motor Function Recovery. Stem Cells Dev 2024; 33:540-550. [PMID: 39001828 DOI: 10.1089/scd.2023.0247] [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] [Indexed: 07/15/2024] Open
Abstract
Traumatic injury to the spinal cord can lead to significant, permanent disability. Mammalian spinal cords are not capable of regeneration; in contrast, adult zebrafish are capable of such regeneration, fully recovering motor function. Understanding the mechanisms underlying zebrafish neuroregeneration may provide useful information regarding endogenous regenerative potential and aid in the development of therapeutic strategies in humans. DELTEX proteins (DTXs) regulate a variety of cellular processes. However, their role in neural regeneration has not been described. We found that zebrafish dtx2, encoding Deltex E3 ubiquitin ligase 2, is expressed in ependymo-radial glial cells in the adult spinal cord. After spinal cord injury, the heterozygous dtx2 mutant fish motor function recovered quicker than that of the wild-type controls. The mutant fish displayed increased ependymo-radial glial cell proliferation and augmented motor neuron formation. Moreover, her gene expression, downstream of Notch signaling, increased in Dtx2 mutants. Notch signaling inactivation by dominant-negative Rbpj abolished the increased ependymo-radial glia proliferation caused by Dtx2 deficiency. These results indicate that ependymo-radial glial proliferation is induced by Dtx2 deficiency by activating Notch-Rbpj signaling to improve spinal cord regeneration and motor function recovery.
Collapse
Affiliation(s)
- Hao-Yuan Chen
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Yin-Cheng Huang
- Department of Neurosurgery, Chang Gung Memorial Hospital at Linkou Medical Center, Taoyuan, Taiwan
- College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Tu-Hsueh Yeh
- Department of Neurology, Taipei Medical University Hospital, Taipei, Taiwan
- School of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Chia-Wei Chang
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Yang-Jin Shen
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan
- School of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Yi-Chieh Chen
- Department of Neurology, Chang Gung Memorial Hospital at Linkou Medical Center, Taoyuan, Taiwan
- Neuroscience Research Center, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Mu-Qun Sun
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Yi-Chuan Cheng
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Neuroscience Research Center, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| |
Collapse
|
4
|
Stefanova EE, Dychiao JVT, Chinn MC, Borhani M, Scott AL. P2X7 regulates ependymo-radial glial cell proliferation in adult Danio rerio following spinal cord injury. Biol Open 2024; 13:bio060270. [PMID: 38526172 PMCID: PMC11033521 DOI: 10.1242/bio.060270] [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: 12/06/2023] [Accepted: 03/11/2024] [Indexed: 03/26/2024] Open
Abstract
In contrast to mammals, zebrafish undergo successful neural regeneration following spinal cord injury. Spinal cord ependymo-radial glia (ERG) undergo injury-induced proliferation and neuronal differentiation to replace damaged cells and restore motor function. However, the molecular cues driving these processes remain elusive. Here, we demonstrate that the evolutionarily conserved P2X7 receptors are widely distributed on neurons and ERG within the zebrafish spinal cord. At the protein level, the P2X7 receptor expressed in zebrafish is a truncated splice variant of the full-length variant found in mammals. The protein expression of this 50 kDa isoform was significantly downregulated at 7 days post-injury (dpi) but returned to basal levels at 14 dpi when compared to naïve controls. Pharmacological activation of P2X7 following SCI resulted in a greater number of proliferating cells around the central canal by 7 dpi but did not affect neuronal differentiation at 14 dpi. Our findings suggest that unlike in mammals, P2X7 signaling may not play a maladaptive role following SCI in adult zebrafish and may also work to curb the proliferative response of ERG following injury.
Collapse
Affiliation(s)
- Eva E. Stefanova
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, Ontario, Canada
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | | | - Mavis C. Chinn
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, Ontario, Canada
| | - Matin Borhani
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Angela L. Scott
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, Ontario, Canada
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| |
Collapse
|
5
|
Purifoy EJ, Mruk K. Differential Roles of Diet on Development and Spinal Cord Regeneration in Larval Zebrafish. Zebrafish 2024; 21:214-222. [PMID: 38621204 PMCID: PMC11035855 DOI: 10.1089/zeb.2023.0042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/17/2024] Open
Abstract
The zebrafish is a powerful model organism for studying development and regeneration. However, there is a lack of a standardized reference diet for developmental and regeneration experiments. Most studies evaluate the rate of growth, survival, and fecundity. In this study, we compare three diets and their effects on growth and regeneration after a spinal cord injury (SCI). Fish were fed daily for 1 week with daily measurements of overall length and width of spinal injury. Fish fed a live rotifer diet grew 32%, whereas a commercially available diet only led to a 4% increase in body length. Similarly, differences in rate of regeneration were observed with over 80% of rotifer-fed larvae forming a glial bridge after injury compared to <10% of zebrafish fed with the commercial diet. Our data highlight the need for establishing a standardized diet for regeneration studies to improve research reproducibility.
Collapse
Affiliation(s)
- Emily J. Purifoy
- Wyoming Research Scholars Program and University of Wyoming, Laramie, Wyoming, USA
| | - Karen Mruk
- School of Pharmacy, University of Wyoming, Laramie, Wyoming, USA
- Department of Pharmacology and Toxicology, Brody School of Medicine, East Carolina University, Greenville, North Carolina, USA
| |
Collapse
|
6
|
Zeng CW, Tsai HJ. The Promising Role of a Zebrafish Model Employed in Neural Regeneration Following a Spinal Cord Injury. Int J Mol Sci 2023; 24:13938. [PMID: 37762240 PMCID: PMC10530783 DOI: 10.3390/ijms241813938] [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: 07/27/2023] [Revised: 09/07/2023] [Accepted: 09/09/2023] [Indexed: 09/29/2023] Open
Abstract
Spinal cord injury (SCI) is a devastating event that results in a wide range of physical impairments and disabilities. Despite the advances in our understanding of the biological response to injured tissue, no effective treatments are available for SCIs at present. Some studies have addressed this issue by exploring the potential of cell transplantation therapy. However, because of the abnormal microenvironment in injured tissue, the survival rate of transplanted cells is often low, thus limiting the efficacy of such treatments. Many studies have attempted to overcome these obstacles using a variety of cell types and animal models. Recent studies have shown the utility of zebrafish as a model of neural regeneration following SCIs, including the proliferation and migration of various cell types and the involvement of various progenitor cells. In this review, we discuss some of the current challenges in SCI research, including the accurate identification of cell types involved in neural regeneration, the adverse microenvironment created by SCIs, attenuated immune responses that inhibit nerve regeneration, and glial scar formation that prevents axonal regeneration. More in-depth studies are needed to fully understand the neural regeneration mechanisms, proteins, and signaling pathways involved in the complex interactions between the SCI microenvironment and transplanted cells in non-mammals, particularly in the zebrafish model, which could, in turn, lead to new therapeutic approaches to treat SCIs in humans and other mammals.
Collapse
Affiliation(s)
- Chih-Wei Zeng
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA;
- Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Huai-Jen Tsai
- Department of Life Science, Fu Jen Catholic University, New Taipei City 242062, Taiwan
| |
Collapse
|
7
|
Hu X, Xu W, Ren Y, Wang Z, He X, Huang R, Ma B, Zhao J, Zhu R, Cheng L. Spinal cord injury: molecular mechanisms and therapeutic interventions. Signal Transduct Target Ther 2023; 8:245. [PMID: 37357239 DOI: 10.1038/s41392-023-01477-6] [Citation(s) in RCA: 217] [Impact Index Per Article: 108.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Revised: 03/22/2023] [Accepted: 05/07/2023] [Indexed: 06/27/2023] Open
Abstract
Spinal cord injury (SCI) remains a severe condition with an extremely high disability rate. The challenges of SCI repair include its complex pathological mechanisms and the difficulties of neural regeneration in the central nervous system. In the past few decades, researchers have attempted to completely elucidate the pathological mechanism of SCI and identify effective strategies to promote axon regeneration and neural circuit remodeling, but the results have not been ideal. Recently, new pathological mechanisms of SCI, especially the interactions between immune and neural cell responses, have been revealed by single-cell sequencing and spatial transcriptome analysis. With the development of bioactive materials and stem cells, more attention has been focused on forming intermediate neural networks to promote neural regeneration and neural circuit reconstruction than on promoting axonal regeneration in the corticospinal tract. Furthermore, technologies to control physical parameters such as electricity, magnetism and ultrasound have been constantly innovated and applied in neural cell fate regulation. Among these advanced novel strategies and technologies, stem cell therapy, biomaterial transplantation, and electromagnetic stimulation have entered into the stage of clinical trials, and some of them have already been applied in clinical treatment. In this review, we outline the overall epidemiology and pathophysiology of SCI, expound on the latest research progress related to neural regeneration and circuit reconstruction in detail, and propose future directions for SCI repair and clinical applications.
Collapse
Affiliation(s)
- Xiao Hu
- Division of Spine, Department of Orthopaedics, Tongji Hospital, Tongji University School of Medicine, 200065, Shanghai, China
- Key Laboratory of Spine and Spinal cord Injury Repair and Regeneration (Tongji University), Ministry of Education, 200065, Shanghai, China
- Clinical Center For Brain And Spinal Cord Research, Tongji University, 200065, Shanghai, China
| | - Wei Xu
- Division of Spine, Department of Orthopaedics, Tongji Hospital, Tongji University School of Medicine, 200065, Shanghai, China
- Key Laboratory of Spine and Spinal cord Injury Repair and Regeneration (Tongji University), Ministry of Education, 200065, Shanghai, China
- Clinical Center For Brain And Spinal Cord Research, Tongji University, 200065, Shanghai, China
| | - Yilong Ren
- Division of Spine, Department of Orthopaedics, Tongji Hospital, Tongji University School of Medicine, 200065, Shanghai, China
- Key Laboratory of Spine and Spinal cord Injury Repair and Regeneration (Tongji University), Ministry of Education, 200065, Shanghai, China
- Clinical Center For Brain And Spinal Cord Research, Tongji University, 200065, Shanghai, China
| | - Zhaojie Wang
- Division of Spine, Department of Orthopaedics, Tongji Hospital, Tongji University School of Medicine, 200065, Shanghai, China
- Key Laboratory of Spine and Spinal cord Injury Repair and Regeneration (Tongji University), Ministry of Education, 200065, Shanghai, China
- Clinical Center For Brain And Spinal Cord Research, Tongji University, 200065, Shanghai, China
| | - Xiaolie He
- Division of Spine, Department of Orthopaedics, Tongji Hospital, Tongji University School of Medicine, 200065, Shanghai, China
- Key Laboratory of Spine and Spinal cord Injury Repair and Regeneration (Tongji University), Ministry of Education, 200065, Shanghai, China
- Clinical Center For Brain And Spinal Cord Research, Tongji University, 200065, Shanghai, China
| | - Runzhi Huang
- Division of Spine, Department of Orthopaedics, Tongji Hospital, Tongji University School of Medicine, 200065, Shanghai, China
- Key Laboratory of Spine and Spinal cord Injury Repair and Regeneration (Tongji University), Ministry of Education, 200065, Shanghai, China
- Clinical Center For Brain And Spinal Cord Research, Tongji University, 200065, Shanghai, China
| | - Bei Ma
- Division of Spine, Department of Orthopaedics, Tongji Hospital, Tongji University School of Medicine, 200065, Shanghai, China
- Key Laboratory of Spine and Spinal cord Injury Repair and Regeneration (Tongji University), Ministry of Education, 200065, Shanghai, China
- Clinical Center For Brain And Spinal Cord Research, Tongji University, 200065, Shanghai, China
| | - Jingwei Zhao
- Division of Spine, Department of Orthopaedics, Tongji Hospital, Tongji University School of Medicine, 200065, Shanghai, China
- Key Laboratory of Spine and Spinal cord Injury Repair and Regeneration (Tongji University), Ministry of Education, 200065, Shanghai, China
- Clinical Center For Brain And Spinal Cord Research, Tongji University, 200065, Shanghai, China
| | - Rongrong Zhu
- Division of Spine, Department of Orthopaedics, Tongji Hospital, Tongji University School of Medicine, 200065, Shanghai, China.
- Key Laboratory of Spine and Spinal cord Injury Repair and Regeneration (Tongji University), Ministry of Education, 200065, Shanghai, China.
- Clinical Center For Brain And Spinal Cord Research, Tongji University, 200065, Shanghai, China.
| | - Liming Cheng
- Division of Spine, Department of Orthopaedics, Tongji Hospital, Tongji University School of Medicine, 200065, Shanghai, China.
- Key Laboratory of Spine and Spinal cord Injury Repair and Regeneration (Tongji University), Ministry of Education, 200065, Shanghai, China.
- Clinical Center For Brain And Spinal Cord Research, Tongji University, 200065, Shanghai, China.
| |
Collapse
|
8
|
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
|
9
|
Miranda-Negrón Y, García-Arrarás JE. Radial glia and radial glia-like cells: Their role in neurogenesis and regeneration. Front Neurosci 2022; 16:1006037. [PMID: 36466166 PMCID: PMC9708897 DOI: 10.3389/fnins.2022.1006037] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Accepted: 10/21/2022] [Indexed: 01/25/2024] Open
Abstract
Radial glia is a cell type traditionally associated with the developing nervous system, particularly with the formation of cortical layers in the mammalian brain. Nonetheless, some of these cells, or closely related types, called radial glia-like cells are found in adult central nervous system structures, functioning as neurogenic progenitors in normal homeostatic maintenance and in response to injury. The heterogeneity of radial glia-like cells is nowadays being probed with molecular tools, primarily by the expression of specific genes that define cell types. Similar markers have identified radial glia-like cells in the nervous system of non-vertebrate organisms. In this review, we focus on adult radial glia-like cells in neurogenic processes during homeostasis and in response to injury. We highlight our results using a non-vertebrate model system, the echinoderm Holothuria glaberrima where we have described a radial glia-like cell that plays a prominent role in the regeneration of the holothurian central nervous system.
Collapse
Affiliation(s)
| | - José E. García-Arrarás
- Department of Biology, College of Natural Sciences, University of Puerto Rico, San Juan, Puerto Rico
| |
Collapse
|
10
|
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
|
11
|
Kiaie N, Gorabi AM, Loveless R, Teng Y, Jamialahmadi T, Sahebkar A. The regenerative potential of glial progenitor cells and reactive astrocytes in CNS injuries. Neurosci Biobehav Rev 2022; 140:104794. [PMID: 35902044 DOI: 10.1016/j.neubiorev.2022.104794] [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: 02/18/2022] [Revised: 07/20/2022] [Accepted: 07/21/2022] [Indexed: 10/16/2022]
Abstract
Cell therapeutic approaches focusing on the regeneration of damaged tissue have been a popular topic among researchers in recent years. In particular, self-repair scarring from the central nervous system (CNS) can significantly complicate the treatment of an injured patient. In CNS regeneration schemes, either glial progenitor cells or reactive glial cells have key roles to play. In this review, the contribution and underlying mechanisms of these progenitor/reactive glial cells during CNS regeneration are discussed, as well as their role in CNS-related diseases.
Collapse
Affiliation(s)
- Nasim Kiaie
- Research Center for Advanced Technologies in Cardiovascular Medicine, Tehran Heart Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Armita Mahdavi Gorabi
- Department of Tissue Engineering and Applied Cell Science, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Reid Loveless
- Department of Oral Biology and Diagnostic Sciences, Dental College of Georgia, Augusta University, Augusta, GA 30912, USA
| | - Yong Teng
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA, USA
| | - Tannaz Jamialahmadi
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Nutrition, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amirhossein Sahebkar
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
12
|
Corrigendum: Purinergic signaling systems across comparative models of spinal cord injury. Neural Regen Res 2022; 18:689-696. [PMID: 36018196 PMCID: PMC9727416 DOI: 10.4103/1673-5374.350234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
[This corrects the article DOI: 10.4103/1673-5374.338993].
Collapse
|
13
|
Radial Glia and Neuronal-like Ependymal Cells Are Present within the Spinal Cord of the Trunk (Body) in the Leopard Gecko (Eublepharis macularius). J Dev Biol 2022; 10:jdb10020021. [PMID: 35735912 PMCID: PMC9224675 DOI: 10.3390/jdb10020021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 05/12/2022] [Accepted: 05/20/2022] [Indexed: 11/28/2022] Open
Abstract
As is the case for many lizards, leopard geckos (Eublepharis macularius) can self-detach a portion of their tail to escape predation, and then regenerate a replacement complete with a spinal cord. Previous research has shown that endogenous populations of neural stem/progenitor cells (NSPCs) reside within the spinal cord of the original tail. In response to tail loss, these NSPCs are activated and contribute to regeneration. Here, we investigate whether similar populations of NSPCs are found within the spinal cord of the trunk (body). Using a long-duration 5-bromo-2′-deoxyuridine pulse-chase experiment, we determined that a population of cells within the ependymal layer are label-retaining following a 20-week chase. Tail loss does not significantly alter rates of ependymal cell proliferation within the trunk spinal cord. Ependymal cells of the trunk spinal cord express SOX2 and represent at least two distinct cell populations: radial glial-like (glial fibrillary acidic protein- and Vimentin-expressing) cells; and neuronal-like (HuCD-expressing) cells. Taken together, these data demonstrate that NSPCs of the trunk spinal cord closely resemble those of the tail and support the use of the tail spinal cord as a less invasive proxy for body spinal cord injury investigations.
Collapse
|
14
|
Slater PG, Domínguez-Romero ME, Villarreal M, Eisner V, Larraín J. Mitochondrial function in spinal cord injury and regeneration. Cell Mol Life Sci 2022; 79:239. [PMID: 35416520 PMCID: PMC11072423 DOI: 10.1007/s00018-022-04261-x] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Revised: 03/16/2022] [Accepted: 03/17/2022] [Indexed: 12/21/2022]
Abstract
Many people around the world suffer from some form of paralysis caused by spinal cord injury (SCI), which has an impact on quality and life expectancy. The spinal cord is part of the central nervous system (CNS), which in mammals is unable to regenerate, and to date, there is a lack of full functional recovery therapies for SCI. These injuries start with a rapid and mechanical insult, followed by a secondary phase leading progressively to greater damage. This secondary phase can be potentially modifiable through targeted therapies. The growing literature, derived from mammalian and regenerative model studies, supports a leading role for mitochondria in every cellular response after SCI: mitochondrial dysfunction is the common event of different triggers leading to cell death, cellular metabolism regulates the immune response, mitochondrial number and localization correlate with axon regenerative capacity, while mitochondrial abundance and substrate utilization regulate neural stem progenitor cells self-renewal and differentiation. Herein, we present a comprehensive review of the cellular responses during the secondary phase of SCI, the mitochondrial contribution to each of them, as well as evidence of mitochondrial involvement in spinal cord regeneration, suggesting that a more in-depth study of mitochondrial function and regulation is needed to identify potential targets for SCI therapeutic intervention.
Collapse
Affiliation(s)
- Paula G Slater
- Center for Aging and Regeneration, Departamento de Biología Celular Y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Alameda 340, 8331150, Santiago, Chile.
| | - Miguel E Domínguez-Romero
- Center for Aging and Regeneration, Departamento de Biología Celular Y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Alameda 340, 8331150, Santiago, Chile
| | - Maximiliano Villarreal
- Center for Aging and Regeneration, Departamento de Biología Celular Y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Alameda 340, 8331150, Santiago, Chile
| | - Verónica Eisner
- Center for Aging and Regeneration, Departamento de Biología Celular Y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Alameda 340, 8331150, Santiago, Chile
- Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Alameda 340, 8331150, Santiago, Chile
| | - Juan Larraín
- Center for Aging and Regeneration, Departamento de Biología Celular Y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Alameda 340, 8331150, Santiago, Chile
| |
Collapse
|
15
|
Vanni V, Salonna M, Gasparini F, Martini M, Anselmi C, Gissi C, Manni L. Yamanaka Factors in the Budding Tunicate Botryllus schlosseri Show a Shared Spatio-Temporal Expression Pattern in Chordates. Front Cell Dev Biol 2022; 10:782722. [PMID: 35342743 PMCID: PMC8948423 DOI: 10.3389/fcell.2022.782722] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Accepted: 02/09/2022] [Indexed: 01/22/2023] Open
Abstract
In vertebrates, the four transcription factors Sox2, c-Myc, Pou5f1 and Klf4 are involved in the differentiation of several tissues during vertebrate embryogenesis; moreover, they are normally co-expressed in embryonic stem cells and play roles in pluripotency, self-renewal, and maintenance of the undifferentiated state in adult cells. The in vitro forced co-expression of these factors, named Yamanaka factors (YFs), induces pluripotency in human or mouse fibroblasts. Botryllus schlosseri is a colonial tunicate undergoing continuous stem cell-mediated asexual development, providing a valuable model system for the study of pluripotency in the closest living relatives of vertebrates. In this study, we identified B. schlosseri orthologs of human Sox2 and c-Myc genes, as well as the closest homologs of the vertebrate-specific Pou5f1 gene, through an in-depth evolutionary analysis of the YF gene families in tunicates and other deuterostomes. Then, we studied the expression of these genes during the asexual cycle of B. schlosseri using in situ hybridization in order to investigate their possible involvement in tissue differentiation and in pluripotency maintenance. Our results show a shared spatio-temporal expression pattern consistent with the reported functions of these genes in invertebrate and vertebrate embryogenesis. Moreover, Myc, SoxB1 and Pou3 were expressed in candidate stem cells residing in their niches, while Pou2 was found expressed exclusively in the immature previtellogenic oocytes, both in gonads and circulating in the colonial vascular system. Our data suggest that Myc, SoxB1 and Pou3 may be individually involved in the differentiation of the same territories seen in other chordates, and that, together, they may play a role in stemness even in this colonial ascidian.
Collapse
Affiliation(s)
- Virginia Vanni
- Department of Biology, University of Padova, Padova, Italy
| | - Marika Salonna
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari "Aldo Moro", Bari, Italy
| | | | | | - Chiara Anselmi
- Stanford University, Hopkins Marine Station, Pacific Grove, CA, United States.,Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, United States
| | - Carmela Gissi
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari "Aldo Moro", Bari, Italy.,IBIOM, Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies, Consiglio Nazionale Delle Ricerche, Bari, Italy.,CoNISMa, Consorzio Nazionale Interuniversitario per le Scienze Del Mare, Roma, Italy
| | - Lucia Manni
- Department of Biology, University of Padova, Padova, Italy
| |
Collapse
|
16
|
Zeng CW, Sheu JC, Tsai HJ. Hypoxia-Responsive Subtype Cells Differentiate Into Neurons in the Brain of Zebrafish Embryos Exposed to Hypoxic Stress. Cell Transplant 2022; 31:9636897221077930. [PMID: 35225023 PMCID: PMC8894973 DOI: 10.1177/09636897221077930] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022] Open
Abstract
Severe hypoxia results in complete loss of central nervous system (CNS) function in mammals, while several other vertebrates, such as zebrafish, can regenerate after hypoxia-induced injury of CNS. Since the cellular mechanism involved in this remarkable feature of other vertebrates is still unclear, we studied the cellular regeneration of zebrafish brain, employing zebrafish embryos from transgenic line huORFZ exposed to hypoxia and then oxygen recovery. GFP-expressing cells, identified in some cells of the CNS, including some brain cells, were termed as hypoxia-responsive recovering cells (HrRCs). After hypoxia, HrRCs did not undergo apoptosis, while most non-GFP-expressing cells, including neurons, did. Major cell types of HrRCs found in the brain of zebrafish embryos induced by hypoxic stress were neural stem/progenitor cells (NSPCs) and radial glia cells (RGs), that is, subtypes of NSPCs (NSPCs-HrRCs) and RGs (RGs-HrRCs) that were induced by and sensitively responded to hypoxic stress. Interestingly, among HrRCs, subtypes of NSPCs- or RGs-HrRCs could proliferate and differentiate into early neurons during oxygen recovery, suggesting that these subtype cells might play a critical role in brain regeneration of zebrafish embryos after hypoxic stress.
Collapse
Affiliation(s)
- Chih-Wei Zeng
- Institute of Molecular and Cellular Biology, College of Life Science, National Taiwan University, Taipei.,Liver Disease Prevention and Treatment Research Foundation, Taipei
| | - Jin-Chuan Sheu
- Liver Disease Prevention and Treatment Research Foundation, Taipei
| | - Huai-Jen Tsai
- School of Medicine, Fu Jen Catholic University, New Taipei City.,Department of Life Science, Fu Jen Catholic University, New Taipei City
| |
Collapse
|
17
|
Camphausen R, Cuevas Á, Duempelmann L, Terborg RA, Wajs E, Tisa S, Ruggeri A, Cusini I, Steinlechner F, Pruneri V. A quantum-enhanced wide-field phase imager. SCIENCE ADVANCES 2021; 7:eabj2155. [PMID: 34788099 PMCID: PMC8598016 DOI: 10.1126/sciadv.abj2155] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/15/2023]
Abstract
Quantum techniques can be used to enhance the signal-to-noise ratio in optical imaging. Leveraging the latest advances in single-photon avalanche diode array cameras and multiphoton detection techniques, here, we introduce a supersensitive phase imager, which uses space-polarization hyperentanglement to operate over a large field of view without the need of scanning operation. We show quantum-enhanced imaging of birefringent and nonbirefringent phase samples over large areas, with sensitivity improvements over equivalent classical measurements carried out with equal number of photons. The potential applicability is demonstrated by imaging a biomedical protein microarray sample. Our technology is inherently scalable to high-resolution images and represents an essential step toward practical quantum-enhanced imaging.
Collapse
Affiliation(s)
- Robin Camphausen
- ICFO-Institut de Ciencies Fotoniques, The Barcelona Institute of Science and Technology, Av. Carl Friedrich Gauss, 3, 08860 Castelldefels, Barcelona, Spain
- Corresponding author. (R.C.); (Á.C.); (V.P.)
| | - Álvaro Cuevas
- ICFO-Institut de Ciencies Fotoniques, The Barcelona Institute of Science and Technology, Av. Carl Friedrich Gauss, 3, 08860 Castelldefels, Barcelona, Spain
- Corresponding author. (R.C.); (Á.C.); (V.P.)
| | - Luc Duempelmann
- ICFO-Institut de Ciencies Fotoniques, The Barcelona Institute of Science and Technology, Av. Carl Friedrich Gauss, 3, 08860 Castelldefels, Barcelona, Spain
| | - Roland A. Terborg
- ICFO-Institut de Ciencies Fotoniques, The Barcelona Institute of Science and Technology, Av. Carl Friedrich Gauss, 3, 08860 Castelldefels, Barcelona, Spain
| | - Ewelina Wajs
- ICFO-Institut de Ciencies Fotoniques, The Barcelona Institute of Science and Technology, Av. Carl Friedrich Gauss, 3, 08860 Castelldefels, Barcelona, Spain
| | - Simone Tisa
- Micro Photon Device SRL, Via Waltraud Gebert Deeg 3f, 39100 Bolzano, Italy
| | - Alessandro Ruggeri
- Micro Photon Device SRL, Via Waltraud Gebert Deeg 3f, 39100 Bolzano, Italy
| | - Iris Cusini
- Dipartimento di Elettronica, Informazione e Bioingegneria, Politecnico di Milano, Via Giuseppe Ponzio, 34, 20133 Milano, Italy
| | - Fabian Steinlechner
- Fraunhofer Institute for Applied Optics and Precision Engineering IOF, Albert-Einstein-Str. 7, 07745 Jena, Germany
- Abbe Center of Photonics, Friedrich Schiller University Jena, Albert-Einstein-Str. 6, 07745 Jena, Germany
| | - Valerio Pruneri
- ICFO-Institut de Ciencies Fotoniques, The Barcelona Institute of Science and Technology, Av. Carl Friedrich Gauss, 3, 08860 Castelldefels, Barcelona, Spain
- ICREA-Institució Catalana de Recerca i Estudis Avançats, Passeig Lluís Companys 23, 08010 Barcelona, Spain
- Corresponding author. (R.C.); (Á.C.); (V.P.)
| |
Collapse
|
18
|
Vandestadt C, Vanwalleghem GC, Khabooshan MA, Douek AM, Castillo HA, Li M, Schulze K, Don E, Stamatis SA, Ratnadiwakara M, Änkö ML, Scott EK, Kaslin J. RNA-induced inflammation and migration of precursor neurons initiates neuronal circuit regeneration in zebrafish. Dev Cell 2021; 56:2364-2380.e8. [PMID: 34428400 DOI: 10.1016/j.devcel.2021.07.021] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Revised: 06/18/2021] [Accepted: 07/27/2021] [Indexed: 11/29/2022]
Abstract
Tissue regeneration and functional restoration after injury are considered as stem- and progenitor-cell-driven processes. In the central nervous system, stem cell-driven repair is slow and problematic because function needs to be restored rapidly for vital tasks. In highly regenerative vertebrates, such as zebrafish, functional recovery is rapid, suggesting a capability for fast cell production and functional integration. Surprisingly, we found that migration of dormant "precursor neurons" to the injury site pioneers functional circuit regeneration after spinal cord injury and controls the subsequent stem-cell-driven repair response. Thus, the precursor neurons make do before the stem cells make new. Furthermore, RNA released from the dying or damaged cells at the site of injury acts as a signal to attract precursor neurons for repair. Taken together, our data demonstrate an unanticipated role of neuronal migration and RNA as drivers of neural repair.
Collapse
Affiliation(s)
- Celia Vandestadt
- Australian Regenerative Medicine Institute, Monash University, Clayton VIC, 3800, Australia
| | - Gilles C Vanwalleghem
- The Queensland Brain Institute, the University of Queensland, St. Lucia, QLD, Australia
| | - Mitra Amiri Khabooshan
- Australian Regenerative Medicine Institute, Monash University, Clayton VIC, 3800, Australia
| | - Alon M Douek
- Australian Regenerative Medicine Institute, Monash University, Clayton VIC, 3800, Australia
| | - Hozana Andrade Castillo
- Australian Regenerative Medicine Institute, Monash University, Clayton VIC, 3800, Australia; Brazilian Biosciences National Laboratory, Brazilian Centre for Research in Energy and Materials, Campinas CEP 13083-100, Brazil
| | - Mei Li
- Australian Regenerative Medicine Institute, Monash University, Clayton VIC, 3800, Australia
| | - Keith Schulze
- Monash Micro Imaging, Monash University, Monash University, Clayton, VIC 3800, Australia
| | - Emily Don
- Centre for Motor Neuron Disease Research, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Macquarie University, Macquarie Park, NSW 2109, Australia
| | | | - Madara Ratnadiwakara
- Centre for Reproductive Health and Center for Cancer Research, Hudson Institute of Medical Research, Clayton, VIC 3168, Australia
| | - Minna-Liisa Änkö
- Centre for Reproductive Health and Center for Cancer Research, Hudson Institute of Medical Research, Clayton, VIC 3168, Australia; Department of Molecular and Translational Sciences, Monash University, Clayton, VIC 3800, Australia
| | - Ethan K Scott
- The Queensland Brain Institute, the University of Queensland, St. Lucia, QLD, Australia
| | - Jan Kaslin
- Australian Regenerative Medicine Institute, Monash University, Clayton VIC, 3800, Australia.
| |
Collapse
|
19
|
Farías-Serratos BM, Lazcano I, Villalobos P, Darras VM, Orozco A. Thyroid hormone deficiency during zebrafish development impairs central nervous system myelination. PLoS One 2021; 16:e0256207. [PMID: 34403440 PMCID: PMC8370640 DOI: 10.1371/journal.pone.0256207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Accepted: 08/02/2021] [Indexed: 11/19/2022] Open
Abstract
Thyroid hormones are messengers that bind to specific nuclear receptors and regulate a wide range of physiological processes in the early stages of vertebrate embryonic development, including neurodevelopment and myelogenesis. We here tested the effects of reduced T3 availability upon the myelination process by treating zebrafish embryos with low concentrations of iopanoic acid (IOP) to block T4 to T3 conversion. Black Gold II staining showed that T3 deficiency reduced the myelin density in the forebrain, midbrain, hindbrain and the spinal cord at 3 and 7 dpf. These observations were confirmed in 3 dpf mbp:egfp transgenic zebrafish, showing that the administration of IOP reduced the fluorescent signal in the brain. T3 rescue treatment restored brain myelination and reversed the changes in myelin-related gene expression induced by IOP exposure. NG2 immunostaining revealed that T3 deficiency reduced the amount of oligodendrocyte precursor cells in 3 dpf IOP-treated larvae. Altogether, the present results show that inhibition of T4 to T3 conversion results in hypomyelination, suggesting that THs are part of the key signaling molecules that control the timing of oligodendrocyte differentiation and myelin synthesis from very early stages of brain development.
Collapse
Affiliation(s)
| | - Iván Lazcano
- Instituto de Neurobiología, Universidad Nacional Autónoma de México (UNAM), Querétaro, Qro., México
| | - Patricia Villalobos
- Instituto de Neurobiología, Universidad Nacional Autónoma de México (UNAM), Querétaro, Qro., México
| | - Veerle M. Darras
- Instituto de Neurobiología, Universidad Nacional Autónoma de México (UNAM), Querétaro, Qro., México
- Biology Department, Laboratory of Comparative Endocrinology, KU Leuven, Leuven, Belgium
| | - Aurea Orozco
- Instituto de Neurobiología, Universidad Nacional Autónoma de México (UNAM), Querétaro, Qro., México
- * E-mail:
| |
Collapse
|
20
|
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
|
21
|
Edwards-Faret G, González-Pinto K, Cebrián-Silla A, Peñailillo J, García-Verdugo JM, Larraín J. Cellular response to spinal cord injury in regenerative and non-regenerative stages in Xenopus laevis. Neural Dev 2021; 16:2. [PMID: 33526076 PMCID: PMC7852093 DOI: 10.1186/s13064-021-00152-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2020] [Accepted: 01/14/2021] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND The efficient regenerative abilities at larvae stages followed by a non-regenerative response after metamorphosis in froglets makes Xenopus an ideal model organism to understand the cellular responses leading to spinal cord regeneration. METHODS We compared the cellular response to spinal cord injury between the regenerative and non-regenerative stages of Xenopus laevis. For this analysis, we used electron microscopy, immunofluorescence and histological staining of the extracellular matrix. We generated two transgenic lines: i) the reporter line with the zebrafish GFAP regulatory regions driving the expression of EGFP, and ii) a cell specific inducible ablation line with the same GFAP regulatory regions. In addition, we used FACS to isolate EGFP+ cells for RNAseq analysis. RESULTS In regenerative stage animals, spinal cord regeneration triggers a rapid sealing of the injured stumps, followed by proliferation of cells lining the central canal, and formation of rosette-like structures in the ablation gap. In addition, the central canal is filled by cells with similar morphology to the cells lining the central canal, neurons, axons, and even synaptic structures. Regeneration is almost completed after 20 days post injury. In non-regenerative stage animals, mostly damaged tissue was observed, without clear closure of the stumps. The ablation gap was filled with fibroblast-like cells, and deposition of extracellular matrix components. No reconstruction of the spinal cord was observed even after 40 days post injury. Cellular markers analysis confirmed these histological differences, a transient increase of vimentin, fibronectin and collagen was detected in regenerative stages, contrary to a sustained accumulation of most of these markers, including chondroitin sulfate proteoglycans in the NR-stage. The zebrafish GFAP transgenic line was validated, and we have demonstrated that is a very reliable and new tool to study the role of neural stem progenitor cells (NSPCs). RNASeq of GFAP::EGFP cells has allowed us to clearly demonstrate that indeed these cells are NSPCs. On the contrary, the GFAP::EGFP transgene is mainly expressed in astrocytes in non-regenerative stages. During regenerative stages, spinal cord injury activates proliferation of NSPCs, and we found that are mainly differentiated into neurons and glial cells. Specific ablation of these cells abolished proper regeneration, confirming that NSPCs cells are necessary for functional regeneration of the spinal cord. CONCLUSIONS The cellular response to spinal cord injury in regenerative and non-regenerative stages is profoundly different between both stages. A key hallmark of the regenerative response is the activation of NSPCs, which massively proliferate, and are differentiated into neurons to reconstruct the spinal cord. Also very notably, no glial scar formation is observed in regenerative stages, but a transient, glial scar-like structure is formed in non-regenerative stage animals.
Collapse
Affiliation(s)
- Gabriela Edwards-Faret
- Center for Aging and Regeneration, Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Alameda 340, Santiago, Chile
| | - Karina González-Pinto
- Center for Aging and Regeneration, Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Alameda 340, Santiago, Chile
| | - Arantxa Cebrián-Silla
- Laboratorio de Neurobiología Comparada, Instituto Cavanilles, Universidad de Valencia, CIBERNED, 46980, Valencia, Spain
| | - Johany Peñailillo
- Center for Aging and Regeneration, Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Alameda 340, Santiago, Chile
| | - José Manuel García-Verdugo
- Laboratorio de Neurobiología Comparada, Instituto Cavanilles, Universidad de Valencia, CIBERNED, 46980, Valencia, Spain
| | - Juan Larraín
- Center for Aging and Regeneration, Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Alameda 340, Santiago, Chile.
| |
Collapse
|
22
|
Panlilio JM, Aluru N, Hahn ME. Developmental Neurotoxicity of the Harmful Algal Bloom Toxin Domoic Acid: Cellular and Molecular Mechanisms Underlying Altered Behavior in the Zebrafish Model. ENVIRONMENTAL HEALTH PERSPECTIVES 2020; 128:117002. [PMID: 33147070 PMCID: PMC7641300 DOI: 10.1289/ehp6652] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
BACKGROUND Harmful algal blooms (HABs) produce potent neurotoxins that threaten human health, but current regulations may not be protective of sensitive populations. Early life exposure to low levels of the HAB toxin domoic acid (DomA) produces long-lasting behavioral deficits in rodent and primate models; however, the mechanisms involved are unknown. The zebrafish is a powerful in vivo vertebrate model system for exploring cellular processes during development and thus may help to elucidate mechanisms of DomA developmental neurotoxicity. OBJECTIVES We used the zebrafish model to investigate how low doses of DomA affect the developing nervous system, including windows of susceptibility to DomA exposure, structural and molecular changes in the nervous system, and the link to behavioral alterations. METHODS To identify potential windows of susceptibility, DomA (0.09-0.18 ng) was delivered to zebrafish through caudal vein microinjection during distinct periods in early neurodevelopment. Following exposure, structural and molecular targets were identified using live imaging of transgenic fish and RNA sequencing. To assess the functional consequences of exposures, we quantified startle behavior in response to acoustic/vibrational stimuli. RESULTS Larvae exposed to DomA at 2 d postfertilization (dpf), but not at 1 or 4 dpf, showed consistent deficits in startle behavior at 7 dpf, including lower responsiveness and altered kinematics. Similarly, myelination in the spinal cord was disorganized after exposure at 2 dpf but not 1 or 4 dpf. Time-lapse imaging revealed disruption of the initial stages of myelination. DomA exposure at 2 dpf down-regulated genes required for maintaining myelin structure and the axonal cytoskeleton. DISCUSSION These results in zebrafish reveal a developmental window of susceptibility to DomA-induced behavioral deficits and identify altered gene expression and disrupted myelin structure as possible mechanisms. The results establish a zebrafish model for investigating the mechanisms of developmental DomA toxicity, including effects with potential relevance to exposed sensitive human populations. https://doi.org/10.1289/EHP6652.
Collapse
Affiliation(s)
- Jennifer M. Panlilio
- Biology Department, Woods Hole Oceanographic Institution (WHOI), Woods Hole, Massachusetts, USA
- Massachusetts Institute of Technology (MIT)–WHOI Joint Graduate Program in Oceanography and Oceanographic Engineering, Department of Earth, Atmospheric and Planetary Sciences, MIT, Cambridge, Massachusetts, USA
- Woods Hole Center for Oceans and Human Health, WHOI, Woods Hole, Massachusetts, USA
| | - Neelakanteswar Aluru
- Biology Department, Woods Hole Oceanographic Institution (WHOI), Woods Hole, Massachusetts, USA
- Woods Hole Center for Oceans and Human Health, WHOI, Woods Hole, Massachusetts, USA
| | - Mark E. Hahn
- Biology Department, Woods Hole Oceanographic Institution (WHOI), Woods Hole, Massachusetts, USA
- Woods Hole Center for Oceans and Human Health, WHOI, Woods Hole, Massachusetts, USA
| |
Collapse
|
23
|
Wang N, He L, Yang Y, Li S, Chen Y, Tian Z, Ji Y, Wang Y, Pang M, Wang Y, Liu B, Rong L. Integrated analysis of competing endogenous RNA (ceRNA) networks in subacute stage of spinal cord injury. Gene 2019; 726:144171. [PMID: 31669638 DOI: 10.1016/j.gene.2019.144171] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2019] [Revised: 10/13/2019] [Accepted: 10/15/2019] [Indexed: 12/26/2022]
Abstract
This study aims to investigate the genetic and epigenetic mechanisms involved in the pathogenesis of subacute stage of spinal cord injury (SCI). Gene-expression datasets associated with SCI were downloaded from the Gene Expression Omnibus (GEO) database, and differential expression analyses were performed in order to identify differentially expressed genes (DEGs). Multiple network types were constructed and analyzed, including protein-protein-interaction (PPI) network, miRNA-target network, lncRNA-associated competing endogenous RNA (ceRNA) network, and miRNA-transcription factor (TF)-target network. Cluster analyses were performed to identify significant modules. To verify the prediction accuracy of the in-silico identified molecules, qRT-PCR experiments were conducted. The results depicted the Ywhae gene as the hub gene with the highest degree in the PPI network. The ceRNA network identified specific genes (Flna, ID3, and HK2), miRNAs (miR-16-5p, miR-1958, and miR-185-5p), and lncRNAs (Neat1, Xist, and Malat1) as playing critical regulating roles in the pathological mechanisms of SCI. The miRNA-TF-gene interaction network identified four important TFs (Sp1, Trp53, Jun, and Rela). The miRNA-gene-TF interaction loops from the significant modules indicated that miR-325-3p can interact with the Asah1 gene and TF-Sp1 by forming a closed loop. The qRT-PCR experiments verified four selected genes (Flna, ID3, HK2, and Ywhae) and two selected TFs (Jun, and Sp1) as significantly up-regulated following SCI. The results indicated that four genes (Flna, ID3, HK2, and Ywhae), four transcription factors (Sp1, Trp53, Jun, and RelA), two miRNAs (miR-16-5p and miR-325-3p), and three lncRNAs (Neat1, Xist, and Malat1) are likely to be involved in the molecular mechanisms underlying the subacute stage of SCI. These findings uncover putative pathogenic mechanisms involved in SCI and might bear translation significance for future research towards therapeutic development.
Collapse
Affiliation(s)
- Nanxiang Wang
- Department of Spine Surgery, The Third Affiliated Hospital of Sun Yat-Sen University, No. 600 Tianhe Road, Tianhe District, Guangzhou, Guangdong Province, People's Republic of China
| | - Lei He
- Department of Spine Surgery, The Third Affiliated Hospital of Sun Yat-Sen University, No. 600 Tianhe Road, Tianhe District, Guangzhou, Guangdong Province, People's Republic of China
| | - Yang Yang
- Department of Spine Surgery, The Third Affiliated Hospital of Sun Yat-Sen University, No. 600 Tianhe Road, Tianhe District, Guangzhou, Guangdong Province, People's Republic of China
| | - Simin Li
- Department of Cariology, Endodontology and Periodontology, University Leipzig, Liebigstr. 12, 04103 Leipzig, Germany
| | - Yuyong Chen
- Department of Spine Surgery, The Third Affiliated Hospital of Sun Yat-Sen University, No. 600 Tianhe Road, Tianhe District, Guangzhou, Guangdong Province, People's Republic of China
| | - Zhenming Tian
- Department of Spine Surgery, The Third Affiliated Hospital of Sun Yat-Sen University, No. 600 Tianhe Road, Tianhe District, Guangzhou, Guangdong Province, People's Republic of China
| | - Ye Ji
- Department of Orthopaedics, The Second Affiliated Hospital of Harbin Medical University, No. 246 Xuefu Road, Nangang District, Harbin, Heilongjiang Province, People's Republic of China
| | - Yufu Wang
- Department of Orthopaedics, The Second Affiliated Hospital of Harbin Medical University, No. 246 Xuefu Road, Nangang District, Harbin, Heilongjiang Province, People's Republic of China
| | - Mao Pang
- Department of Spine Surgery, The Third Affiliated Hospital of Sun Yat-Sen University, No. 600 Tianhe Road, Tianhe District, Guangzhou, Guangdong Province, People's Republic of China
| | - Yang Wang
- Department of Spine Surgery, The Third Affiliated Hospital of Sun Yat-Sen University, No. 600 Tianhe Road, Tianhe District, Guangzhou, Guangdong Province, People's Republic of China
| | - Bin Liu
- Department of Spine Surgery, The Third Affiliated Hospital of Sun Yat-Sen University, No. 600 Tianhe Road, Tianhe District, Guangzhou, Guangdong Province, People's Republic of China.
| | - Limin Rong
- Department of Spine Surgery, The Third Affiliated Hospital of Sun Yat-Sen University, No. 600 Tianhe Road, Tianhe District, Guangzhou, Guangdong Province, People's Republic of China.
| |
Collapse
|
24
|
Subbarayan R, Barathidasan R, Raja STK, Arumugam G, Kuruvilla S, Shanthi P, Ranga Rao S. Human gingival derived neuronal cells in the optimized caffeic acid hydrogel for hemitransection spinal cord injury model. J Cell Biochem 2019; 121:2077-2088. [PMID: 31646674 DOI: 10.1002/jcb.29452] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Accepted: 10/08/2019] [Indexed: 01/17/2023]
Abstract
Spinal cord injury induces scar formation causes axonal damage that leads to the degeneration of axonal function. Still, there is no robust conceptual design to regenerate the damaged axon after spinal injury. Therefore, the present study demonstrates that human gingival derived neuronal stem cells (GNSCs) transplants in the injectable caffeic acid bioconjugated hydrogel (CBGH) helps to bridge the cavity and promote the engraftment and repopulation of transplants in the injured spinal tissue. Our study reports that the bioluminescence imaging in vivo imaging system (IVIS) provides a satisfactory progression in CBGH-GNSCs transplants compare to lesion control and CBGH alone. Immune regulators interleukin-6 (IL-6), tumor necrosis factor-α, neutrophil elastase are decreased, IL-10 is increased. Likewise, immunostaining (TAU/TUJ-1, SOX-2/NeuN, MAP-2/PSD93, NSE, S100b, and GFAP) shown repopulated cells. Also, TRA-1-81 expression confirms the absence of immune rejection in the CBGH-GNSCs transplants. However, locomotor recovery test, gene (IL-6, CASPASE3, p14-ARF, VEGF, LCAM, BDNF, NT3, NGN2, TrKc, FGF2, Sox-2, TUJ-1, MAP-2, Nestin, and NeuN) and protein expression (TAU, TUJ-1, SOX-2 MAP-2, PSD93, NeuN, TRA-1-81, GFAP, TAU, and MBP) shows functional improvements in the CBGH-GNSCs group. Further, GABA and glutamine level demonstrates the new synaptic vesicle formation. Hence, the CBGH scaffold enhances GNSCs transplants to restore the injured spinal tissue.
Collapse
Affiliation(s)
- Rajasekaran Subbarayan
- Centre for Regenerative Medicine and Stem Cell Research, Central Research Facility, Sri Ramachandra Medical College and Research Institute, Chennai, India
| | - Rajamani Barathidasan
- Centre for toxicology and Developmental Research (CEFT), Sri Ramachandra Medical College and Research Institute, Chennai, India
| | - Selvaraj T K Raja
- Biological Material Laboratory, Central Leather Research Institute Adyar, Chennai, Tamil Nadu, India
| | - Gnanamani Arumugam
- Biological Material Laboratory, Central Leather Research Institute Adyar, Chennai, Tamil Nadu, India
| | | | - Palanivelu Shanthi
- Department of Pathology, Dr ALM PGIBMS, University of Madras Taramani Campus, Chennai, India
| | - Suresh Ranga Rao
- Department of Periodontology and Implantology, Faculty of Dental Sciences and Centre for Regenerative Medicine and Stem Cell Research, Sri Ramachandra Medical College and Research Institute, Chennai, India
| |
Collapse
|
25
|
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
|
26
|
Becker CG, Becker T, Hugnot JP. The spinal ependymal zone as a source of endogenous repair cells across vertebrates. Prog Neurobiol 2018; 170:67-80. [DOI: 10.1016/j.pneurobio.2018.04.002] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2017] [Revised: 01/30/2018] [Accepted: 04/05/2018] [Indexed: 02/07/2023]
|
27
|
Mechanical Mapping of Spinal Cord Growth and Repair in Living Zebrafish Larvae by Brillouin Imaging. Biophys J 2018; 115:911-923. [PMID: 30122291 PMCID: PMC6127462 DOI: 10.1016/j.bpj.2018.07.027] [Citation(s) in RCA: 103] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Revised: 07/03/2018] [Accepted: 07/25/2018] [Indexed: 12/21/2022] Open
Abstract
The mechanical properties of biological tissues are increasingly recognized as important factors in developmental and pathological processes. Most existing mechanical measurement techniques either necessitate destruction of the tissue for access or provide insufficient spatial resolution. Here, we show for the first time to our knowledge a systematic application of confocal Brillouin microscopy to quantitatively map the mechanical properties of spinal cord tissues during biologically relevant processes in a contact-free and nondestructive manner. Living zebrafish larvae were mechanically imaged in all anatomical planes during development and after spinal cord injury. These experiments revealed that Brillouin microscopy is capable of detecting the mechanical properties of distinct anatomical structures without interfering with the animal’s natural development. The Brillouin shift within the spinal cord remained comparable during development and transiently decreased during the repair processes after spinal cord transection. By taking into account the refractive index distribution, we explicitly determined the apparent longitudinal modulus and viscosity of different larval zebrafish tissues. Importantly, mechanical properties differed between tissues in situ and in excised slices. The presented work constitutes the first step toward an in vivo assessment of spinal cord tissue mechanics during regeneration, provides a methodical basis to identify key determinants of mechanical tissue properties, and allows us to test their relative importance in combination with biochemical and genetic factors during developmental and regenerative processes.
Collapse
|
28
|
Berg EM, Bertuzzi M, Ampatzis K. Complementary expression of calcium binding proteins delineates the functional organization of the locomotor network. Brain Struct Funct 2018; 223:2181-2196. [PMID: 29423637 PMCID: PMC5968073 DOI: 10.1007/s00429-018-1622-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Accepted: 01/30/2018] [Indexed: 12/18/2022]
Abstract
Neuronal networks in the spinal cord generate and execute all locomotor-related movements by transforming descending signals from supraspinal areas into appropriate rhythmic activity patterns. In these spinal networks, neurons that arise from the same progenitor domain share similar distribution patterns, neurotransmitter phenotypes, morphological and electrophysiological features. However, subgroups of them participate in different functionally distinct microcircuits to produce locomotion at different speeds and of different modalities. To better understand the nature of this network complexity, here we characterized the distribution of parvalbumin (PV), calbindin D-28 k (CB) and calretinin (CR) which are regulators of intracellular calcium levels and can serve as anatomical markers for morphologically and potential functionally distinct neuronal subpopulations. We observed wide expression of CBPs in the adult zebrafish, in several spinal and reticulospinal neuronal populations with a diverse neurotransmitter phenotype. We also found that several spinal motoneurons express CR and PV. However, only the motoneuron pools that are responsible for generation of fast locomotion were CR-positive. CR can thus be used as a marker for fast motoneurons and might potentially label the fast locomotor module. Moreover, CB was mainly observed in the neuronal progenitor cells that are distributed around the central canal. Thus, our results suggest that during development the spinal neurons utilize CB and as the neurons mature and establish a neurotransmitter phenotype they use CR or/and PV. The detailed characterization of CBPs expression, in the spinal cord and brainstem neurons, is a crucial step toward a better understanding of the development and functionality of neuronal locomotor networks.
Collapse
Affiliation(s)
- Eva M Berg
- Department of Neuroscience, Karolinska Institutet, 171 77, Stockholm, Sweden
| | - Maria Bertuzzi
- Department of Neuroscience, Karolinska Institutet, 171 77, Stockholm, Sweden
| | | |
Collapse
|
29
|
Ribeiro A, Monteiro JF, Certal AC, Cristovão AM, Saúde L. Foxj1a is expressed in ependymal precursors, controls central canal position and is activated in new ependymal cells during regeneration in zebrafish. Open Biol 2018; 7:rsob.170139. [PMID: 29162726 PMCID: PMC5717339 DOI: 10.1098/rsob.170139] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Accepted: 10/27/2017] [Indexed: 01/06/2023] Open
Abstract
Zebrafish are able to regenerate the spinal cord and recover motor and sensory functions upon severe injury, through the activation of cells located at the ependymal canal. Here, we show that cells surrounding the ependymal canal in the adult zebrafish spinal cord express Foxj1a. We demonstrate that ependymal cells express Foxj1a from their birth in the embryonic neural tube and that Foxj1a activity is required for the final positioning of the ependymal canal. We also show that in response to spinal cord injury, Foxj1a ependymal cells actively proliferate and contribute to the restoration of the spinal cord structure. Finally, this study reveals that Foxj1a expression in the injured spinal cord is regulated by regulatory elements activated during regeneration. These data establish Foxj1a as a pan-ependymal marker in development, homeostasis and regeneration and may help identify the signals that enable this progenitor population to replace lost cells after spinal cord injury.
Collapse
Affiliation(s)
- Ana Ribeiro
- Instituto de Medicina Molecular, Faculdade de Medicina da Universidade de Lisboa, 1649-028 Lisboa, Portugal
| | - Joana F Monteiro
- Champalimaud Research, Champalimaud Centre for the Unknown, Lisboa, Portugal
| | - Ana C Certal
- Champalimaud Research, Champalimaud Centre for the Unknown, Lisboa, Portugal
| | - Ana M Cristovão
- Instituto de Medicina Molecular, Faculdade de Medicina da Universidade de Lisboa, 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
|
30
|
Ghosh S, Hui SP. Axonal regeneration in zebrafish spinal cord. REGENERATION (OXFORD, ENGLAND) 2018; 5:43-60. [PMID: 29721326 PMCID: PMC5911453 DOI: 10.1002/reg2.99] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Revised: 03/09/2018] [Accepted: 03/13/2018] [Indexed: 12/12/2022]
Abstract
In the present review we discuss two interrelated events-axonal damage and repair-known to occur after spinal cord injury (SCI) in the zebrafish. Adult zebrafish are capable of regenerating axonal tracts and can restore full functionality after SCI. Unlike fish, axon regeneration in the adult mammalian central nervous system is extremely limited. As a consequence of an injury there is very little repair of disengaged axons and therefore functional deficit persists after SCI in adult mammals. In contrast, peripheral nervous system axons readily regenerate following injury and hence allow functional recovery both in mammals and fish. A better mechanistic understanding of these three scenarios could provide a more comprehensive insight into the success or failure of axonal regeneration after SCI. This review summarizes the present understanding of the cellular and molecular basis of axonal regeneration, in both the peripheral nervous system and the central nervous system, and large scale gene expression analysis is used to focus on different events during regeneration. The discovery and identification of genes involved in zebrafish spinal cord regeneration and subsequent functional experimentation will provide more insight into the endogenous mechanism of myelination and remyelination. Furthermore, precise knowledge of the mechanism underlying the extraordinary axonal regeneration process in zebrafish will also allow us to unravel the potential therapeutic strategies to be implemented for enhancing regrowth and remyelination of axons in mammals.
Collapse
Affiliation(s)
- Sukla Ghosh
- Department of BiophysicsMolecular Biology and BioinformaticsUniversity of Calcutta92 A. P. C. RoadKolkata 700009India
| | - Subhra Prakash Hui
- Department of BiophysicsMolecular Biology and BioinformaticsUniversity of Calcutta92 A. P. C. RoadKolkata 700009India
- Victor Chang Cardiac Research InstituteLowy Packer Building, 405 Liverpool StDarlinghurstNSW 2010Australia.
| |
Collapse
|
31
|
Shimizu Y, Ueda Y, Ohshima T. Wnt signaling regulates proliferation and differentiation of radial glia in regenerative processes after stab injury in the optic tectum of adult zebrafish. Glia 2018; 66:1382-1394. [PMID: 29411422 DOI: 10.1002/glia.23311] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Revised: 01/18/2018] [Accepted: 01/29/2018] [Indexed: 01/03/2023]
Abstract
Zebrafish have superior abilities to generate new neurons in the adult brain and to regenerate brain tissue after brain injury compared with mammals. There exist two types of neural stem cells (NSCs): neuroepithelial-like stem cells (NE) and radial glia (RG) in the optic tectum. We established an optic tectum stab injury model to analyze the function of NSCs in the regenerative condition and confirmed that the injury induced the proliferation of RG, but not NE and that the proliferated RG differentiated into new neurons after the injury. We then analyzed the involvement of Wnt signaling after the injury, using a Wnt reporter line in which canonical Wnt signaling activation induced GFP expression and confirmed that GFP expression was induced specifically in RG after the injury. We also analyzed the expression level of genes related to Wnt signaling, and confirmed that endogenous Wnt antagonist dkk1b expression was significantly decreased after the injury. We observed that Wnt signal inhibitor IWR1 treatment suppressed the proliferation and differentiation of RG after the injury, suggesting that up-regulation of Wnt signaling in RG after the stab injury was required for optic tectum regeneration. We also confirmed that Wnt activation by treatment with GSK3β inhibitor BIO in uninjured zebrafish induced proliferation of RG in the optic tectum. This optic tectum stab injury model is useful for the study of the molecular mechanisms of brain regeneration and analysis of the RG functions in physiological and regenerative conditions.
Collapse
Affiliation(s)
- Yuki Shimizu
- Department of Life Science and Medical Bio-Science, Waseda University, 2-2 Wakamatsu-cho, Shinjuku-ku, Tokyo, 162-8480, Japan
| | - Yuto Ueda
- Department of Life Science and Medical Bio-Science, Waseda University, 2-2 Wakamatsu-cho, Shinjuku-ku, Tokyo, 162-8480, Japan
| | - Toshio Ohshima
- Department of Life Science and Medical Bio-Science, Waseda University, 2-2 Wakamatsu-cho, Shinjuku-ku, Tokyo, 162-8480, Japan
| |
Collapse
|
32
|
Méndez-Olivos EE, Muñoz R, Larraín J. Spinal Cord Cells from Pre-metamorphic Stages Differentiate into Neurons and Promote Axon Growth and Regeneration after Transplantation into the Injured Spinal Cord of Non-regenerative Xenopus laevis Froglets. Front Cell Neurosci 2017; 11:398. [PMID: 29326551 PMCID: PMC5733487 DOI: 10.3389/fncel.2017.00398] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Accepted: 11/28/2017] [Indexed: 11/13/2022] Open
Abstract
Mammals are unable to regenerate its spinal cord after a lesion, meanwhile, anuran amphibians are capable of spinal cord regeneration only as larvae, and during metamorphosis, this capability is lost. Sox2/3+ cells present in the spinal cord of regenerative larvae are required for spinal cord regeneration. Here we evaluate the effect of the transplantation of spinal cord cells from regenerative larvae into the resected spinal cord of non-regenerative stages (NR-stage). Donor cells were able to survive up to 60 days after transplantation in the injury zone. During the first 3-weeks, transplanted cells organize in neural tube-like structures formed by Sox2/3+ cells. This was not observed when donor cells come from non-regenerative froglets. Mature neurons expressing NeuN and Neurofilament-H were detected in the grafted tissue 4 weeks after transplantation concomitantly with the appearance of axons derived from the donor cells growing into the host spinal cord, suggesting that Sox2/3+ cells behave as neural stem progenitor cells. We also found that cells from regenerative animals provide a permissive environment that promotes growth and regeneration of axons coming from the host. These results suggest that Sox2/3 cells present in the spinal cord of regenerative stage (R-stage) larvae are most probably neural stem progenitor cells that are able to survive, proliferate, self-organize and differentiate into neurons in the environment of the non-regenerative host. In addition, we have established an experimental paradigm to study the biology of neural stem progenitor cells in spinal cord regeneration.
Collapse
Affiliation(s)
- Emilio E Méndez-Olivos
- Center for Aging and Regeneration, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Rosana Muñoz
- Center for Aging and Regeneration, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Juan Larraín
- Center for Aging and Regeneration, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile
| |
Collapse
|
33
|
Hui SP, Sheng DZ, Sugimoto K, Gonzalez-Rajal A, Nakagawa S, Hesselson D, Kikuchi K. Zebrafish Regulatory T Cells Mediate Organ-Specific Regenerative Programs. Dev Cell 2017; 43:659-672.e5. [DOI: 10.1016/j.devcel.2017.11.010] [Citation(s) in RCA: 125] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2017] [Revised: 10/20/2017] [Accepted: 11/13/2017] [Indexed: 12/12/2022]
|
34
|
Go and stop signals for glial regeneration. Curr Opin Neurobiol 2017; 47:182-187. [PMID: 29126016 PMCID: PMC6419527 DOI: 10.1016/j.conb.2017.10.011] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Revised: 08/16/2017] [Accepted: 10/10/2017] [Indexed: 12/22/2022]
Abstract
The regenerative response of ensheating glia to central nervous system (CNS) injury involves proliferation and differentiation, axonal re-enwrapment and some recovery of behaviour. Understanding this limited response could enable the enhancement of it. In Drosophila, the glial progenitor state is maintained by Notch, an activator of cell division and Prospero (Pros), a repressor. Injury provokes the activation of NFκB and up-regulation of Kon-tiki (Kon), driving cell proliferation. Homeostatic switch-off comes about as two negative feedback loops involving Pros terminate the response. Importantly, the functions of the kon and pros homologues NG2 and prox1, respectively, are conserved in mammalian NG2 glia. Controlling these genes is key for therapeutic manipulation of progenitors and stem cells to promote regeneration of the damaged CNS.
Collapse
|
35
|
Sarasamma S, Varikkodan MM, Liang ST, Lin YC, Wang WP, Hsiao CD. Zebrafish: A Premier Vertebrate Model for Biomedical Research in Indian Scenario. Zebrafish 2017; 14:589-605. [PMID: 29023224 DOI: 10.1089/zeb.2017.1447] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The zebrafish (Danio rerio) is a versatile model organism that has been used in biomedical research for several decades to study a wide range of biological phenomena. There are many technical advantages of using zebrafish over other vertebrate models. They are readily available, hardy, easy, and inexpensive to maintain in the laboratory, have a short life cycle, and have excellent fecundity. Due to its optical clarity and reproducible capabilities, it has become one of the predominant models of human genetic diseases. Zebrafish research has made rapid strides in the United States and Europe, but in India the field is at an early stage and many researchers still remain unaware of the full research potential of this tiny fish. The zebrafish model system was introduced into India in the early 2000s. Up to now, more than 200 scientific referred articles have been published by Indian researchers. This review gives an overview of the current state of knowledge for zebrafish research in India, with the aim of promoting wider utilization of zebrafish for high level biological studies.
Collapse
Affiliation(s)
- Sreeja Sarasamma
- 1 Department of Chemistry, Chung Yuan Christian University , Chung-Li, Taiwan .,2 Department of Bioscience Technology, Chung Yuan Christian University , Chung-Li, Taiwan .,3 Department of Chemical Biology, Rajiv Gandhi Centre for Biotechnology , Thiruvananthapuram, Kerala, India
| | - Muhammed Muhsin Varikkodan
- 1 Department of Chemistry, Chung Yuan Christian University , Chung-Li, Taiwan .,2 Department of Bioscience Technology, Chung Yuan Christian University , Chung-Li, Taiwan .,4 Department of Biotechnology and Genetic Engineering, Bharathidasan University , Tiruchirapalli, India
| | - Sung-Tzu Liang
- 1 Department of Chemistry, Chung Yuan Christian University , Chung-Li, Taiwan
| | - Yen-Chang Lin
- 5 Graduate Institute of Biotechnology, Chinese Culture University , Taipei, Taiwan
| | - Wen-Pin Wang
- 6 Institute of Medical Sciences, Tzu-Chi University , Hualien, Taiwan .,7 Department of Molecular Biology and Human Genetics, Tzu-Chi University , Hualien, Taiwan
| | - Chung-Der Hsiao
- 1 Department of Chemistry, Chung Yuan Christian University , Chung-Li, Taiwan .,8 Center for Biomedical Technology, Chung Yuan Christian University , Chung-Li, Taiwan .,9 Center for Nanotechnology, Chung Yuan Christian University , Chung-Li, Taiwan
| |
Collapse
|
36
|
Fu Q, Liu Y, Liu X, Zhang Q, Chen L, Peng J, Ao J, Li Y, Wang S, Song G, Yu L, Liu J, Zhang T. Engrafted peripheral blood-derived mesenchymal stem cells promote locomotive recovery in adult rats after spinal cord injury. Am J Transl Res 2017; 9:3950-3966. [PMID: 28979672 PMCID: PMC5622241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2017] [Accepted: 08/03/2017] [Indexed: 06/07/2023]
Abstract
Spinal cord injury (SCI) is a severe trauma of central nervous system (CNS). Numerous stem cells have been applied for SCI therapy. Peripheral blood-derived mesenchymal stem cells (PBMSCs) have captured researchers' attention by virtue of pluripotency and effectiveness. However, little work has been performed on whether PBMSCs play roles and what role, if any, in the lesion microenvironment. Through the investigation of the differentiation, neuroprotection and immunoloregulation of engrafted PBMSCs, we found that the expression of glial fibrillary acidic protein (GFAP) was inhibited. Meanwhile, myelin basic protein (MBP), neurofilament protein-200 (NF-200) and microtubule associated protein-2 (MAP-2) were promoted after PBMSC transplantation (PBMSCT) by immunohistochemistry. Though engrafted PKH26+PBMSCs could survive in vivo for at least 8 w, they could not respectively express GFAP, MBP and neuronal specific neucleoprotein (NeuN) by immunofluorescence. Additionally, Flow cytometry demonstrated that the number of CD4+IL17+Th17 cells decreased while CD4+CD25+Foxp3+Treg ones increased after PBMSCT (P < 0.01). Immunohistochemistry and Elisa both showed a lower expression of IL-6 and IL-17a while a higher expression of TGF-β after PBMSCT (P < 0.05). RT-PCR indicated that Th17-relevant genes including RORγT, IL-6 and IL-21 were inhibited and resulted in the decrease of IL-23a and IL-22 secretion (P < 0.05); Treg-relevant genes including FoxP3 and TGF-β and the secretion of IL-10 were improved (P < 0.05). Accordingly, we concluded that the PBMSCT-relevant therapy took effect not through the differentiation of PBMSCs into CNS cells, but through regulating Th17/Treg-relevant gene expression, inhibiting Th17-relevant gene expression and meanwhile promoting Treg-relevant gene expression, and eventually resulted in promotion of the functional recovery of SCI rats.
Collapse
Affiliation(s)
- Qiang Fu
- Key Laboratory of Cell Engineering of Guizhou Province and Regenerative Medicine Centre, Affiliated Hospital of Zunyi Medical CollegeZunyi, Guizhou, China
- Department of Orthopedics, Affiliated Hospital of Zunyi Medical CollegeZunyi, Guizhou, China
- Sichuan Academy of Medical Sciences and Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of ChinaChengdu, Sichuan, China
| | - Yi Liu
- Department of Orthopedics, Affiliated Hospital of Zunyi Medical CollegeZunyi, Guizhou, China
| | - Xiu Liu
- Key Laboratory of Cell Engineering of Guizhou Province and Regenerative Medicine Centre, Affiliated Hospital of Zunyi Medical CollegeZunyi, Guizhou, China
| | - Qian Zhang
- Department of Human Anatomy, Zunyi Medical CollegeZunyi, Guizhou, China
| | - Long Chen
- Key Laboratory of Cell Engineering of Guizhou Province and Regenerative Medicine Centre, Affiliated Hospital of Zunyi Medical CollegeZunyi, Guizhou, China
- Experimental Centre, Affiliated Dongfeng General Hospital of Hubei University of MedicineShiyan, Hubei, China
| | - Jiachen Peng
- Department of Orthopedics, Affiliated Hospital of Zunyi Medical CollegeZunyi, Guizhou, China
| | - Jun Ao
- Department of Orthopedics, Affiliated Hospital of Zunyi Medical CollegeZunyi, Guizhou, China
| | - Yuwan Li
- Department of Orthopedics, Affiliated Hospital of Zunyi Medical CollegeZunyi, Guizhou, China
| | - Shengmin Wang
- Department of Orthopedics, Affiliated Hospital of Zunyi Medical CollegeZunyi, Guizhou, China
| | - Gongyu Song
- Department of Human Anatomy, Zunyi Medical CollegeZunyi, Guizhou, China
| | - Limei Yu
- Key Laboratory of Cell Engineering of Guizhou Province and Regenerative Medicine Centre, Affiliated Hospital of Zunyi Medical CollegeZunyi, Guizhou, China
| | - Jinwei Liu
- Key Laboratory of Cell Engineering of Guizhou Province and Regenerative Medicine Centre, Affiliated Hospital of Zunyi Medical CollegeZunyi, Guizhou, China
| | - Tao Zhang
- Key Laboratory of Cell Engineering of Guizhou Province and Regenerative Medicine Centre, Affiliated Hospital of Zunyi Medical CollegeZunyi, Guizhou, China
| |
Collapse
|
37
|
Cocktail of chemical compounds robustly promoting cell reprogramming protects liver against acute injury. Protein Cell 2017; 8:273-283. [PMID: 28190217 PMCID: PMC5359186 DOI: 10.1007/s13238-017-0373-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Accepted: 01/08/2017] [Indexed: 01/06/2023] Open
Abstract
Tissue damage induces cells into reprogramming-like cellular state, which contributes to tissue regeneration. However, whether factors promoting the cell reprogramming favor tissue regeneration remains elusive. Here we identified combination of small chemical compounds including drug cocktails robustly promoting in vitro cell reprogramming. We then administrated the drug cocktails to mice with acute liver injuries induced by partial hepatectomy or toxic treatment. Our results demonstrated that the drug cocktails which promoted cell reprogramming in vitro improved liver regeneration and hepatic function in vivo after acute injuries. The underlying mechanism could be that expression of pluripotent genes activated after injury is further upregulated by drug cocktails. Thus our study offers proof-of-concept evidence that cocktail of clinical compounds improving cell reprogramming favors tissue recovery after acute damages, which is an attractive strategy for regenerative purpose.
Collapse
|
38
|
Zhang L, Tao W, Feng H, Chen Y. Transcriptional and Genomic Targets of Neural Stem Cells for Functional Recovery after Hemorrhagic Stroke. Stem Cells Int 2017; 2017:2412890. [PMID: 28133486 PMCID: PMC5241497 DOI: 10.1155/2017/2412890] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Accepted: 12/21/2016] [Indexed: 01/27/2023] Open
Abstract
Hemorrhagic stroke is a life-threatening disease characterized by a sudden rupture of cerebral blood vessels, and it is widely believed that neural cell death occurs after exposure to blood metabolites or subsequently damaged cells. Neural stem cells (NSCs), which maintain neurogenesis and are found in subgranular zone and subventricular zone, are thought to be an endogenous neuroprotective mechanism for these brain injuries. However, due to the complexity of NSCs and their microenvironment, current strategies cannot satisfactorily enhance functional recovery after hemorrhagic stroke. It is well known that transcriptional and genomic pathways play important roles in ensuring the normal functions of NSCs, including proliferation, migration, differentiation, and neural reconnection. Recently, emerging evidence from the use of new technologies such as next-generation sequencing and transcriptome profiling has provided insight into our understanding of genomic function and regulation of NSCs. In the present article, we summarize and present the current data on the control of NSCs at both the transcriptional and genomic levels. Using bioinformatics methods, we sought to predict novel therapeutic targets of endogenous neurogenesis and exogenous NSC transplantation for functional recovery after hemorrhagic stroke, which could also advance our understanding of its pathophysiology.
Collapse
Affiliation(s)
- Le Zhang
- College of Computer and Information Science, Southwest University, Chongqing, China
| | - Wenjing Tao
- College of Computer and Information Science, Southwest University, Chongqing, China
| | - Hua Feng
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Yujie Chen
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University, Chongqing, China
| |
Collapse
|
39
|
Rost F, Rodrigo Albors A, Mazurov V, Brusch L, Deutsch A, Tanaka EM, Chara O. Accelerated cell divisions drive the outgrowth of the regenerating spinal cord in axolotls. eLife 2016; 5:20357. [PMID: 27885987 PMCID: PMC5182066 DOI: 10.7554/elife.20357] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2016] [Accepted: 11/14/2016] [Indexed: 01/25/2023] Open
Abstract
Axolotls are unique in their ability to regenerate the spinal cord. However, the mechanisms that underlie this phenomenon remain poorly understood. Previously, we showed that regenerating stem cells in the axolotl spinal cord revert to a molecular state resembling embryonic neuroepithelial cells and functionally acquire rapid proliferative divisions (Rodrigo Albors et al., 2015). Here, we refine the analysis of cell proliferation in space and time and identify a high-proliferation zone in the regenerating spinal cord that shifts posteriorly over time. By tracking sparsely-labeled cells, we also quantify cell influx into the regenerate. Taking a mathematical modeling approach, we integrate these quantitative datasets of cell proliferation, neural stem cell activation and cell influx, to predict regenerative tissue outgrowth. Our model shows that while cell influx and neural stem cell activation play a minor role, the acceleration of the cell cycle is the major driver of regenerative spinal cord outgrowth in axolotls. DOI:http://dx.doi.org/10.7554/eLife.20357.001
Collapse
Affiliation(s)
- Fabian Rost
- Center for Information Services and High Performance Computing, Technische Universität Dresden, Dresden, Germany
| | - Aida Rodrigo Albors
- Deutsche Forschungsgemeinschaft - Center for Regenerative Therapies Dresden, Dresden, Germany.,Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Vladimir Mazurov
- Deutsche Forschungsgemeinschaft - Center for Regenerative Therapies Dresden, Dresden, Germany.,Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Lutz Brusch
- Center for Information Services and High Performance Computing, Technische Universität Dresden, Dresden, Germany.,Center for Advancing Electronics Dresden, Dresden, Germany
| | - Andreas Deutsch
- Center for Information Services and High Performance Computing, Technische Universität Dresden, Dresden, Germany.,Center for Advancing Electronics Dresden, Dresden, Germany
| | - Elly M Tanaka
- Deutsche Forschungsgemeinschaft - Center for Regenerative Therapies Dresden, Dresden, Germany.,Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Osvaldo Chara
- Center for Information Services and High Performance Computing, Technische Universität Dresden, Dresden, Germany.,Systems Biology Group, Institute of Physics of Liquids and Biological Systems, National Scientific and Technical Research Council, University of La Plata, La Plata, Argentina
| |
Collapse
|
40
|
Zeng CW, Kamei Y, Wang CT, Tsai HJ. Subtypes of hypoxia-responsive cells differentiate into neurons in spinal cord of zebrafish embryos after hypoxic stress. Biol Cell 2016; 108:357-377. [PMID: 27539672 DOI: 10.1111/boc.201600015] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2016] [Revised: 08/11/2016] [Accepted: 08/15/2016] [Indexed: 11/27/2022]
Abstract
BACKGROUND INFORMATION Neuron stem/progenitor cells (NSPCs) of zebrafish central nervous system (CNS) are known to thrive during oxygen recovery after hypoxia, but not all cell types have been fully characterised due to their heterogeneities. In addition, an in vivo model system is not available that can help us to identify what type-specific cell populations that are involved in neural regeneration and to track their cell fate after regeneration. To solve these issues, we employed a zebrafish transgenic line, huORFZ, which harbours an inhibitory upstream open reading frame of human chop mRNA fused downstream with GFP reporter and driven by cytomegalovirus promoter. When huORFZ embryos were exposure to hypoxic stress, followed by oxygen recovery, GFP was exclusively expressed in some particular cells of CNS. Unlike GFP-negative cells, all GFP-expressing cells were not apoptotic, indicating that cell populations that are able to survive after hypoxia can be identified through this approach. RESULTS When GFP-expressing cells of spinal cord were studied, we found mostly NSPCs and radial glia cells (RGs), along with a few oligodendrocyte progenitor cells and oligodendrocytes, all termed as hypoxia-responsive recovering cells (HrRCs). After hypoxic stress, these GFP-positive HrRCs did not undergo apoptosis, but GFP-negative neurons did. Prolonged recovery time after hypoxia was correlated with higher proportions of GFP(+)-NSPCs and GFP(+)-RGs, in contrast to lower proportions of proliferating/differentiating GFP(-)-NSPCs and GFP(-)-RGs. Among HrRCs subtypes, only GFP(+)-NSPCs and GFP(+)-RGs proliferated, migrated and differentiated into functional neurons during oxygen recovery. When some HrRCs were ablated in the spinal cord of hypoxia-exposed huORFZ embryos, swimming performance was impaired, suggesting that HrRCs are involved in neuronal regeneration. CONCLUSION We demonstrated type-specific cell populations able to respond sensitively to hypoxic stress in the spinal cord of zebrafish embryos and that these type-specific populations play a role in neural regeneration. SIGNIFICANCE Among heterogeneous cell types that exist in the spinal cord of zebrafish embryos after hypoxia, the particular cells that are resistant to hypoxia and also involved in neuronal regeneration can be clearly identified and dynamically traced using a transgenic model fish.
Collapse
Affiliation(s)
- Chih-Wei Zeng
- Institute of Molecular and Cellular Biology, National Taiwan University, Taipei, 10617, Taiwan
| | - Yasuhiro Kamei
- National Institute for Basic Biology, Okazaki, 444-8585, Aichi, Japan
| | - Chih-Tien Wang
- Institute of Molecular and Cellular Biology, National Taiwan University, Taipei, 10617, Taiwan
| | - Huai-Jen Tsai
- Institute of Molecular and Cellular Biology, National Taiwan University, Taipei, 10617, Taiwan.,Institute of Biomedical Science, Mackay Medical College, New Taipei City, 252, Taiwan
| |
Collapse
|
41
|
Ghosh S, Hui SP. Regeneration of Zebrafish CNS: Adult Neurogenesis. Neural Plast 2016; 2016:5815439. [PMID: 27382491 PMCID: PMC4921647 DOI: 10.1155/2016/5815439] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2016] [Accepted: 04/18/2016] [Indexed: 12/13/2022] Open
Abstract
Regeneration in the animal kingdom is one of the most fascinating problems that have allowed scientists to address many issues of fundamental importance in basic biology. However, we came to know that the regenerative capability may vary across different species. Among vertebrates, fish and amphibians are capable of regenerating a variety of complex organs through epimorphosis. Zebrafish is an excellent animal model, which can repair several organs like damaged retina, severed spinal cord, injured brain and heart, and amputated fins. The focus of the present paper is on spinal cord regeneration in adult zebrafish. We intend to discuss our current understanding of the cellular and molecular mechanism(s) that allows formation of proliferating progenitors and controls neurogenesis, which involve changes in epigenetic and transcription programs. Unlike mammals, zebrafish retains radial glia, a nonneuronal cell type in their adult central nervous system. Injury induced proliferation involves radial glia which proliferate, transcribe embryonic genes, and can give rise to new neurons. Recent technological development of exquisite molecular tools in zebrafish, such as cell ablation, lineage analysis, and novel and substantial microarray, together with advancement in stem cell biology, allowed us to investigate how progenitor cells contribute to the generation of appropriate structures and various underlying mechanisms like reprogramming.
Collapse
Affiliation(s)
- Sukla Ghosh
- Department of Biophysics, Molecular Biology and Bioinformatics, University of Calcutta, 92 A. P. C. Road, Kolkata 700009, India
| | - Subhra Prakash Hui
- Department of Biophysics, Molecular Biology and Bioinformatics, University of Calcutta, 92 A. P. C. Road, Kolkata 700009, India
| |
Collapse
|
42
|
Rooj AK, Bronisz A, Godlewski J. The role of octamer binding transcription factors in glioblastoma multiforme. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2016; 1859:805-11. [PMID: 26968235 DOI: 10.1016/j.bbagrm.2016.03.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/14/2016] [Revised: 03/04/2016] [Accepted: 03/05/2016] [Indexed: 12/24/2022]
Abstract
A group of transcription factors (TF) that are master developmental regulators of the establishment and maintenance of pluripotency during embryogenesis play additional roles to control tissue homeostasis and regeneration in adults. Among these TFs, members of the octamer-binding transcription factor (OCT) gene family are well documented as major regulators controlling the self-renewal and pluripotency of stem cells isolated from different adult organs including the brain. In the last few years a large number of studies show the aberrant expression and dysfunction of OCT in different types of cancers including glioblastoma multiforme (GBM). GBM is the most common malignant primary brain tumor, and contains a subpopulation of undifferentiated stem cells (GSCs), with self-renewal and tumorigenic potential that contribute to tumor initiation, invasion, recurrence, and therapeutic resistance. In this review, we have summarized the current knowledge about OCT family in GBM and their crucial role in the initiation, maintenance and drug resistance properties of GSCs. This article is part of a Special Issue entitled: The Oct Transcription Factor Family, edited by Dr. Dean Tantin.
Collapse
Affiliation(s)
- A K Rooj
- Harvey Cushing Neuro-Oncology Laboratories, Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States
| | - A Bronisz
- Harvey Cushing Neuro-Oncology Laboratories, Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States
| | - J Godlewski
- Harvey Cushing Neuro-Oncology Laboratories, Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
43
|
Ghosh S. Human regeneration: An achievable goal or a dream? J Biosci 2016; 41:157-65. [PMID: 26949097 DOI: 10.1007/s12038-016-9589-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
The main objective of regenerative medicine is to replenish cells or tissues or even to restore different body parts that are lost or damaged due to disease, injury and aging. Several avenues have been explored over many decades to address the fascinating problem of regeneration at the cell, tissue and organ levels. Here we discuss some of the primary approaches adopted by researchers in the context of enhancing the regenerating ability of mammals. Natural regeneration can occur in different animal species, and the underlying mechanism is highly relevant to regenerative medicine-based intervention. Significant progress has been achieved in understanding the endogenous regeneration in urodeles and fishes with the hope that they could help to reach our goal of designing future strategies for human regeneration.
Collapse
Affiliation(s)
- Sukla Ghosh
- Department of Biophysics, Molecular Biology and Bioinformatics, University of Calcutta, 92, A. P.C. Road, Kolkata 700 009, India,
| |
Collapse
|