1
|
Marconi A, Vernaz G, Karunaratna A, Ngochera MJ, Durbin R, Santos ME. Genetic and Developmental Divergence in the Neural Crest Program between Cichlid Fish Species. Mol Biol Evol 2024; 41:msae217. [PMID: 39412298 PMCID: PMC11558072 DOI: 10.1093/molbev/msae217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Revised: 10/07/2024] [Accepted: 10/09/2024] [Indexed: 10/30/2024] Open
Abstract
Neural crest (NC) is a vertebrate-specific embryonic progenitor cell population at the basis of important vertebrate features such as the craniofacial skeleton and pigmentation patterns. Despite the wide-ranging variation of NC-derived traits across vertebrates, the contribution of NC to species diversification remains underexplored. Here, leveraging the adaptive diversity of African Great Lakes' cichlid species, we combined comparative transcriptomics and population genomics to investigate the evolution of the NC genetic program in the context of their morphological divergence. Our analysis revealed substantial differences in transcriptional landscapes across somitogenesis, an embryonic period coinciding with NC development and migration. This included dozens of genes with described functions in the vertebrate NC gene regulatory network, several of which showed signatures of positive selection. Among candidates showing between-species expression divergence, we focused on teleost-specific paralogs of the NC-specifier sox10 (sox10a and sox10b) as prime candidates to influence NC development. These genes, expressed in NC cells, displayed remarkable spatio-temporal variation in cichlids, suggesting their contribution to interspecific morphological differences, such as craniofacial structures and pigmentation. Finally, through CRISPR/Cas9 mutagenesis, we demonstrated the functional divergence between cichlid sox10 paralogs, with the acquisition of a novel skeletogenic function by sox10a. When compared with teleost models zebrafish and medaka, our findings reveal that sox10 duplication, although retained in most teleost lineages, had variable functional fates across their phylogeny. Altogether, our study suggests that NC-related processes-particularly those controlled by sox10s-are involved in generating morphological diversification between species and lays the groundwork for further investigations into the mechanisms underpinning vertebrate NC diversification.
Collapse
Affiliation(s)
| | - Grégoire Vernaz
- Zoological Institute, University of Basel, Basel, Switzerland
| | | | - Maxon J Ngochera
- Malawi Fisheries Department, Senga Bay Fisheries Research Center, P.O. Box 316, Salima, Malawi
| | - Richard Durbin
- Department of Genetics, University of Cambridge, Cambridge, UK
| | - M Emília Santos
- Department of Zoology, University of Cambridge, Cambridge, UK
| |
Collapse
|
2
|
Anneser L, Satou C, Hotz HR, Friedrich RW. Molecular organization of neuronal cell types and neuromodulatory systems in the zebrafish telencephalon. Curr Biol 2024; 34:298-312.e4. [PMID: 38157860 PMCID: PMC10808507 DOI: 10.1016/j.cub.2023.12.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 11/30/2023] [Accepted: 12/04/2023] [Indexed: 01/03/2024]
Abstract
The function of neuronal networks is determined not only by synaptic connectivity but also by neuromodulatory systems that broadcast information via distributed connections and volume transmission. To understand the molecular constraints that organize neuromodulatory signaling in the telencephalon of adult zebrafish, we used transcriptomics and additional approaches to delineate cell types, to determine their phylogenetic conservation, and to map the expression of marker genes at high granularity. The combinatorial expression of GPCRs and cell-type markers indicates that all neuronal cell types are subject to modulation by multiple monoaminergic systems and distinct combinations of neuropeptides. Individual cell types were associated with multiple (typically >30) neuromodulatory signaling networks but expressed only a few diagnostic GPCRs at high levels, suggesting that different neuromodulatory systems act in combination, albeit with unequal weights. These results provide a detailed map of cell types and brain areas in the zebrafish telencephalon, identify core components of neuromodulatory networks, highlight the cell-type specificity of neuropeptides and GPCRs, and begin to decipher the logic of combinatorial neuromodulation.
Collapse
Affiliation(s)
- Lukas Anneser
- Friedrich Miescher Institute for Biomedical Research, Maulbeerstrasse 66, 4058 Basel, Switzerland
| | - Chie Satou
- Friedrich Miescher Institute for Biomedical Research, Maulbeerstrasse 66, 4058 Basel, Switzerland
| | - Hans-Rudolf Hotz
- Friedrich Miescher Institute for Biomedical Research, Maulbeerstrasse 66, 4058 Basel, Switzerland
| | - Rainer W Friedrich
- Friedrich Miescher Institute for Biomedical Research, Maulbeerstrasse 66, 4058 Basel, Switzerland; University of Basel, 4003 Basel, Switzerland.
| |
Collapse
|
3
|
Altbürger C, Rath M, Armbruster D, Driever W. Neurog1 and Olig2 integrate patterning and neurogenesis signals in development of zebrafish dopaminergic and glutamatergic dual transmitter neurons. Dev Biol 2024; 505:85-98. [PMID: 37944224 DOI: 10.1016/j.ydbio.2023.10.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 10/02/2023] [Accepted: 10/20/2023] [Indexed: 11/12/2023]
Abstract
Dopaminergic neurons develop in distinct neural domains by integrating local patterning and neurogenesis signals. While the proneural proteins Neurog1 and Olig2 have been previously linked to development of dopaminergic neurons, their dependence on local prepatterning and specific contributions to dopaminergic neurogenesis are not well understood. Here, we show that both transcription factors are differentially required for the development of defined dopaminergic glutamatergic subpopulations in the zebrafish posterior tuberculum, which are homologous to A11 dopaminergic neurons in mammals. Both Olig2 and Neurog1 are expressed in otpa expressing progenitor cells and appear to act upstream of Otpa during dopaminergic neurogenesis. Our epistasis analysis confirmed that Neurog1 acts downstream of Notch signaling, while Olig2 acts downstream of Shh, but upstream and/or in parallel to Notch signaling during neurogenesis of A11-type dopaminergic clusters. Furthermore, we identified Olig2 to be an upstream regulator of neurog1 in dopaminergic neurogenesis. This regulation occurs through Olig2-dependent repression of the proneural repressor and Notch target gene her2. Our study reveals how Neurog1 and Olig2 integrate local patterning signals, including Shh, with Notch neurogenic selection signaling, to specify the progenitor population and initiate neurogenesis and differentiation of A11-type dopaminergic neurons.
Collapse
Affiliation(s)
- Christian Altbürger
- Department of Developmental Biology, Faculty of Biology, Albert-Ludwigs-University of Freiburg, 79104 Freiburg, Germany; Signalling Research Centres BIOSS and CIBSS, University of Freiburg, 79104 Freiburg, Germany
| | - Meta Rath
- Department of Developmental Biology, Faculty of Biology, Albert-Ludwigs-University of Freiburg, 79104 Freiburg, Germany
| | - Daniel Armbruster
- Department of Developmental Biology, Faculty of Biology, Albert-Ludwigs-University of Freiburg, 79104 Freiburg, Germany; MeInBio Research Training Group, University of Freiburg, 79104 Freiburg, Germany
| | - Wolfgang Driever
- Department of Developmental Biology, Faculty of Biology, Albert-Ludwigs-University of Freiburg, 79104 Freiburg, Germany; Signalling Research Centres BIOSS and CIBSS, University of Freiburg, 79104 Freiburg, Germany.
| |
Collapse
|
4
|
Masson MA, Nait-Oumesmar B. Emerging concepts in oligodendrocyte and myelin formation, inputs from the zebrafish model. Glia 2023; 71:1147-1163. [PMID: 36645033 DOI: 10.1002/glia.24336] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 12/20/2022] [Accepted: 12/29/2022] [Indexed: 01/17/2023]
Abstract
Oligodendrocytes (OLs) are the myelinating cells of the central nervous system (CNS), which are derived from OL precursor cells. Myelin insulates axons allowing the saltatory conduction of action potentials and also provides trophic and metabolic supports to axons. Interestingly, oligodendroglial cells have the capacity to sense neuronal activity, which regulates myelin sheath formation via the vesicular release of neurotransmitters. Neuronal activity-dependent regulation of myelination is mediated by specialized interaction between axons and oligodendroglia, involving both synaptic and extra-synaptic modes of communications. The zebrafish has provided key advantages for the study of the myelination process in the CNS. External development and transparent larval stages of this vertebrate specie combined with the existence of several transgenic reporter lines provided key advances in oligodendroglial cell biology, axo-glial interactions and CNS myelination. In this publication, we reviewed and discussed the most recent knowledge on OL development and myelin formation, with a focus on mechanisms regulating these fundamental biological processes in the zebrafish. Especially, we highlighted the critical function of axons and oligodendroglia modes of communications and calcium signaling in myelin sheath formation and growth. Finally, we reviewed the relevance of these knowledge's in demyelinating diseases and drug discovery of pharmacological compounds favoring myelin regeneration.
Collapse
Affiliation(s)
- Mary-Amélie Masson
- Sorbonne Université, Institut du Cerveau, Paris Brain Institute - ICM, Inserm, CNRS, APHP, Hôpital de la Pitié-Salpêtrière, Paris, France
| | - Brahim Nait-Oumesmar
- Sorbonne Université, Institut du Cerveau, Paris Brain Institute - ICM, Inserm, CNRS, APHP, Hôpital de la Pitié-Salpêtrière, Paris, France
| |
Collapse
|
5
|
Cell–Cell Contact Mediates Gene Expression and Fate Choice of Human Neural Stem/Progenitor Cells. Cells 2022; 11:cells11111741. [PMID: 35681435 PMCID: PMC9179342 DOI: 10.3390/cells11111741] [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: 03/14/2022] [Revised: 04/21/2022] [Accepted: 05/12/2022] [Indexed: 02/04/2023] Open
Abstract
Transplantation of Neural Stem/Progenitor Cells (NPCs) is a promising regenerative strategy to promote neural repair following injury and degeneration because of the ability of these cells to proliferate, migrate, and integrate with the host tissue. Precise in vitro control of NPC proliferation without compromising multipotency and differentiation ability is critical in stem cell maintenance. This idea was highlighted in recent clinical trials, where discrepancies in NPC culturing protocols produced inconsistent therapeutic benefits. Of note, cell density plays an important role in regulating the survival, proliferation, differentiation, and fate choice of stem cells. To determine the extent of variability produced by inconsistent culturing densities, the present study cultured human-induced pluripotent NPCs (hiPSC-NPCs) at either a low or high plating density. hiPSC-NPCs were then isolated for transcriptomic analysis or differentiation in vitro. Following sequencing analysis, genes involved in cell–cell contact-mediated pathways, including Hippo-signaling, NOTCH, and WNT were differentially expressed. Modulation of these pathways was highly associated with the regulation of pro-neuronal transcription factors, which were also upregulated in response to higher-density hiPSC-NPC culture. Moreover, higher plating density translated into a greater neuronal and less astrocytic differentiation in vitro. This study highlights the importance of precisely controlling culture conditions during the development of NPC transplantation therapies.
Collapse
|
6
|
Garcia-Martin G, Alcover-Sanchez B, Wandosell F, Cubelos B. Pathways Involved in Remyelination after Cerebral Ischemia. Curr Neuropharmacol 2022; 20:751-765. [PMID: 34151767 PMCID: PMC9878953 DOI: 10.2174/1570159x19666210610093658] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 05/05/2021] [Accepted: 05/12/2021] [Indexed: 11/22/2022] Open
Abstract
Brain ischemia, also known as ischemic stroke, occurs when there is a lack of blood supply into the brain. When an ischemic insult appears, both neurons and glial cells can react in several ways that will determine the severity and prognosis. This high heterogeneity of responses has been a major obstacle in developing effective treatments or preventive methods for stroke. Although white matter pathophysiology has not been deeply assessed in stroke, its remodelling can greatly influence the clinical outcome and the disability degree. Oligodendrocytes, the unique cell type implied in CNS myelination, are sensible to ischemic damage. Loss of myelin sheaths can compromise axon survival, so new Oligodendrocyte Precursor Cells are required to restore brain function. Stroke can, therefore, enhance oligodendrogenesis to regenerate those new oligodendrocytes that will ensheath the damaged axons. Given that myelination is a highly complex process that requires coordination of multiple pathways such as Sonic Hedgehog, RTKs or Wnt/β-catenin, we will analyse new research highlighting their importance after brain ischemia. In addition, oligodendrocytes are not isolated cells inside the brain, but rather form part of a dynamic environment of interactions between neurons and glial cells. For this reason, we will put some context into how microglia and astrocytes react against stroke and influence oligodendrogenesis to highlight the relevance of remyelination in the ischemic brain. This will help to guide future studies to develop treatments focused on potentiating the ability of the brain to repair the damage.
Collapse
Affiliation(s)
- Gonzalo Garcia-Martin
- Departamento de Biología Molecular and Centro Biología Molecular “Severo Ochoa”, Universidad Autónoma de Madrid-Consejo Superior de Investigaciones Científicas, 28049 Madrid, Spain
| | - Berta Alcover-Sanchez
- Departamento de Biología Molecular and Centro Biología Molecular “Severo Ochoa”, Universidad Autónoma de Madrid-Consejo Superior de Investigaciones Científicas, 28049 Madrid, Spain
| | - Francisco Wandosell
- Departamento de Biología Molecular and Centro Biología Molecular “Severo Ochoa”, Universidad Autónoma de Madrid-Consejo Superior de Investigaciones Científicas, 28049 Madrid, Spain
| | - Beatriz Cubelos
- Departamento de Biología Molecular and Centro Biología Molecular “Severo Ochoa”, Universidad Autónoma de Madrid-Consejo Superior de Investigaciones Científicas, 28049 Madrid, Spain,Address correspondence to this author at the Departamento de Biología Molecular, Centro de Biología Molecular Severo Ochoa, Nicolás Cabrera 1, Universidad Autónoma de Madrid, 28049 Madrid, Spain; Tel: 34-91-1964561; Fax: 34-91-1964420; E-mail:
| |
Collapse
|
7
|
Brown RI, Kawakami K, Kucenas S. A novel gene trap line for visualization and manipulation of erbb3b + neural crest and glial cells in zebrafish. Dev Biol 2022; 482:114-123. [PMID: 34932993 DOI: 10.1016/j.ydbio.2021.12.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 12/08/2021] [Accepted: 12/16/2021] [Indexed: 12/22/2022]
Abstract
Glia are a diverse and essential cell type in the vertebrate nervous system. Transgenic tools and fluorescent reporter lines are critical resources to investigate how glial subtypes develop and function. However, despite the many lines available in zebrafish, the community still lacks the ability to label all unique stages of glial development and specific subpopulations of cells. To address this issue, we screened zebrafish gene and enhancer trap lines to find a novel reporter for peripheral glial subtypes. From these, we generated the gSAIzGFFD37A transgenic line that expresses GFP in neural crest cells and central and peripheral glia. We found that the gene trap construct is located within an intron of erbb3b, a gene essential for glial development. Additionally, we confirmed that GFP+ cells express erbb3b along with sox10, a known glial marker. From our screen, we have identified the gSAIzGFFD37A line as a novel and powerful tool for studying glia in the developing zebrafish, as well as a new resource to manipulate erbb3b+ cells.
Collapse
Affiliation(s)
- Robin Isadora Brown
- Department of Biology, University of Virginia, Charlottesville, VA, 22904, USA; Program in Fundamental Neuroscience, University of Virginia, Charlottesville, VA, 22904, USA
| | - Koichi Kawakami
- Laboratory of Molecular and Developmental Biology, National Institute of Genetics, and Department of Genetics, SOKENDAI The Graduate University for Advanced Studies, Mishima, Shizuoka, 444-8540, Japan
| | - Sarah Kucenas
- Department of Biology, University of Virginia, Charlottesville, VA, 22904, USA; Program in Fundamental Neuroscience, University of Virginia, Charlottesville, VA, 22904, USA.
| |
Collapse
|
8
|
Scott K, O'Rourke R, Winkler CC, Kearns CA, Appel B. Temporal single-cell transcriptomes of zebrafish spinal cord pMN progenitors reveal distinct neuronal and glial progenitor populations. Dev Biol 2021; 479:37-50. [PMID: 34303700 PMCID: PMC8410680 DOI: 10.1016/j.ydbio.2021.07.010] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 07/19/2021] [Accepted: 07/21/2021] [Indexed: 11/20/2022]
Abstract
Ventral spinal cord progenitor cells, which express the basic helix loop helix transcription factor Olig2, sequentially produce motor neurons and oligodendrocyte precursor cells (OPCs). Following specification some OPCs differentiate as myelinating oligodendrocytes while others persist as OPCs. Though a considerable amount of work has described the molecular profiles that define motor neurons, OPCs, and oligodendrocytes, less is known about the progenitors that produce them. To identify the developmental origins and transcriptional profiles of motor neurons and OPCs, we performed single-cell RNA sequencing on isolated pMN cells from embryonic zebrafish trunk tissue at stages that encompassed motor neurogenesis, OPC specification, and initiation of oligodendrocyte differentiation. Downstream analyses revealed two distinct pMN progenitor populations: one that appears to produce neurons and one that appears to produce OPCs. This latter population, called Pre-OPCs, is marked by expression of GS Homeobox 2 (gsx2), a gene that encodes a homeobox transcription factor. Using fluorescent in situ hybridizations, we identified gsx2-expressing Pre-OPCs in the spinal cord prior to expression of canonical OPC marker genes. Our data therefore reveal heterogeneous gene expression profiles among pMN progenitors, supporting prior fate mapping evidence.
Collapse
Affiliation(s)
- Kayt Scott
- Department of Pediatrics, Section of Developmental Biology, Colorado, 80045, USA; Cell Biology, Stem Cells and Development Training Program, Colorado, 80045, USA
| | - Rebecca O'Rourke
- Department of Pediatrics, Section of Developmental Biology, Colorado, 80045, USA
| | - Caitlin C Winkler
- Department of Pediatrics, Section of Developmental Biology, Colorado, 80045, USA; RNA Bioscience Initiative and Department of Biochemistry and Molecular Genetics, Colorado, 80045, USA
| | - Christina A Kearns
- Department of Pediatrics, Section of Developmental Biology, Colorado, 80045, USA
| | - Bruce Appel
- Department of Pediatrics, Section of Developmental Biology, Colorado, 80045, USA; Children's Hospital Colorado, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA.
| |
Collapse
|
9
|
The Effects of the Olig Family on the Regulation of Spinal Cord Development and Regeneration. Neurochem Res 2021; 46:2776-2782. [PMID: 34228233 DOI: 10.1007/s11064-021-03383-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 06/15/2021] [Accepted: 06/17/2021] [Indexed: 12/22/2022]
Abstract
Neurons and glial cells in the central nervous system (CNS) are generated from neuroepithelial cells in the ventricular zone that surrounds the embryonic neural tube. The proliferation and distinct differentiation of neural precursors occurs at certain stages and are regulated by a series of transcription factors leading to the generation of neuronal and glial cell subtypes. In this manuscript, we review the effects of the Olig family, namely, members Olig1, Olig2 and Olig3, on the distinct differentiation of glial and neuronal cells in the developing spinal cord and injured neural tissue.
Collapse
|
10
|
Szu J, Wojcinski A, Jiang P, Kesari S. Impact of the Olig Family on Neurodevelopmental Disorders. Front Neurosci 2021; 15:659601. [PMID: 33859549 PMCID: PMC8042229 DOI: 10.3389/fnins.2021.659601] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Accepted: 03/08/2021] [Indexed: 12/13/2022] Open
Abstract
The Olig genes encode members of the basic helix-loop-helix (bHLH) family of transcription factors. Olig1, Olig2, and Olig3 are expressed in both the developing and mature central nervous system (CNS) and strictly regulate cellular specification and differentiation. Extensive studies have established functional roles of Olig1 and Olig2 in directing neuronal and glial formation during different stages in development. Recently, Olig2 overexpression was implicated in neurodevelopmental disorders down syndrome (DS) and autism spectrum disorder (ASD) but its influence on cognitive and intellectual defects remains unknown. In this review, we summarize the biological functions of the Olig family and how it uniquely promotes cellular diversity in the CNS. This is followed up with a discussion on how abnormal Olig2 expression impacts brain development and function in DS and ASD. Collectively, the studies described here emphasize vital features of the Olig members and their distinctive potential roles in neurodevelopmental disease states.
Collapse
Affiliation(s)
- Jenny Szu
- Department of Translational Neurosciences and Neurotherapeutics, Saint John's Cancer Institute, Providence Saint John's Health Center, Santa Monica, CA, United States
| | - Alexandre Wojcinski
- Department of Translational Neurosciences and Neurotherapeutics, Saint John's Cancer Institute, Providence Saint John's Health Center, Santa Monica, CA, United States
| | - Peng Jiang
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ, United States
| | - Santosh Kesari
- Department of Translational Neurosciences and Neurotherapeutics, Saint John's Cancer Institute, Providence Saint John's Health Center, Santa Monica, CA, United States.,Pacific Neuroscience Institute, Providence Saint John's Health Center, Santa Monica, CA, United States
| |
Collapse
|
11
|
Zhang T, Guan P, Liu W, Zhao G, Fang Y, Fu H, Gui JF, Li G, Liu JX. Copper stress induces zebrafish central neural system myelin defects via WNT/NOTCH-hoxb5b signaling and pou3f1/fam168a/fam168b DNA methylation. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2020; 1863:194612. [PMID: 32745624 DOI: 10.1016/j.bbagrm.2020.194612] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Revised: 07/18/2020] [Accepted: 07/27/2020] [Indexed: 12/21/2022]
Abstract
Unbalanced copper (Cu) homeostasis is associated with neurological development defects and diseases. However, the molecular mechanisms remain elusive. Here, central neural system (CNS) myelin defects and the down-regulated expression of WNT/NOTCH signaling and its down-stream mediator hoxb5b were observed in Cu2+ stressed zebrafish larvae. The loss/knockdown-of-function of hoxb5b phenocopied the myelin and axon defects observed in Cu2+ stressed embryos. Meanwhile, the activation of WNT/NOTCH signaling and ectopic expression of hoxb5b could rescue Cu induced myelin defects. Additionally, fam168b, similar to pou3f1/2, exhibited significant promoter hypermethylation and reduced expression in Cu2+ stressed embryos. The hypermethylated locus in fam168b promoter acted pivotally in its transcription, and the loss/knockdown of fam168b/pou3f1 also induced myelin defects. This study also demonstrated that fam168b/pou3f1 and hoxb5b axis acted in a seesaw manner during fish embryogenesis: Cu induced the down-regulated expression of the WNT&NOTCH-hoxb5b axis through the function of copper transporter cox17, coupled with the promoter methylation of genes fam168b/pou3f1, and its subsequent down-regulated expression through the function of another transporter atp7b, making joint contributions to myelin defects in embryos.
Collapse
Affiliation(s)
- Ting Zhang
- College of Fisheries, Key Laboratory of Freshwater Animal Breeding, Ministry of Agriculture, Huazhong Agricultural University, Wuhan 430070, China; Key Laboratory of Tropical Marine Bio-resources and Ecology, Guangdong Provincial Key Laboratory of Applied Marine Biology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou 510301, China
| | - PengPeng Guan
- College of Informatics, Agricultural Bioinformatics Key Laboratory of Hubei Province, Hubei Engineering Technology Research Center of Agricultural Big Data, Huazhong Agricultural University, Wuhan 430070, China
| | - WenYe Liu
- College of Fisheries, Key Laboratory of Freshwater Animal Breeding, Ministry of Agriculture, Huazhong Agricultural University, Wuhan 430070, China
| | - Guang Zhao
- College of Fisheries, Key Laboratory of Freshwater Animal Breeding, Ministry of Agriculture, Huazhong Agricultural University, Wuhan 430070, China
| | - YaPing Fang
- College of Informatics, Agricultural Bioinformatics Key Laboratory of Hubei Province, Hubei Engineering Technology Research Center of Agricultural Big Data, Huazhong Agricultural University, Wuhan 430070, China
| | - Hui Fu
- Department of Anatomy, School of Basic Medical Science, Wuhan University, Wuhan 430072, China
| | - Jian-Fang Gui
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China
| | - GuoLiang Li
- College of Informatics, Agricultural Bioinformatics Key Laboratory of Hubei Province, Hubei Engineering Technology Research Center of Agricultural Big Data, Huazhong Agricultural University, Wuhan 430070, China.
| | - Jing-Xia Liu
- College of Fisheries, Key Laboratory of Freshwater Animal Breeding, Ministry of Agriculture, Huazhong Agricultural University, Wuhan 430070, China.
| |
Collapse
|
12
|
Functionally distinct subgroups of oligodendrocyte precursor cells integrate neural activity and execute myelin formation. Nat Neurosci 2020; 23:363-374. [PMID: 32066987 PMCID: PMC7292734 DOI: 10.1038/s41593-019-0581-2] [Citation(s) in RCA: 151] [Impact Index Per Article: 30.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Accepted: 12/20/2019] [Indexed: 01/06/2023]
Abstract
Recent reports have revealed oligodendrocyte precursor cell (OPC)
heterogeneity. It remains unclear if such heterogeneity reflects different
subtypes of cells with distinct functions, or rather transiently acquired states
of cells with the same function. By integrating lineage formation of individual
OPC clones, single-cell transcriptomics, calcium imaging and neural activity
manipulation, we show that OPCs in the zebrafish spinal cord can be divided into
two functionally distinct groups. One subgroup forms elaborate networks of
processes and exhibits a high degree of calcium signalling, but infrequently
differentiates, despite contact with permissive axons. Instead, these OPCs
divide in an activity and calcium dependent manner to produce another subgroup
with higher process motility and less calcium signaling, which readily
differentiates. Our data show that OPC subgroups are functionally diverse in
responding to neurons and reveal that activity regulates proliferation of a
subset of OPCs that is distinct from the cells that generate differentiated
oligodendrocytes.
Collapse
|
13
|
Triclosan induces zebrafish neurotoxicity by abnormal expression of miR-219 targeting oligodendrocyte differentiation of central nervous system. Arch Toxicol 2020; 94:857-871. [PMID: 32060586 DOI: 10.1007/s00204-020-02661-1] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Accepted: 02/03/2020] [Indexed: 12/21/2022]
Abstract
Triclosan (TCS) is ubiquitous in a wide range of personal care and consumer products, and it is acute/chronic exposure may result in several nervous system disorders. Previous studies demonstrated TCS-induced abnormal expression of miRNAs, but no investigations focused on upstream changes of miRNAs and associated molecular mechanisms. Herein, phenotype observation and behavioral analysis confirmed that TCS exposure (0, 62.5, 125, 250 μg/L) led to developmental neurotoxicity in zebrafish larvae, especially for oligodendrocyte precursor cells (OPCs). High-throughput sequencing demonstrated the critical role of miR-219 in the differentiation of OPCs. Larvae with miR-219 depletion showed the same phenotype caused by TCS. Functional tests with miR-219 knock-down and over-expression showed that miR-219 promoted differentiation of OPCs by acting on myelination inhibitors. The miR-219 also protected against TCS-induced inhibition of cell differentiation. Several epigenetic features were identified to reveal potential upstream regulatory mechanisms of miR-219. In particular, five CpG islands hyper-methylated with increasing TCS concentrations in the promoter region of miR-219. TCS inhibited OPC differentiation by influencing epigenetic effects on miR-219-related pathways, contributing to severe neurotoxicity. These findings enhance our understanding of epigenetic mechanisms affecting demyelination diseases due to TCS exposure, and also provide theoretical guidance for early intervention and gene therapy of environmentally induced diseases.
Collapse
|
14
|
Laouarem Y, Traiffort E. Developmental and Repairing Production of Myelin: The Role of Hedgehog Signaling. Front Cell Neurosci 2018; 12:305. [PMID: 30237763 PMCID: PMC6135882 DOI: 10.3389/fncel.2018.00305] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Accepted: 08/22/2018] [Indexed: 11/13/2022] Open
Abstract
Since the discovery of its role as a morphogen directing ventral patterning of the spinal cord, the secreted protein Sonic Hedgehog (Shh) has been implicated in a wide array of events contributing to the development, maintenance and repair of the central nervous system (CNS). One of these events is the generation of oligodendrocytes, the glial cells of the CNS responsible for axon myelination. In embryo, the first oligodendroglial cells arise from the ventral ventricular zone in the developing brain and spinal cord where Shh induces the basic helix-loop-helix transcription factors Olig1 and Olig2 both necessary and sufficient for oligodendrocyte production. Later on, Shh signaling participates in the production of oligodendroglial cells in the dorsal ventricular-subventricular zone in the postnatal forebrain. Finally, the modulation of Hedgehog signaling activity promotes the repair of demyelinated lesions. This mini-review article focuses on the Shh-dependent molecular mechanisms involved in the spatial and temporal control of oligodendrocyte lineage appearance. The apparent intricacy of the roles of two essential components of Shh signaling, Smoothened and Gli1, in the postnatal production of myelin and its regeneration following a demyelinating event is also highlighted. A deeper understanding of the implication of each of the components that regulate oligodendrogenesis and myelination should beneficially influence the therapeutic strategies in the field of myelin diseases.
Collapse
Affiliation(s)
| | - Elisabeth Traiffort
- Small Molecules of Neuroprotection, Neuroregeneration and Remyelination – U1195, INSERM, University Paris-Sud/Paris-Saclay, Kremlin-Bicêtre, France
| |
Collapse
|
15
|
刘 川, 林 春, 郭 培, 张 昕, 朱 晓. [Exposure to propofol down-regulates myelin basic protein expression in zebrafish embryos: its neurotoxicity on oligodendrocytes and the molecular mechanisms]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2018; 38:1115-1120. [PMID: 30377113 PMCID: PMC6744183 DOI: 10.12122/j.issn.1673-4254.2018.09.15] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Indexed: 11/24/2022]
Abstract
OBJECTIVE To investigate the mechanism underlying propofol- induced down-regulation of myelin basic protein (MBP) in zebrafish embryos. METHODS Zebrafish embryos (6-48 h post-fertilization [hpf]) were randomized into 4 equal groups for exposure to dimethyl sulfoxide (DMSO), 20 μg/mL propofol, 30 μg/mL propofol, or no particular treatment (control group). The larvae were collected at 48 or 72 hpf for detecting the mRNA levels of MBP, Olig1, Olig2, and Sox10 using qRT-PCR (n=80). The protein expression of MBP was quantitatively detected using Western blotting (n=80), and the apoptosis of the oligodendrocytes was investigated using TUNEL staining (n=6). RESULTS Exposure to 20 and 30 μg/mL propofol caused significant reductions in the mRNA expressions of Olig1, Olig2, and Sox10 at 48 and 72 hpf (P < 0.05) and also in MBP mRNA and protein levels at 72 hpf (P < 0.05). Exposure to 30 μg/mL propofol induced more obvious reduction in MBP protein expression than 20 μg/mL propofol at 72 hpf (P < 0.05), and the exposures resulted in a significant increase of oligodendrocyte apoptosis at 72 hpf (P < 0.05). CONCLUSIONS Propofol exposure reduces MBP expression at both the mRNA and protein levels in zebrafish embryos by down-regulating the expressions of Olig1, Olig2 and Sox10 mRNA levels and increasing apoptosis of the oligodendrocytes.
Collapse
Affiliation(s)
- 川 刘
- />南方医科大学南方医院麻醉科,广东 广州 510515Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - 春水 林
- />南方医科大学南方医院麻醉科,广东 广州 510515Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - 培培 郭
- />南方医科大学南方医院麻醉科,广东 广州 510515Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - 昕 张
- />南方医科大学南方医院麻醉科,广东 广州 510515Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - 晓勤 朱
- />南方医科大学南方医院麻醉科,广东 广州 510515Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| |
Collapse
|
16
|
Fuller TD, Westfall TA, Das T, Dawson DV, Slusarski DC. High-throughput behavioral assay to investigate seizure sensitivity in zebrafish implicates ZFHX3 in epilepsy. J Neurogenet 2018; 32:92-105. [PMID: 29718741 DOI: 10.1080/01677063.2018.1445247] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Epilepsy, which affects ∼1% of the population, is caused by abnormal synchronous neural activity in the central nervous system (CNS). While there is a significant genetic contribution to epilepsy, the underlying causes for the majority of genetic cases remain unknown. The NIH Undiagnosed Diseases Project (UDP) utilized exome sequencing to identify genetic variants in patients affected by various conditions with undefined etiology, including epilepsy. Confirming the functional relevance of the candidate genes identified by exome sequencing in a timely manner is crucial to translating exome data into clinically useful information. To this end, we developed a high throughput version of a seizure-sensitivity assay in zebrafish (Danio rerio) to rapidly evaluate candidate genes found by exome sequencing. We developed open access software, Studying Epilepsy In Zebrafish using R (SEIZR), to efficiently analyze the data. SEIZR was validated by disrupting function of a known epilepsy gene, prickle 1. Next, using SEIZR, we analyzed a candidate gene from the UDP screen (Zinc Finger Homeobox 3, ZFHX3), and showed that reduced ZFHX3 function in zebrafish results in a significant hyperactive response to the convulsant drug pentylenetetrazol (PTZ). We find that ZFHX3 shows strong expression in the CNS during neurogenesis including in the pallium, thalamus, tegmentum, reticular formation, and medulla oblongata - all regions which have roles in motor control and coordination. Our findings in the zebrafish confirm human ZFHX3 is a strong candidate for further neurological studies. We offer SEIZR to other researchers as a tool to rapidly and efficiently analyze large behavioral data sets.
Collapse
Affiliation(s)
- Tyson D Fuller
- a Department of Biology , University of Iowa , Iowa City , IA , USA.,b Interdisciplinary Graduate Program in Genetics , University of Iowa , Iowa City , IA , USA
| | - Trudi A Westfall
- a Department of Biology , University of Iowa , Iowa City , IA , USA
| | - Tirthasree Das
- a Department of Biology , University of Iowa , Iowa City , IA , USA
| | - Deborah V Dawson
- b Interdisciplinary Graduate Program in Genetics , University of Iowa , Iowa City , IA , USA.,c Iowa Institute for Oral Health Research , University of Iowa , Iowa City , IA , USA.,d Department of Biostatistics , University of Iowa , Iowa City , IA , USA
| | | |
Collapse
|
17
|
Matsuoka RL, Rossi A, Stone OA, Stainier DYR. CNS-resident progenitors direct the vascularization of neighboring tissues. Proc Natl Acad Sci U S A 2017; 114:10137-10142. [PMID: 28855341 PMCID: PMC5617242 DOI: 10.1073/pnas.1619300114] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Organ growth requires the coordinated invasion and expansion of blood vessel networks directed by tissue-resident cells and morphogenetic cues. A striking example of this intercellular communication is the vascularization of the central nervous system (CNS), which is driven by neuronal progenitors, including neuroepithelial cells and radial glia. Although the importance of neuronal progenitors in vascular development within the CNS is well recognized, how these progenitors regulate the vasculature outside the CNS remains largely unknown. Here we show that CNS-resident radial glia direct the vascularization of neighboring tissues during development. We find that genetic ablation of radial glia in zebrafish larvae leads to a complete loss of the bilateral vertebral arteries (VTAs) that extend along the ventrolateral sides of the spinal cord. Importantly, VTA formation is not affected by ablation of other CNS cell types, and radial glia ablation also compromises the subsequent formation of the peri-neural vascular plexus (PNVP), a vascular network that surrounds the CNS and is critical for CNS angiogenesis. Mechanistically, we find that radial glia control these processes via Vegfab/Vegfr2 signaling: vegfab is expressed by radial glia, and genetic or pharmacological inhibition of Vegfab/Vegfr2 signaling blocks the formation of the VTAs and subsequently of the PNVP. Moreover, mosaic overexpression of Vegfab in radial glia is sufficient to partially rescue the VTA formation defect in vegfab mutants. Thus, our findings identify a critical function for CNS-resident progenitors in the regulation of vascularization outside the CNS, serving as a paradigm for cross-tissue coordination of vascular morphogenesis and growth.
Collapse
Affiliation(s)
- Ryota L Matsuoka
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim 61231, Germany
| | - Andrea Rossi
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim 61231, Germany
| | - Oliver A Stone
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim 61231, Germany
| | - Didier Y R Stainier
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim 61231, Germany
| |
Collapse
|
18
|
Anand SK, Mondal AC. Cellular and molecular attributes of neural stem cell niches in adult zebrafish brain. Dev Neurobiol 2017; 77:1188-1205. [PMID: 28589616 DOI: 10.1002/dneu.22508] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2016] [Revised: 04/05/2017] [Accepted: 06/02/2017] [Indexed: 12/20/2022]
Abstract
Adult neurogenesis is a complex, presumably conserved phenomenon in vertebrates with a broad range of variations regarding neural progenitor/stem cell niches, cellular composition of these niches, migratory patterns of progenitors and so forth among different species. Current understanding of the reasons underlying the inter-species differences in adult neurogenic potential, the identification and characterization of various neural progenitors, characterization of the permissive environment of neural stem cell niches and other important aspects of adult neurogenesis is insufficient. In the last decade, zebrafish has emerged as a very useful model for addressing these questions. In this review, we have discussed the present knowledge regarding the neural stem cell niches in adult zebrafish brain as well as their cellular and molecular attributes. We have also highlighted their similarities and differences with other vertebrate species. In the end, we shed light on some of the known intrinsic and extrinsic factors that are assumed to regulate the neurogenic process in adult zebrafish brain. © 2017 Wiley Periodicals, Inc. Develop Neurobiol 77: 1188-1205, 2017.
Collapse
Affiliation(s)
- Surendra Kumar Anand
- Cellular and Molecular Neurobiology Lab, School of Life Sciences, Jawaharlal Nehru University, New Mehrauli Road, New Delhi, India, 110067
| | - Amal Chandra Mondal
- Cellular and Molecular Neurobiology Lab, School of Life Sciences, Jawaharlal Nehru University, New Mehrauli Road, New Delhi, India, 110067
| |
Collapse
|
19
|
Abstract
Myelination by oligodendrocytes in the central nervous system (CNS) and Schwann cells in the peripheral nervous system is essential for nervous system function and health. Despite its importance, we have a relatively poor understanding of the molecular and cellular mechanisms that regulate myelination in the living animal, particularly in the CNS. This is partly due to the fact that myelination commences around birth in mammals, by which time the CNS is complex and largely inaccessible, and thus very difficult to image live in its intact form. As a consequence, in recent years much effort has been invested in the use of smaller, simpler, transparent model organisms to investigate mechanisms of myelination in vivo. Although the majority of such studies have employed zebrafish, the Xenopus tadpole also represents an important complementary system with advantages for investigating myelin biology in vivo. Here we review how the natural features of zebrafish embryos and larvae and Xenopus tadpoles make them ideal systems for experimentally interrogating myelination by live imaging. We outline common transgenic technologies used to generate zebrafish and Xenopus that express fluorescent reporters, which can be used to image myelination. We also provide an extensive overview of the imaging modalities most commonly employed to date to image the nervous system in these transparent systems, and also emerging technologies that we anticipate will become widely used in studies of zebrafish and Xenopus myelination in the near future.
Collapse
Affiliation(s)
- Jenea M Bin
- Centre for Neuroregeneration, MS Society Centre for Translational Research, Euan MacDonald Centre for Motor Neurone Disease Research, University of Edinburgh, Edinburgh, UK
| | - David A Lyons
- Centre for Neuroregeneration, MS Society Centre for Translational Research, Euan MacDonald Centre for Motor Neurone Disease Research, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
20
|
Abstract
Myelin is a lipid-rich sheath formed by the spiral wrapping of specialized glial cells around axon segments. Myelinating glia allow for rapid transmission of nerve impulses and metabolic support of axons, and the absence of or disruption to myelin results in debilitating motor, cognitive, and emotional deficits in humans. Because myelin is a jawed vertebrate innovation, zebrafish are one of the simplest vertebrate model systems to study the genetics and development of myelinating glia. The morphogenetic cellular movements and genetic program that drive myelination are conserved between zebrafish and mammals, and myelin develops rapidly in zebrafish larvae, within 3-5days postfertilization. Myelin ultrastructure can be visualized in the zebrafish from larval to adult stages via transmission electron microscopy, and the dynamic development of myelinating glial cells may be observed in vivo via transgenic reporter lines in zebrafish larvae. Zebrafish are amenable to genetic and pharmacological screens, and screens for myelinating glial phenotypes have revealed both genes and drugs that promote myelin development, many of which are conserved in mammalian glia. Recently, zebrafish have been employed as a model to understand the complex dynamics of myelinating glia during development and regeneration. In this chapter, we describe these key methodologies and recent insights into mechanisms that regulate myelination using the zebrafish model.
Collapse
Affiliation(s)
- M D'Rozario
- Washington University School of Medicine, St. Louis, MO, United States
| | - K R Monk
- Washington University School of Medicine, St. Louis, MO, United States; Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO, United States
| | | |
Collapse
|
21
|
Traiffort E, Zakaria M, Laouarem Y, Ferent J. Hedgehog: A Key Signaling in the Development of the Oligodendrocyte Lineage. J Dev Biol 2016; 4:jdb4030028. [PMID: 29615592 PMCID: PMC5831774 DOI: 10.3390/jdb4030028] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2016] [Revised: 08/26/2016] [Accepted: 08/31/2016] [Indexed: 01/11/2023] Open
Abstract
The Hedgehog morphogen aroused an enormous interest since it was characterized as an essential signal for ventral patterning of the spinal cord two decades ago. The pathway is notably implicated in the initial appearance of the progenitors of oligodendrocytes (OPCs), the glial cells of the central nervous system which after maturation are responsible for axon myelination. In accordance with the requirement for Hedgehog signaling in ventral patterning, the earliest identifiable cells in the oligodendrocyte lineage are derived from the ventral ventricular zone of the developing spinal cord and brain. Here, we present the current knowledge about the involvement of Hedgehog signaling in the strict spatial and temporal regulation which characterizes the initiation and progression of the oligodendrocyte lineage. We notably describe the ability of the Hedgehog signaling to tightly orchestrate the appearance of specific combinations of genes in concert with other pathways. We document the molecular mechanisms controlling Hedgehog temporal activity during OPC specification. The contribution of the pathway to aspects of OPC development different from their specification is also highlighted especially in the optic nerve. Finally, we report the data demonstrating that Hedgehog signaling-dependency is not a universal situation for oligodendrocyte generation as evidenced in the dorsal spinal cord in contrast to the dorsal forebrain.
Collapse
Affiliation(s)
- Elisabeth Traiffort
- Neuroprotective, Neuroregenerative and Remyelinating Small Molecules' U1195, INSERM-Université Paris-Sud, Université Paris-Saclay, 80 rue du Général Leclerc, Kremlin-Bicêtre F-94276, France.
| | - Mary Zakaria
- Neuroprotective, Neuroregenerative and Remyelinating Small Molecules' U1195, INSERM-Université Paris-Sud, Université Paris-Saclay, 80 rue du Général Leclerc, Kremlin-Bicêtre F-94276, France.
| | - Yousra Laouarem
- Neuroprotective, Neuroregenerative and Remyelinating Small Molecules' U1195, INSERM-Université Paris-Sud, Université Paris-Saclay, 80 rue du Général Leclerc, Kremlin-Bicêtre F-94276, France.
| | - Julien Ferent
- IRCM, Molecular Biology of Neural Development, 110 Pine Avenue West, Montreal, QC H2W 1R7, Canada.
| |
Collapse
|
22
|
Abstract
In the nervous system, axons transmit information in the form of electrical impulses over long distances. The speed of impulse conduction is enhanced by myelin, a lipid-rich membrane that wraps around axons. Myelin also is required for the long-term health of axons by providing metabolic support. Accordingly, myelin deficiencies are implicated in a wide range of neurodevelopmental and neuropsychiatric disorders, intellectual disabilities, and neurodegenerative conditions. Central nervous system myelin is formed by glial cells called oligodendrocytes. During development, oligodendrocyte precursor cells migrate from their origins to their target axons, extend long membrane processes that wrap axons, and produce the proteins and lipids that provide myelin membrane with its unique characteristics. Myelination is a dynamic process that involves intricate interactions between multiple cell types. Therefore, an in vivo myelination model, such as the zebrafish, which allows for live observation of cell dynamics and cell-to-cell interactions, is well suited for investigating oligodendrocyte development. Zebrafish offer several advantages to investigating myelination, including the use of transgenic reporter lines, live imaging, forward genetic screens, chemical screens, and reverse genetic approaches. This chapter will describe how these tools and approaches have provided new insights into the regulatory mechanisms that guide myelination.
Collapse
Affiliation(s)
- E S Mathews
- University of Colorado School of Medicine, Aurora, CO, United States
| | - B Appel
- University of Colorado School of Medicine, Aurora, CO, United States
| |
Collapse
|
23
|
Czopka T. Insights into mechanisms of central nervous system myelination using zebrafish. Glia 2015; 64:333-49. [PMID: 26250418 DOI: 10.1002/glia.22897] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2015] [Revised: 07/14/2015] [Accepted: 07/15/2015] [Indexed: 12/12/2022]
Abstract
Myelin is the multi-layered membrane that surrounds most axons and is produced by oligodendrocytes in the central nervous system (CNS). In addition to its important role in enabling rapid nerve conduction, it has become clear in recent years that myelin plays additional vital roles in CNS function. Myelinating oligodendrocytes provide metabolic support to axons and active myelination is even involved in regulating forms of learning and memory formation. However, there are still large gaps in our understanding of how myelination by oligodendrocytes is regulated. The small tropical zebrafish has become an increasingly popular model organism to investigate many aspects of nervous system formation, function, and regeneration. This is mainly due to two approaches for which the zebrafish is an ideally suited vertebrate model--(1) in vivo live cell imaging using vital dyes and genetically encoded reporters, and (2) gene and target discovery using unbiased screens. This review summarizes how the use of zebrafish has helped understand mechanisms of oligodendrocyte behavior and myelination in vivo and discusses the potential use of zebrafish to shed light on important future questions relating to myelination in the context of CNS development, function and repair.
Collapse
Affiliation(s)
- Tim Czopka
- Institute of Neuronal Cell Biology, Technische Universität München, Munich, Germany
| |
Collapse
|
24
|
Abstract
The zebrafish is a premier vertebrate model system that offers many experimental advantages for in vivo imaging and genetic studies. This review provides an overview of glial cell types in the central and peripheral nervous system of zebrafish. We highlight some recent work that exploited the strengths of the zebrafish system to increase the understanding of the role of Gpr126 in Schwann cell myelination and illuminate the mechanisms controlling oligodendrocyte development and myelination. We also summarize similarities and differences between zebrafish radial glia and mammalian astrocytes and consider the possibility that their distinct characteristics may represent extremes in a continuum of cell identity. Finally, we focus on the emergence of zebrafish as a model for elucidating the development and function of microglia. These recent studies have highlighted the power of the zebrafish system for analyzing important aspects of glial development and function.
Collapse
Affiliation(s)
- David A Lyons
- Centre for Neuroregeneration, University of Edinburgh, Edinburgh EH16 4SB, United Kingdom
| | - William S Talbot
- Department of Developmental Biology, Stanford University, Stanford, California 94305
| |
Collapse
|
25
|
Preston MA, Macklin WB. Zebrafish as a model to investigate CNS myelination. Glia 2014; 63:177-93. [PMID: 25263121 DOI: 10.1002/glia.22755] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2014] [Accepted: 09/12/2014] [Indexed: 12/18/2022]
Abstract
Myelin plays a critical role in proper neuronal function by providing trophic and metabolic support to axons and facilitating energy-efficient saltatory conduction. Myelination is influenced by numerous molecules including growth factors, hormones, transmembrane receptors and extracellular molecules, which activate signaling cascades that drive cellular maturation. Key signaling molecules and downstream signaling cascades controlling myelination have been identified in cell culture systems. However, in vitro systems are not able to faithfully replicate the complex in vivo signaling environment that occurs during development or following injury. Currently, it remains time-consuming and expensive to investigate myelination in vivo in rodents, the most widely used model for studying mammalian myelination. As such, there is a need for alternative in vivo myelination models, particularly ones that can test molecular mechanisms without removing oligodendrocyte lineage cells from their native signaling environment or disrupting intercellular interactions with other cell types present during myelination. Here, we review the ever-increasing role of zebrafish in studies uncovering novel mechanisms controlling vertebrate myelination. These innovative studies range from observations of the behavior of single cells during in vivo myelination as well as mutagenesis- and pharmacology-based screens in whole animals. Additionally, we discuss recent efforts to develop novel models of demyelination and oligodendrocyte cell death in adult zebrafish for the study of cellular behavior in real time during repair and regeneration of damaged nervous systems.
Collapse
Affiliation(s)
- Marnie A Preston
- Department of Cell and Developmental Biology, University of Colorado School of Medicine, Aurora, Colorado
| | | |
Collapse
|
26
|
Zada D, Tovin A, Lerer-Goldshtein T, Vatine GD, Appelbaum L. Altered behavioral performance and live imaging of circuit-specific neural deficiencies in a zebrafish model for psychomotor retardation. PLoS Genet 2014; 10:e1004615. [PMID: 25255244 PMCID: PMC4177677 DOI: 10.1371/journal.pgen.1004615] [Citation(s) in RCA: 71] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2014] [Accepted: 07/18/2014] [Indexed: 11/28/2022] Open
Abstract
The mechanisms and treatment of psychomotor retardation, which includes motor and cognitive impairment, are indefinite. The Allan-Herndon-Dudley syndrome (AHDS) is an X-linked psychomotor retardation characterized by delayed development, severe intellectual disability, muscle hypotonia, and spastic paraplegia, in combination with disturbed thyroid hormone (TH) parameters. AHDS has been associated with mutations in the monocarboxylate transporter 8 (mct8/slc16a2) gene, which is a TH transporter. In order to determine the pathophysiological mechanisms of AHDS, MCT8 knockout mice were intensively studied. Although these mice faithfully replicated the abnormal serum TH levels, they failed to exhibit the neurological and behavioral symptoms of AHDS patients. Here, we generated an mct8 mutant (mct8−/−) zebrafish using zinc-finger nuclease (ZFN)-mediated targeted gene editing system. The elimination of MCT8 decreased the expression levels of TH receptors; however, it did not affect the expression of other TH-related genes. Similar to human patients, mct8−/− larvae exhibited neurological and behavioral deficiencies. High-throughput behavioral assays demonstrated that mct8−/− larvae exhibited reduced locomotor activity, altered response to external light and dark transitions and an increase in sleep time. These deficiencies in behavioral performance were associated with altered expression of myelin-related genes and neuron-specific deficiencies in circuit formation. Time-lapse imaging of single-axon arbors and synapses in live mct8−/− larvae revealed a reduction in filopodia dynamics and axon branching in sensory neurons and decreased synaptic density in motor neurons. These phenotypes enable assessment of the therapeutic potential of three TH analogs that can enter the cells in the absence of MCT8. The TH analogs restored the myelin and axon outgrowth deficiencies in mct8−/− larvae. These findings suggest a mechanism by which MCT8 regulates neural circuit assembly, ultimately mediating sensory and motor control of behavioral performance. We also propose that the administration of TH analogs early during embryo development can specifically reduce neurological damage in AHDS patients. In a wide range of brain disorders, mutations in specific genes cause alterations in the development and function of neural circuits that ultimately affect behavior. A major challenge is to uncover the mechanism and provide treatment which is capable of preventing brain damage. Allan-Herndon-Dudley syndrome (AHDS) is a severe psychomotor retardation characterized by intellectual disabilities, neurological impairment and abnormal thyroid hormone (TH) levels. Mutations in the TH transporter MCT8 are associated with AHDS. Mice that lack the MCT8 protein exhibited impaired TH levels, as is the case in human patients; however, they lack neurological defects. Here, we generated an mct8 mutant (mct8−/−) zebrafish, which exhibited neurological and behavioral deficiencies and mimics pathological conditions of AHDS patients. The zebrafish is a simple transparent vertebrate and its nervous system is conserved with mammals. Time-lapse live imaging of single axons and synapses, and video-tracking of behavior revealed deficiencies in neural circuit assembly, which are associated with disturbed sleep and altered locomotor activity. In addition, since the mct8−/− larvae provides a highthroughput platform for testing therapeutic drugs, we showed that TH analogs can recover neurological deficiencies in an animal model for psychomotor retardation.
Collapse
Affiliation(s)
- David Zada
- The Mina & Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan, Israel
- The Leslie and Susan Gonda Multidisciplinary Brain Research Center, Bar-Ilan University, Ramat-Gan, Israel
| | - Adi Tovin
- The Mina & Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan, Israel
- The Leslie and Susan Gonda Multidisciplinary Brain Research Center, Bar-Ilan University, Ramat-Gan, Israel
| | - Tali Lerer-Goldshtein
- The Mina & Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan, Israel
- The Leslie and Susan Gonda Multidisciplinary Brain Research Center, Bar-Ilan University, Ramat-Gan, Israel
| | - Gad David Vatine
- The Mina & Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan, Israel
- The Leslie and Susan Gonda Multidisciplinary Brain Research Center, Bar-Ilan University, Ramat-Gan, Israel
- Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, California, United States of America
| | - Lior Appelbaum
- The Mina & Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan, Israel
- The Leslie and Susan Gonda Multidisciplinary Brain Research Center, Bar-Ilan University, Ramat-Gan, Israel
- * E-mail:
| |
Collapse
|
27
|
Tan B, Yu J, Yin Y, Jia G, Jiang W, Yu L. The Olig family affects central nervous system development and disease. Neural Regen Res 2014; 9:329-36. [PMID: 25206819 PMCID: PMC4146145 DOI: 10.4103/1673-5374.128232] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/15/2013] [Indexed: 11/04/2022] Open
Abstract
Neural cell differentiation and maturation is a critical step during central nervous system development. The oligodendrocyte transcription family (Olig family) is known to be an important factor in regulating neural cell differentiation. Because of this, the Olig family also affects acute and chronic central nervous system diseases, including brain injury, multiple sclerosis, and even gliomas. Improved understanding about the functions of the Olig family in central nervous system development and disease will greatly aid novel breakthroughs in central nervous system diseases. This review investigates the role of the Olig family in central nervous system development and related diseases.
Collapse
Affiliation(s)
- Botao Tan
- Department of Rehabilitation Medicine, Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Jing Yu
- Department of Rehabilitation Medicine, Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Ying Yin
- Department of Rehabilitation Medicine, Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Gongwei Jia
- Department of Rehabilitation Medicine, Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Wei Jiang
- Department of Rehabilitation Medicine, Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Lehua Yu
- Department of Rehabilitation Medicine, Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| |
Collapse
|
28
|
García-Cerro S, Martínez P, Vidal V, Corrales A, Flórez J, Vidal R, Rueda N, Arbonés ML, Martínez-Cué C. Overexpression of Dyrk1A is implicated in several cognitive, electrophysiological and neuromorphological alterations found in a mouse model of Down syndrome. PLoS One 2014; 9:e106572. [PMID: 25188425 PMCID: PMC4154723 DOI: 10.1371/journal.pone.0106572] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2014] [Accepted: 07/30/2014] [Indexed: 01/09/2023] Open
Abstract
Down syndrome (DS) phenotypes result from the overexpression of several dosage-sensitive genes. The DYRK1A (dual-specificity tyrosine-(Y)-phosphorylation regulated kinase 1A) gene, which has been implicated in the behavioral and neuronal alterations that are characteristic of DS, plays a role in neuronal progenitor proliferation, neuronal differentiation and long-term potentiation (LTP) mechanisms that contribute to the cognitive deficits found in DS. The purpose of this study was to evaluate the effect of Dyrk1A overexpression on the behavioral and cognitive alterations in the Ts65Dn (TS) mouse model, which is the most commonly utilized mouse model of DS, as well as on several neuromorphological and electrophysiological properties proposed to underlie these deficits. In this study, we analyzed the phenotypic differences in the progeny obtained from crosses of TS females and heterozygous Dyrk1A (+/-) male mice. Our results revealed that normalization of the Dyrk1A copy number in TS mice improved working and reference memory based on the Morris water maze and contextual conditioning based on the fear conditioning test and rescued hippocampal LTP. Concomitant with these functional improvements, normalization of the Dyrk1A expression level in TS mice restored the proliferation and differentiation of hippocampal cells in the adult dentate gyrus (DG) and the density of GABAergic and glutamatergic synapse markers in the molecular layer of the hippocampus. However, normalization of the Dyrk1A gene dosage did not affect other structural (e.g., the density of mature hippocampal granule cells, the DG volume and the subgranular zone area) or behavioral (i.e., hyperactivity/attention) alterations found in the TS mouse. These results suggest that Dyrk1A overexpression is involved in some of the cognitive, electrophysiological and neuromorphological alterations, but not in the structural alterations found in DS, and suggest that pharmacological strategies targeting this gene may improve the treatment of DS-associated learning disabilities.
Collapse
Affiliation(s)
- Susana García-Cerro
- Department of Physiology and Pharmacology, Faculty of Medicine, University of Cantabria, Santander, Spain
| | - Paula Martínez
- Department of Physiology and Pharmacology, Faculty of Medicine, University of Cantabria, Santander, Spain
| | - Verónica Vidal
- Department of Physiology and Pharmacology, Faculty of Medicine, University of Cantabria, Santander, Spain
| | - Andrea Corrales
- Department of Physiology and Pharmacology, Faculty of Medicine, University of Cantabria, Santander, Spain
| | - Jesús Flórez
- Department of Physiology and Pharmacology, Faculty of Medicine, University of Cantabria, Santander, Spain
| | - Rebeca Vidal
- Department of Physiology and Pharmacology, Faculty of Medicine, University of Cantabria, Santander, Spain
- Institute of Biomedicine and Biotechnology (IBBITEC), (University of Cantabria- Consejo Superior de Investigaciones Científicas (CSIC) and Investigación, Desarrollo e Investigación Cantabria (IDICAN)), Santander, Spain
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Instituto de Salud Carlos III, Madrid, Spain
| | - Noemí Rueda
- Department of Physiology and Pharmacology, Faculty of Medicine, University of Cantabria, Santander, Spain
| | - María L. Arbonés
- Barcelona Institute of Molecular Biology, Centro Superior de Investigaciones Científicas (CSIC) and Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Barcelona, Spain
| | - Carmen Martínez-Cué
- Department of Physiology and Pharmacology, Faculty of Medicine, University of Cantabria, Santander, Spain
| |
Collapse
|
29
|
The housekeeping gene hypoxanthine guanine phosphoribosyltransferase (HPRT) regulates multiple developmental and metabolic pathways of murine embryonic stem cell neuronal differentiation. PLoS One 2013; 8:e74967. [PMID: 24130677 PMCID: PMC3794013 DOI: 10.1371/journal.pone.0074967] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2013] [Accepted: 08/09/2013] [Indexed: 11/19/2022] Open
Abstract
The mechanisms by which mutations of the purinergic housekeeping gene hypoxanthine guanine phosphoribosyltransferase (HPRT) cause the severe neurodevelopmental Lesch Nyhan Disease (LND) are poorly understood. The best recognized neural consequences of HPRT deficiency are defective basal ganglia expression of the neurotransmitter dopamine (DA) and aberrant DA neuronal function. We have reported that HPRT deficiency leads to dysregulated expression of multiple DA-related developmental functions and cellular signaling defects in a variety of HPRT-deficient cells, including human induced pluripotent stem (iPS) cells. We now describe results of gene expression studies during neuronal differentiation of HPRT-deficient murine ESD3 embryonic stem cells and report that HPRT knockdown causes a marked switch from neuronal to glial gene expression and dysregulates expression of Sox2 and its regulator, genes vital for stem cell pluripotency and for the neuronal/glial cell fate decision. In addition, HPRT deficiency dysregulates many cellular functions controlling cell cycle and proliferation mechanisms, RNA metabolism, DNA replication and repair, replication stress, lysosome function, membrane trafficking, signaling pathway for platelet activation (SPPA) multiple neurotransmission systems and sphingolipid, sulfur and glycan metabolism. We propose that the neural aberrations of HPRT deficiency result from combinatorial effects of these multi-system metabolic errors. Since some of these aberrations are also found in forms of Alzheimer's and Huntington's disease, we predict that some of these systems defects play similar neuropathogenic roles in diverse neurodevelopmental and neurodegenerative diseases in common and may therefore provide new experimental opportunities for clarifying pathogenesis and for devising new potential therapeutic targets in developmental and genetic disease.
Collapse
|
30
|
de Oliveira-Carlos V, Ganz J, Hans S, Kaslin J, Brand M. Notch receptor expression in neurogenic regions of the adult zebrafish brain. PLoS One 2013; 8:e73384. [PMID: 24039926 PMCID: PMC3767821 DOI: 10.1371/journal.pone.0073384] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2012] [Accepted: 07/22/2013] [Indexed: 12/21/2022] Open
Abstract
The adult zebrash brain has a remarkable constitutive neurogenic capacity. The regulation and maintenance of its adult neurogenic niches are poorly understood. In mammals, Notch signaling is involved in stem cell maintenance both in embryonic and adult CNS. To better understand how Notch signaling is involved in stem cell maintenance during adult neurogenesis in zebrafish we analysed Notch receptor expression in five neurogenic zones of the adult zebrafish brain. Combining proliferation and glial markers we identified several subsets of Notch receptor expressing cells. We found that 90 of proliferating radial glia express notch1a, notch1b and notch3. In contrast, the proliferating non-glial populations of the dorsal telencephalon and hypothalamus rarely express notch3 and about half express notch1a/1b. In the non-proliferating radial glia notch3 is the predominant receptor throughout the brain. In the ventral telencephalon and in the mitotic area of the optic tectum, where cells have neuroepithelial properties, notch1a/1b/3 are expressed in most proliferating cells. However, in the cerebellar niche, although progenitors also have neuroepithelial properties, only notch1a/1b are expressed in a high number of PCNA cells. In this region notch3 expression is mostly in Bergmann glia and at low levels in few PCNA cells. Additionally, we found that in the proliferation zone of the ventral telencephalon, Notch receptors display an apical high to basal low gradient of expression. Notch receptors are also expressed in subpopulations of oligodendrocytes, neurons and endothelial cells. We suggest that the partial regional heterogeneity observed for Notch expression in progenitor cells might be related to the cellular diversity present in each of these neurogenic niches.
Collapse
Affiliation(s)
- Vanessa de Oliveira-Carlos
- Biotechnology Center and Center for Regenerative Therapies Dresden, Technische Universität Dresden, Dresden, Germany
| | - Julia Ganz
- Biotechnology Center and Center for Regenerative Therapies Dresden, Technische Universität Dresden, Dresden, Germany
| | - Stefan Hans
- Biotechnology Center and Center for Regenerative Therapies Dresden, Technische Universität Dresden, Dresden, Germany
| | - Jan Kaslin
- Biotechnology Center and Center for Regenerative Therapies Dresden, Technische Universität Dresden, Dresden, Germany
| | - Michael Brand
- Biotechnology Center and Center for Regenerative Therapies Dresden, Technische Universität Dresden, Dresden, Germany
- * E-mail:
| |
Collapse
|
31
|
Fleming A, Alderton W. Zebrafish in pharmaceutical industry research: finding the best fit. ACTA ACUST UNITED AC 2013. [DOI: 10.1016/j.ddmod.2012.02.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
|
32
|
Yuelling LW, Waggener CT, Afshari FS, Lister JA, Fuss B. Autotaxin/ENPP2 regulates oligodendrocyte differentiation in vivo in the developing zebrafish hindbrain. Glia 2012; 60:1605-18. [PMID: 22821873 DOI: 10.1002/glia.22381] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2011] [Accepted: 06/05/2012] [Indexed: 01/12/2023]
Abstract
During development, progenitors that are committed to differentiate into oligodendrocytes, the myelinating cells of the central nervous system (CNS), are generated within discrete regions of the neuroepithelium. More specifically, within the developing spinal cord and hindbrain ventrally located progenitor cells that are characterized by the expression of the transcription factor olig2 give temporally rise to first motor neurons and then oligodendrocyte progenitors. The regulation of this temporal neuron-glial switch has been found complex and little is known about the extrinsic factors regulating it. Our studies described here identified a zebrafish ortholog to mammalian atx, which displays evolutionarily conserved expression pattern characteristics. Most interestingly, atx was found to be expressed by cells of the cephalic floor plate during a time period when ventrally-derived oligodendrocyte progenitors arise in the developing hindbrain of the zebrafish. Knock-down of atx expression resulted in a delay and/or inhibition of the timely appearance of oligodendrocyte progenitors and subsequent developmental stages of the oligodendrocyte lineage. This effect of atx knock-down was not accompanied by changes in the number of olig2-positive progenitor cells, the overall morphology of the axonal network or the number of somatic abducens motor neurons. Thus, our studies identified Atx as an extrinsic factor that is likely secreted by cells from the floor plate and that is involved in regulating specifically the progression of olig2-positive progenitor cells into lineage committed oligodendrocyte progenitors.
Collapse
Affiliation(s)
- Larra W Yuelling
- Department of Anatomy and Neurobiology, Virginia Commonwealth University School of Medicine, Richmond, Virginia 23298, USA
| | | | | | | | | |
Collapse
|
33
|
Zannino DA, Sagerström CG, Appel B. olig2-Expressing hindbrain cells are required for migrating facial motor neurons. Dev Dyn 2012; 241:315-26. [PMID: 22275004 DOI: 10.1002/dvdy.23718] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
The complicated trajectory of facial motor neuron migration requires coordination of intrinsic signals and cues from the surrounding environment. Migration begins in rhombomere (r) 4 where facial motor neurons are born and proceeds in a caudal direction. Once facial motor neurons reach their target rhombomeres, they migrate laterally and radially from the ventral neural tube. In zebrafish, as facial motor neurons migrate through r5/r6, they pass near cells that express olig2, which encodes a bHLH transcription factor. In this study, we found that olig2 function is required for facial motor neurons to complete their caudal migration into r6 and r7 and form stereotypical clusters. Additionally, embryos that lack mafba function, in which facial motor neurons also fail to complete caudal migration, lack olig2 expression in r5 and r6. Our data raise the possibility that cells expressing olig2 are intermediate targets that help guide facial motor neuron migration.
Collapse
Affiliation(s)
- Denise A Zannino
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, Colorado 80045, USA
| | | | | |
Collapse
|
34
|
|
35
|
Abstract
The myelin sheath is an essential component of the vertebrate nervous system, and its disruption causes numerous diseases, including multiple sclerosis (MS), and neurodegeneration. Although we understand a great deal about the early development of the glial cells that make myelin (Schwann cells in the peripheral nervous system and oligodendrocytes in the central nervous system), we know much less about the cellular and molecular mechanisms that regulate the later stages of differentiation that orchestrate myelin formation. Over the past decade, the zebrafish has been employed as a model with which to dissect the development of myelinated axons. Forward genetic screens have revealed new genes essential for myelination, as well as new roles for genes previously implicated in myelinated axon formation in other systems. High-resolution in vivo imaging in zebrafish has also begun to illuminate novel cell behaviors during myelinating glial cell development. Here we review the contribution of zebrafish research to our understanding of myelinated axon formation to date. We also describe and discuss many of the methodologies used in these studies and preview future endeavors that will ensure that the zebrafish remains at the cutting edge of this important area of research.
Collapse
Affiliation(s)
- Tim Czopka
- Centre for Neuroregeneration, University of Edinburgh, Chancellor's Building, 49 Little France Crescent, Edinburgh, UK
| | | |
Collapse
|
36
|
Buckley CE, Marguerie A, Alderton WK, Franklin RJM. Temporal dynamics of myelination in the zebrafish spinal cord. Glia 2010; 58:802-12. [PMID: 20140960 DOI: 10.1002/glia.20964] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Knowledge of the precise timing of myelination is critical to the success of zebrafish-based in vivo screening strategies for potential remyelination therapies. This study provides a systematic review of the timing of myelination in the zebrafish spinal cord and a critique of techniques by which it may be accurately assessed. The onset of myelination was found to be 3 days postfertilization (d.p.f.); earlier than previously reported. This coincided with the dorsal migration and differentiation of oligodendrocytes and the expression of myelin basic protein (Mbp) transcripts and protein. Our data suggests that immunohistochemistry with zebrafish-specific anti-Mbp from 3 d.p.f. is the optimal histological method for myelin visualization, while quantification of myelination is more reliably achieved by quantitative polymerase chain reaction (qPCR) for mbp from 5 d.p.f.. Transgenic fluorescent lines such as olig2:EGFP can be used to assess oligodendrocyte cell number at 3 d.p.f. and the development of new, more specific lines may enable real time visualization of myelin itself. Quantitative ultrastructural analysis revealed that the myelination of zebrafish axons is regulated according to axonal growth and not absolute axonal size. This study confirms the use of the zebrafish larvae as a versatile and efficient in vivo model of myelination and provides a platform on which future myelination screening studies can be based.
Collapse
Affiliation(s)
- Clare E Buckley
- MRC Centre for Stem Cell Biology and Regenerative Medicine and Department of Veterinary Medicine, University of Cambridge, Cambridge, United Kingdom
| | | | | | | |
Collapse
|
37
|
Wen CM, Wang CS, Chin TC, Cheng ST, Nan FH. Immunochemical and molecular characterization of a novel cell line derived from the brain of Trachinotus blochii (Teleostei, Perciformes): A fish cell line with oligodendrocyte progenitor cell and tanycyte characteristics. Comp Biochem Physiol A Mol Integr Physiol 2010; 156:224-31. [PMID: 20167281 DOI: 10.1016/j.cbpa.2010.02.003] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2009] [Revised: 02/03/2010] [Accepted: 02/10/2010] [Indexed: 01/24/2023]
Abstract
Ependymal radial glial cells, also called tanycytes, are the predominant glial fibrillary acidic protein (GFAP)- and vimentin (VIM)-expressing cells in fish ependyma. Radial glial cells have been proposed to be neural stem cells but their molecular expression is not well understood. Previous studies revealed that fish neural progenitor and neural stem cells have A2B5, a marker for oligodendrocyte progenitor cells (OPCs). In this study, an A2B5(+) cell line, SPB, was isolated from the brain of the teleost Trachinotus blochii and characterized. SPB cells usually grew as polygonal epithelial cells, but at high density, long processes were commonly observed. Using immunocytochemistry, SPB cells were shown to exhibit oligodendrocyte markers such as galactocerebroside and Olig2, and radial glial cell markers such as brain lipid-binding protein, GFAP, Sox2, and VIM. SPB cells were also observed to have DARPP-32, a marker for tanycytes in mammals, and primary cilia. RT-PCR additionally revealed expression of bone morphogenetic protein 4, connexin35, Noggin2, and proteolipid protein in SPB cells. Results of this study suggest that SPB cells are OPCs that can display tanycyte characteristics. Fish tanycytes can be neural stem cells suggesting that SPB cells are neural stem cells. SPB is the first fish cell line showing primary cilia and markers for both OPCs and tanycytes.
Collapse
Affiliation(s)
- Chiu-Ming Wen
- Department of Life Sciences, National University of Kaohsiung, Kaohsiung, Taiwan.
| | | | | | | | | |
Collapse
|
38
|
Lamont RE, Vu W, Carter AD, Serluca FC, MacRae CA, Childs SJ. Hedgehog signaling via angiopoietin1 is required for developmental vascular stability. Mech Dev 2010; 127:159-68. [PMID: 20156556 DOI: 10.1016/j.mod.2010.02.001] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2009] [Revised: 02/05/2010] [Accepted: 02/10/2010] [Indexed: 12/20/2022]
Abstract
The molecular pathways by which newly formed, immature endothelial cell tubes remodel to form a mature network of vessels supported by perivascular mural cells are not well understood. The zebrafish iguana (igu) genetic mutant has a mutation in the daz-interacting protein 1 (dzip1), a member of the hedgehog signaling pathway. Loss of dzip1 results in decreased size of the cranial dorsal aortae, ultrastructural defects in perivascular mural cell recruitment and subsequent hemorrhage. Although hedgehog signaling is disrupted in igu mutants, we find no defects in vessel patterning or artery-vein specification. Rather, we show that the loss of angiopoietin1 (angpt1) expression in ventral perivascular mesenchyme is responsible for vascular instability in igu mutants. Over-expression of angpt1 or partial down-regulation of the endogenous Angpt1 antagonist angpt2 rescues hemorrhage. This is the first direct in vivo link between hedgehog signaling and the induction of vascular stability by recruitment of perivascular mural cells through angiopoietin signaling.
Collapse
Affiliation(s)
- Ryan E Lamont
- Biochemistry and Molecular Biology, University of Calgary, 3330 Hospital Dr NW, Calgary, AB, Canada T2N 4N1
| | | | | | | | | | | |
Collapse
|
39
|
Abstract
For more than a decade, the zebrafish has proven to be an excellent model organism to investigate the mechanisms of neurogenesis during development. The often cited advantages, namely external development, genetic, and optical accessibility, have permitted direct examination and experimental manipulations of neurogenesis during development. Recent studies have begun to investigate adult neurogenesis, taking advantage of its widespread occurrence in the mature zebrafish brain to investigate the mechanisms underlying neural stem cell maintenance and recruitment. Here we provide a comprehensive overview of the tools and techniques available to study neurogenesis in zebrafish both during development and in adulthood. As useful resources, we provide tables of available molecular markers, transgenic, and mutant lines. We further provide optimized protocols for studying neurogenesis in the adult zebrafish brain, including in situ hybridization, immunohistochemistry, in vivo lipofection and electroporation methods to deliver expression constructs, administration of bromodeoxyuridine (BrdU), and finally slice cultures. These currently available tools have put zebrafish on par with other model organisms used to investigate neurogenesis.
Collapse
Affiliation(s)
- Prisca Chapouton
- Institute of Developmental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | | |
Collapse
|
40
|
Jung SH, Kim S, Chung AY, Kim HT, So JH, Ryu J, Park HC, Kim CH. Visualization of myelination in GFP-transgenic zebrafish. Dev Dyn 2009; 239:592-7. [DOI: 10.1002/dvdy.22166] [Citation(s) in RCA: 87] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
|