1
|
Yuikawa T, Sato T, Ikeda M, Tsuruoka M, Yasuda K, Sato Y, Nasu K, Yamasu K. Elongation of the developing spinal cord is driven by Oct4-type transcription factor-mediated regulation of retinoic acid signaling in zebrafish embryos. Dev Dyn 2024; 253:404-422. [PMID: 37850839 DOI: 10.1002/dvdy.666] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 10/04/2023] [Accepted: 10/05/2023] [Indexed: 10/19/2023] Open
Abstract
BACKGROUND Elongation of the spinal cord is dependent on neural development from neuromesodermal progenitors in the tail bud. We previously showed the involvement of the Oct4-type gene, pou5f3, in this process in zebrafish mainly by dominant-interference gene induction, but, to compensate for the limitation of this transgene approach, mutant analysis was indispensable. pou5f3 involvement in the signaling pathways was another unsolved question. RESULTS We examined the phenotypes of pou5f3 mutants and the effects of Pou5f3 activation by the tamoxifen-ERT2 system in the posterior neural tube, together confirming the involvement of pou5f3. The reporter assays using P19 cells implicated tail bud-related transcription factors in pou5f3 expression. Regulation of tail bud development by retinoic acid (RA) signaling was confirmed by treatment of embryos with RA and the synthesis inhibitor, and in vitro reporter assays further showed that RA signaling regulated pou5f3 expression. Importantly, the expression of the RA degradation enzyme gene, cyp26a1, was down-regulated in embryos with disrupted pou5f3 activity. CONCLUSIONS The involvement of pou5f3 in spinal cord extension was supported by using mutants and the gain-of-function approach. Our findings further suggest that pou5f3 regulates the RA level, contributing to neurogenesis in the posterior neural tube.
Collapse
Affiliation(s)
- Tatsuya Yuikawa
- Division of Life Science, Graduate School of Science and Engineering, Saitama University, Saitama City, Saitama, Japan
| | - Takehisa Sato
- Division of Life Science, Graduate School of Science and Engineering, Saitama University, Saitama City, Saitama, Japan
| | - Masaaki Ikeda
- Division of Life Science, Graduate School of Science and Engineering, Saitama University, Saitama City, Saitama, Japan
| | - Momo Tsuruoka
- Division of Life Science, Graduate School of Science and Engineering, Saitama University, Saitama City, Saitama, Japan
| | - Kaede Yasuda
- Division of Life Science, Graduate School of Science and Engineering, Saitama University, Saitama City, Saitama, Japan
| | - Yuto Sato
- Division of Life Science, Graduate School of Science and Engineering, Saitama University, Saitama City, Saitama, Japan
| | - Kouhei Nasu
- Division of Life Science, Graduate School of Science and Engineering, Saitama University, Saitama City, Saitama, Japan
| | - Kyo Yamasu
- Division of Life Science, Graduate School of Science and Engineering, Saitama University, Saitama City, Saitama, Japan
| |
Collapse
|
2
|
Belmonte-Mateos C, Meister L, Pujades C. Hindbrain rhombomere centers harbor a heterogenous population of dividing progenitors which rely on Notch signaling. Front Cell Dev Biol 2023; 11:1268631. [PMID: 38020924 PMCID: PMC10652760 DOI: 10.3389/fcell.2023.1268631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Accepted: 10/11/2023] [Indexed: 12/01/2023] Open
Abstract
Tissue growth and morphogenesis are interrelated processes, whose tight coordination is essential for the production of different cell fates and the timely precise allocation of stem cell capacities. The zebrafish embryonic brainstem, the hindbrain, exemplifies such coupling between spatiotemporal cell diversity acquisition and tissue growth as the neurogenic commitment is differentially distributed over time. Here, we combined cell lineage and in vivo imaging approaches to reveal the emergence of specific cell population properties within the rhombomeres. We studied the molecular identity of hindbrain rhombomere centers and showed that they harbor different progenitor capacities that change over time. By clonal analysis, we revealed that cells within the center of rhombomeres decrease the proliferative capacity to remain mainly in the G1 phase. Proliferating progenitors give rise to neurons by asymmetric and symmetric neurogenic divisions while maintaining the pool of progenitors. The proliferative capacity of these cells differs from their neighbors, and they are delayed in the onset of Notch activity. Through functional studies, we demonstrated that they rely on Notch3 signaling to be maintained as non-committed progenitors. In this study, we show that cells in rhombomere centers, despite the neurogenic asynchrony, might share steps of a similar program with the rhombomere counterparts, to ensure proper tissue growth.
Collapse
|
3
|
Yuikawa T, Ikeda M, Tsuda S, Saito S, Yamasu K. Involvement of Oct4-type transcription factor Pou5f3 in posterior spinal cord formation in zebrafish embryos. Dev Growth Differ 2021; 63:306-322. [PMID: 34331767 DOI: 10.1111/dgd.12742] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2021] [Revised: 07/13/2021] [Accepted: 07/27/2021] [Indexed: 12/21/2022]
Abstract
In vertebrate embryogenesis, elongation of the posterior body is driven by de novo production of the axial and paraxial mesoderm as well as the neural tube at the posterior end. This process is presumed to depend on the stem cell-like population in the tail bud region, but the details of the gene regulatory network involved are unknown. Previous studies suggested the involvement of pou5f3, an Oct4-type POU gene in zebrafish, in axial elongation. In the present study, we first found that pou5f3 is expressed mainly in the dorsal region of the tail bud immediately after gastrulation, and that this expression is restricted to the posterior-most region of the elongating neural tube during somitogenesis. This pou5f3 expression was complementary to the broad expression of sox3 in the neural tube, and formed a sharp boundary with specific expression of tbxta (orthologue of mammalian T/Brachyury) in the tail bud, implicating pou5f3 in the specification of tail bud-derived cells toward neural differentiation in the spinal cord. When pou5f3 was functionally impaired after gastrulation by induction of a dominant-interfering pou5f3 mutant gene (en-pou5f3), trunk and tail elongation were markedly disturbed at distinct positions along the axis depending on the stage. This finding showed involvement of pou5f3 in de novo generation of the body from the tail bud. Conditional functional abrogation also showed that pou5f3 downregulates mesoderm-forming genes but promotes neural development by activating neurogenesis genes around the tail bud. These results suggest that pou5f3 is involved in formation of the posterior spinal cord.
Collapse
Affiliation(s)
- Tatsuya Yuikawa
- Division of Life Science, Graduate School of Science and Engineering, Saitama University, Saitama City, Japan
| | - Masaaki Ikeda
- Division of Life Science, Graduate School of Science and Engineering, Saitama University, Saitama City, Japan
| | - Sachiko Tsuda
- Division of Life Science, Graduate School of Science and Engineering, Saitama University, Saitama City, Japan
| | - Shinji Saito
- Division of Life Science, Graduate School of Science and Engineering, Saitama University, Saitama City, Japan
| | - Kyo Yamasu
- Division of Life Science, Graduate School of Science and Engineering, Saitama University, Saitama City, Japan
| |
Collapse
|
4
|
Raj B, Farrell JA, Liu J, El Kholtei J, Carte AN, Navajas Acedo J, Du LY, McKenna A, Relić Đ, Leslie JM, Schier AF. Emergence of Neuronal Diversity during Vertebrate Brain Development. Neuron 2020; 108:1058-1074.e6. [PMID: 33068532 PMCID: PMC8286448 DOI: 10.1016/j.neuron.2020.09.023] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 08/05/2020] [Accepted: 09/17/2020] [Indexed: 01/03/2023]
Abstract
Neurogenesis comprises many highly regulated processes including proliferation, differentiation, and maturation. However, the transcriptional landscapes underlying brain development are poorly characterized. We describe a developmental single-cell catalog of ∼220,000 zebrafish brain cells encompassing 12 stages from embryo to larva. We characterize known and novel gene markers for ∼800 clusters and provide an overview of the diversification of neurons and progenitors across these time points. We also introduce an optimized GESTALT lineage recorder that enables higher expression and recovery of Cas9-edited barcodes to query lineage segregation. Cell type characterization indicates that most embryonic neural progenitor states are transitory and transcriptionally distinct from neural progenitors of post-embryonic stages. Reconstruction of cell specification trajectories reveals that late-stage retinal neural progenitors transcriptionally overlap cell states observed in the embryo. The zebrafish brain development atlas provides a resource to define and manipulate specific subsets of neurons and to uncover the molecular mechanisms underlying vertebrate neurogenesis.
Collapse
Affiliation(s)
- Bushra Raj
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA 02138, USA; Allen Discovery Center for Cell Lineage Tracing, Seattle, WA, USA.
| | - Jeffrey A Farrell
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA 02138, USA; Unit on Cell Specification and Differentiation, National Institute of Child Health and Human Development, NIH, Bethesda, MD 20814, USA
| | - Jialin Liu
- Allen Discovery Center for Cell Lineage Tracing, Seattle, WA, USA; Biozentrum, University of Basel, 4056 Basel, Switzerland
| | - Jakob El Kholtei
- Allen Discovery Center for Cell Lineage Tracing, Seattle, WA, USA; Biozentrum, University of Basel, 4056 Basel, Switzerland
| | - Adam N Carte
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA 02138, USA; Biozentrum, University of Basel, 4056 Basel, Switzerland; Systems, Synthetic, and Quantitative Biology Program, Harvard University, Cambridge, MA 02138, USA
| | - Joaquin Navajas Acedo
- Allen Discovery Center for Cell Lineage Tracing, Seattle, WA, USA; Biozentrum, University of Basel, 4056 Basel, Switzerland
| | - Lucia Y Du
- Allen Discovery Center for Cell Lineage Tracing, Seattle, WA, USA; Biozentrum, University of Basel, 4056 Basel, Switzerland
| | - Aaron McKenna
- Department of Molecular and Systems Biology, Dartmouth Geisel School of Medicine, Lebanon, NH 03756, USA
| | - Đorđe Relić
- Biozentrum, University of Basel, 4056 Basel, Switzerland; Swiss Institute of Bioinformatics (SIB), 4056 Basel, Switzerland
| | - Jessica M Leslie
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA 02138, USA
| | - Alexander F Schier
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA 02138, USA; Allen Discovery Center for Cell Lineage Tracing, Seattle, WA, USA; Biozentrum, University of Basel, 4056 Basel, Switzerland; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Harvard Stem Cell Institute, Harvard University, Cambridge, MA 02138, USA; Center for Brain Science, Harvard University, Cambridge, MA 02138, USA.
| |
Collapse
|
5
|
Expression patterns of activating transcription factor 5 (atf5a and atf5b) in zebrafish. Gene Expr Patterns 2020; 37:119126. [PMID: 32663618 DOI: 10.1016/j.gep.2020.119126] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Revised: 07/07/2020] [Accepted: 07/07/2020] [Indexed: 11/20/2022]
Abstract
The Activating Transcription Factor 5 (ATF5) is a basic leucine-zipper (bZIP) transcription factor (TF) with proposed stress-protective, anti-apoptotic and oncogenic roles which were all established in cell systems. In whole animals, Atf5 function seems highly context dependent. Atf5 is strongly expressed in the rodent nose and mice knockout (KO) pups have defective olfactory sensory neurons (OSNs), smaller olfactory bulbs (OB), while adults are smell deficient. It was therefore proposed that Atf5 plays an important role in maturation and maintenance of OSNs. Atf5 expression was also described in murine liver and bones where it appears to promote differentiation of progenitor cells. By contrast in the rodent brain, Atf5 was first described as uniquely expressed in neuroprogenitors and thus, proposed to drive their proliferation and inhibit their differentiation. However, it was later also found in mature neurons stressing the need for additional work in whole animals. ATF5 is well conserved with two paralogs, atf5a and atf5b in zebrafish. Here, we present the expression patterns for both from 6 h (hpf) to 5day post-fertilization (dpf). We found early expression for both genes, and from 1dpf onwards overlapping expression patterns in the inner ear and the developing liver. In the brain, at 24hpf both atf5a and atf5b were expressed in the forebrain, midbrain, and hindbrain. However, from 2dpf and onwards we only detected atf5a expression namely in the olfactory bulbs, the mesencephalon, and the metencephalon. We further evidenced additional differential expression for atf5a in the sensory neurons of the olfactory organs, and for atf5b in the neuromasts, that form the superficial sensory organ called the lateral line (LL). Our results establish the basis for future functional analyses in this lower vertebrate.
Collapse
|
6
|
Pushchina EV, Zharikova EI, Varaksin AA, Prudnikov IM, Tsyvkin VN. Proliferation, Adult Neuronal Stem Cells and Cells Migration in Pallium during Constitutive Neurogenesis and after Traumatic Injury of Telencephalon of Juvenile Masu Salmon, Oncorhynchus masou. Brain Sci 2020; 10:brainsci10040222. [PMID: 32276413 PMCID: PMC7226367 DOI: 10.3390/brainsci10040222] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 03/31/2020] [Accepted: 04/06/2020] [Indexed: 12/11/2022] Open
Abstract
A study of the lateral pallium in zebrafish and the visual tectum of the medaka revealed a population of adult neuroepithelial (NE) cells supported from the early stage of development to various postembryonic stages of ontogenesis. These data emphasize the importance of non-radial glial stem cells in the neurogenesis of adult animals, in particular fish. However, the distribution, cell cycle features, and molecular markers of NE cells and glial progenitors in fish are still poorly understood at the postembryonic stages of ontogenesis. Fetalization predominates in the ontogenetic development of salmon fish, which is associated with a delay in development and preservation of the features of the embryonic structure of the brain during the first year of life. In the present work, we studied the features of proliferation and the migration of neuronal precursors in the pallial proliferative zone of juvenile Oncorhynchus masou. The aim of the study is a comparative analysis of the distribution of glial-type aNSCs markers, such as vimentin and glial fibrillar acid protein GFAP, as well as the proliferation marker BrdU and migratory neuronal precursor doublecortin, in the pallial zone of the intact telencephalon in juvenile O. masou normal and after mechanical injury. The immunohistochemical IHC labeling with antibodies to vimentin, GFAP and doublecortin in the pallium of intact fish revealed single, small, round and oval immunopositive cells, that correspond to a persistent pool of neuronal and/or glial progenitors. After the injury, heterogeneous cell clusters, radial glia processes, single and small intensely labeled GFAP+ cells in the parenchyma of Dd and lateral part of pallium (Dl) appeared, corresponding to reactive neurogenic niches containing glial aNSCs. A multifold increase in the pool of Vim+ neuronal precursor cells (NPCs) resulting from the injury was observed. Vim+ cells of the neuroepithelial type in Dd and Dm and cells of the glial type were identified in Dl after the injury. Doublecortine (Dc) immunolabeling after the injury revealed the radial migration of neuroblasts into Dm from the neurogenic zone of the pallium. The appearance of intensely labeled Dc+ cells in the brain parenchyma might indicate the activation of resident aNSCs as a consequence of the traumatic process.
Collapse
Affiliation(s)
- Evgeniya V. Pushchina
- Zhirmunsky National Scientific Center of Marine Biology, Far East Branch, Russian Academy of Sciences, 690041 Vladivostok, Russia; (E.I.Z.); (A.A.V.)
- Bogomoletz Institute of Physiology, National Academy of Sciences of Ukraine, 01024 Kyiv, Ukraine; (I.M.P.); (V.N.T.)
- Correspondence: ; Tel.: +79-149680177
| | - Eva I. Zharikova
- Zhirmunsky National Scientific Center of Marine Biology, Far East Branch, Russian Academy of Sciences, 690041 Vladivostok, Russia; (E.I.Z.); (A.A.V.)
| | - Anatoly A. Varaksin
- Zhirmunsky National Scientific Center of Marine Biology, Far East Branch, Russian Academy of Sciences, 690041 Vladivostok, Russia; (E.I.Z.); (A.A.V.)
| | - Igor M. Prudnikov
- Bogomoletz Institute of Physiology, National Academy of Sciences of Ukraine, 01024 Kyiv, Ukraine; (I.M.P.); (V.N.T.)
| | - Vladimir N. Tsyvkin
- Bogomoletz Institute of Physiology, National Academy of Sciences of Ukraine, 01024 Kyiv, Ukraine; (I.M.P.); (V.N.T.)
| |
Collapse
|
7
|
Inomata C, Yuikawa T, Nakayama-Sadakiyo Y, Kobayashi K, Ikeda M, Chiba M, Konishi C, Ishioka A, Tsuda S, Yamasu K. Involvement of an Oct4-related PouV gene, pou5f3/pou2, in neurogenesis in the early neural plate of zebrafish embryos. Dev Biol 2020; 457:30-42. [DOI: 10.1016/j.ydbio.2019.09.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Revised: 09/09/2019] [Accepted: 09/10/2019] [Indexed: 01/03/2023]
|
8
|
Comprehensive analysis of target genes in zebrafish embryos reveals gbx2 involvement in neurogenesis. Dev Biol 2017; 430:237-248. [DOI: 10.1016/j.ydbio.2017.07.015] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2017] [Revised: 07/17/2017] [Accepted: 07/24/2017] [Indexed: 11/21/2022]
|
9
|
Dambroise E, Simion M, Bourquard T, Bouffard S, Rizzi B, Jaszczyszyn Y, Bourge M, Affaticati P, Heuzé A, Jouralet J, Edouard J, Brown S, Thermes C, Poupon A, Reiter E, Sohm F, Bourrat F, Joly JS. Postembryonic Fish Brain Proliferation Zones Exhibit Neuroepithelial-Type Gene Expression Profile. Stem Cells 2017; 35:1505-1518. [PMID: 28181357 DOI: 10.1002/stem.2588] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2016] [Revised: 12/19/2016] [Accepted: 12/20/2016] [Indexed: 01/04/2023]
Abstract
In mammals, neuroepithelial cells play an essential role in embryonic neurogenesis, whereas glial stem cells are the principal source of neurons at postembryonic stages. By contrast, neuroepithelial-like stem/progenitor (NE) cells have been shown to be present throughout life in teleosts. We used three-dimensional (3D) reconstructions of cleared transgenic wdr12:GFP medaka brains to demonstrate that this cell type is widespread in juvenile and to identify new regions containing NE cells. We established the gene expression profile of optic tectum (OT) NE cells by cell sorting followed by RNA-seq. Our results demonstrate that most OT NE cells are indeed active stem cells and that some of them exhibit long G2 phases. We identified several novel pathways (e.g., DNA repair pathways) potentially involved in NE cell homeostasis. In situ hybridization studies showed that all NE populations in the postembryonic medaka brain have a similar molecular signature. Our findings highlight the importance of NE progenitors in medaka and improve our understanding of NE-cell biology. These cells are potentially useful not only for neural stem cell studies but also for improving the characterization of neurodevelopmental diseases, such as microcephaly. Stem Cells 2017;35:1505-1518.
Collapse
Affiliation(s)
- Emilie Dambroise
- INRA CASBAH Group, Neuro-PSI, UMR 9197, CNRS, Gif-sur-Yvette, France
| | - Matthieu Simion
- INRA CASBAH Group, Neuro-PSI, UMR 9197, CNRS, Gif-sur-Yvette, France
| | | | | | - Barbara Rizzi
- Tefor Core Facility, TEFOR Infrastructure, Neuro-PSI, CNRS, Gif-sur-Yvette, France
| | | | | | - Pierre Affaticati
- Tefor Core Facility, TEFOR Infrastructure, Neuro-PSI, CNRS, Gif-sur-Yvette, France
| | - Aurélie Heuzé
- INRA CASBAH Group, Neuro-PSI, UMR 9197, CNRS, Gif-sur-Yvette, France
| | - Julia Jouralet
- Plateforme BM-Gif, Imagif, UMR 9198, CNRS, Gif-sur-Yvette, France
| | - Joanne Edouard
- UMS AMAGEN CNRS, INRA, Université Paris-Saclay, Gif-sur-Yvette, France
| | | | | | | | | | - Frédéric Sohm
- UMS AMAGEN CNRS, INRA, Université Paris-Saclay, Gif-sur-Yvette, France
| | - Franck Bourrat
- INRA CASBAH Group, Neuro-PSI, UMR 9197, CNRS, Gif-sur-Yvette, France
| | | |
Collapse
|
10
|
Galant S, Furlan G, Coolen M, Dirian L, Foucher I, Bally-Cuif L. Embryonic origin and lineage hierarchies of the neural progenitor subtypes building the zebrafish adult midbrain. Dev Biol 2016; 420:120-135. [PMID: 27693369 PMCID: PMC5156517 DOI: 10.1016/j.ydbio.2016.09.022] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2016] [Revised: 08/31/2016] [Accepted: 09/26/2016] [Indexed: 01/11/2023]
Abstract
Neurogenesis in the post-embryonic vertebrate brain varies in extent and efficiency between species and brain territories. Distinct neurogenesis modes may account for this diversity, and several neural progenitor subtypes, radial glial cells (RG) and neuroepithelial progenitors (NE), have been identified in the adult zebrafish brain. The neurogenic sequences issued from these progenitors, and their contribution to brain construction, remain incompletely understood. Here we use genetic tracing techniques based on conditional Cre recombination and Tet-On neuronal birthdating to unravel the neurogenic sequence operating from NE progenitors in the zebrafish post-embryonic optic tectum. We reveal that a subpopulation of her5-positive NE cells of the posterior midbrain layer stands at the top of a neurogenic hierarchy involving, in order, the amplification pool of the tectal proliferation zone (TPZ), followed by her4-positive RG cells with transient neurogenic activity. We further demonstrate that the adult her5-positive NE pool is issued in lineage from an identically located NE pool expressing the same gene in the embryonic neural tube. Finally, we show that these features are reminiscent of the neurogenic sequence and embryonic origin of the her9-positive progenitor NE pool involved in the construction of the lateral pallium at post-embryonic stages. Together, our results highlight the shared recruitment of an identical neurogenic strategy by two remote brain territories, where long-lasting NE pools serve both as a growth zone and as the life-long source of young neurogenic RG cells. Zebrafish post-embryonic tectal neurogenesis is driven by neuroepithelial progenitors. The neuroepithelial progenitor pool is long-lasting and expresses Her5 life long. Tectal radial glia originate from the her5-positive pool and are transiently neurogenic. The post-embryonic neurogenic sequences of the tectum and lateral pallium are similar.
Collapse
Affiliation(s)
- Sonya Galant
- Paris-Saclay Institute for Neuroscience, CNRS UMR9197 - Université Paris-Sud, Team Zebrafish Neurogenetics, Avenue de la Terrasse, Bldg 5, F-91198 Gif-sur-Yvette, France
| | - Giacomo Furlan
- Paris-Saclay Institute for Neuroscience, CNRS UMR9197 - Université Paris-Sud, Team Zebrafish Neurogenetics, Avenue de la Terrasse, Bldg 5, F-91198 Gif-sur-Yvette, France
| | - Marion Coolen
- Paris-Saclay Institute for Neuroscience, CNRS UMR9197 - Université Paris-Sud, Team Zebrafish Neurogenetics, Avenue de la Terrasse, Bldg 5, F-91198 Gif-sur-Yvette, France; Department of Developmental and Stem Cell Biology and CNRS UMR 3738, Institut Pasteur, 25 rue du Dr Roux, 75015 Paris, France
| | - Lara Dirian
- Paris-Saclay Institute for Neuroscience, CNRS UMR9197 - Université Paris-Sud, Team Zebrafish Neurogenetics, Avenue de la Terrasse, Bldg 5, F-91198 Gif-sur-Yvette, France
| | - Isabelle Foucher
- Paris-Saclay Institute for Neuroscience, CNRS UMR9197 - Université Paris-Sud, Team Zebrafish Neurogenetics, Avenue de la Terrasse, Bldg 5, F-91198 Gif-sur-Yvette, France; Department of Developmental and Stem Cell Biology and CNRS UMR 3738, Institut Pasteur, 25 rue du Dr Roux, 75015 Paris, France.
| | - Laure Bally-Cuif
- Paris-Saclay Institute for Neuroscience, CNRS UMR9197 - Université Paris-Sud, Team Zebrafish Neurogenetics, Avenue de la Terrasse, Bldg 5, F-91198 Gif-sur-Yvette, France; Department of Developmental and Stem Cell Biology and CNRS UMR 3738, Institut Pasteur, 25 rue du Dr Roux, 75015 Paris, France.
| |
Collapse
|
11
|
Peretz Y, Eren N, Kohl A, Hen G, Yaniv K, Weisinger K, Cinnamon Y, Sela-Donenfeld D. A new role of hindbrain boundaries as pools of neural stem/progenitor cells regulated by Sox2. BMC Biol 2016; 14:57. [PMID: 27392568 PMCID: PMC4938926 DOI: 10.1186/s12915-016-0277-y] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2016] [Accepted: 06/21/2016] [Indexed: 01/28/2023] Open
Abstract
Background Compartment boundaries are an essential developmental mechanism throughout evolution, designated to act as organizing centers and to regulate and localize differently fated cells. The hindbrain serves as a fascinating example for this phenomenon as its early development is devoted to the formation of repetitive rhombomeres and their well-defined boundaries in all vertebrates. Yet, the actual role of hindbrain boundaries remains unresolved, especially in amniotes. Results Here, we report that hindbrain boundaries in the chick embryo consist of a subset of cells expressing the key neural stem cell (NSC) gene Sox2. These cells co-express other neural progenitor markers such as Transitin (the avian Nestin), GFAP, Pax6 and chondroitin sulfate proteoglycan. The majority of the Sox2+ cells that reside within the boundary core are slow-dividing, whereas nearer to and within rhombomeres Sox2+ cells are largely proliferating. In vivo analyses and cell tracing experiments revealed the contribution of boundary Sox2+ cells to neurons in a ventricular-to-mantle manner within the boundaries, as well as their lateral contribution to proliferating Sox2+ cells in rhombomeres. The generation of boundary-derived neurospheres from hindbrain cultures confirmed the typical NSC behavior of boundary cells as a multipotent and self-renewing Sox2+ cell population. Inhibition of Sox2 in boundaries led to enhanced and aberrant neural differentiation together with inhibition in cell-proliferation, whereas Sox2 mis-expression attenuated neurogenesis, confirming its significant function in hindbrain neuronal organization. Conclusions Data obtained in this study deciphers a novel role of hindbrain boundaries as repetitive pools of neural stem/progenitor cells, which provide proliferating progenitors and differentiating neurons in a Sox2-dependent regulation. Electronic supplementary material The online version of this article (doi:10.1186/s12915-016-0277-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Yuval Peretz
- Koret School of Veterinary Medicine, The Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot, 76100, Israel
| | - Noa Eren
- Koret School of Veterinary Medicine, The Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot, 76100, Israel
| | - Ayelet Kohl
- Koret School of Veterinary Medicine, The Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot, 76100, Israel
| | - Gideon Hen
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel
| | - Karina Yaniv
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel
| | - Karen Weisinger
- Department of Stem Cell and Regenerative Biology, Harvard Stem Cell Institute, Harvard University, Cambridge, MA, USA
| | - Yuval Cinnamon
- Institute of Animal Sciences, Department of Poultry and Aquaculture Sciences, Agricultural Research Organization, The Volcani Center, Bet Dagan, Israel
| | - Dalit Sela-Donenfeld
- Koret School of Veterinary Medicine, The Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot, 76100, Israel.
| |
Collapse
|
12
|
Abstract
The foundation of the diverse metazoan nervous systems is laid by embryonic patterning mechanisms, involving the generation and movement of neural progenitors and their progeny. Here we divide early neurogenesis into discrete elements, including origin, pattern, proliferation, and movement of neuronal progenitors, which are controlled by conserved gene cassettes. We review these neurogenetic mechanisms in representatives of the different metazoan clades, with the goal to build a conceptual framework in which one can ask specific questions, such as which of these mechanisms potentially formed part of the developmental "toolkit" of the bilaterian ancestor and which evolved later.
Collapse
Affiliation(s)
- Volker Hartenstein
- Department of Molecular Cell and Developmental Biology, University of California, Los Angeles, Los Angeles, CA 90095, USA.
| | - Angelika Stollewerk
- School of Biological and Chemical Sciences, Queen Mary University of London, Mile End Road, London E1 4NS, UK.
| |
Collapse
|
13
|
Zhang Z, Gao F, Kang X, Li J, Zhang L, Dong W, Jin Z, Li F, Gao N, Cai X, Yang S, Zhang J, Ren X, Yang X. Exploring the potential relationship between Notch pathway genes expression and their promoter methylation in mice hippocampal neurogenesis. Brain Res Bull 2015; 113:8-16. [PMID: 25701255 DOI: 10.1016/j.brainresbull.2015.02.003] [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: 12/13/2014] [Revised: 02/08/2015] [Accepted: 02/10/2015] [Indexed: 12/11/2022]
Abstract
The Notch pathway is a highly conserved pathway that regulates hippocampal neurogenesis during embryonic development and adulthood. It has become apparent that intracellular epigenetic modification including DNA methylation is deeply involved in fate specification of neural stem cells (NSCs). However, it is still unclear whether the Notch pathway regulates hippocampal neurogenesis by changing the Notch genes' DNA methylation status. Here, we present the evidence from DNA methylation profiling of Notch1, Hes1 and Ngn2 promoters during neurogenesis in the dentate gyrus (DG) of postnatal, adult and traumatic brains. We observed the expression of Notch1, Hes1 and Ngn2 in hippocampal DG with qPCR, Western blot and immunofluorescence staining. In addition, we investigated the methylation status of Notch pathway genes using the bisulfite sequencing PCR (BSP) method. The number of Notch1 or Hes1 (+) and BrdU (+) cells decreased in the subgranular zone (SGZ) of the DG in the hippocampus following TBI. Nevertheless, the number of Ngn2-positive cells in the DG of injured mice was markedly higher than in the DG of non-TBI mice. Accordingly, the DNA methylation level of the three gene promoters changed with their expression in the DG. These findings suggest that the strict spatio-temporal expression of Notch effector genes plays an important role during hippocampal neurogenesis and suggests the possibility that Notch1, Hes1 and Ngn2 were regulated by changing some specific CpG sites of their promoters to further orchestrate neurogenesis in vivo.
Collapse
Affiliation(s)
- Zhen Zhang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin 300052, PR China; Tianjin Neurological Institute, Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, 154 Anshan Road, Heping District, Tianjin 300052, PR China
| | - Feng Gao
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin 300052, PR China; Tianjin Neurological Institute, Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, 154 Anshan Road, Heping District, Tianjin 300052, PR China
| | - Xiaokui Kang
- Department of Neurosurgery, Liaocheng People's Hospital, 67 Dongchang West Road, Liaocheng 252000, PR China
| | - Jia Li
- Department of Neurosurgery, Baoding No. 1 Hospital, Baoding 071000, PR China
| | - Litong Zhang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin 300052, PR China; Tianjin Neurological Institute, Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, 154 Anshan Road, Heping District, Tianjin 300052, PR China
| | - Wentao Dong
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin 300052, PR China; Tianjin Neurological Institute, Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, 154 Anshan Road, Heping District, Tianjin 300052, PR China
| | - Zhangning Jin
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin 300052, PR China; Tianjin Neurological Institute, Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, 154 Anshan Road, Heping District, Tianjin 300052, PR China
| | - Fan Li
- Department of Neurosurgery, Heji Hospital affiliated Changzhi Medical College, 271 Taihang East Road, Changzhi 046000, PR China
| | - Nannan Gao
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin 300052, PR China; Tianjin Neurological Institute, Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, 154 Anshan Road, Heping District, Tianjin 300052, PR China
| | - Xinwang Cai
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin 300052, PR China; Tianjin Neurological Institute, Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, 154 Anshan Road, Heping District, Tianjin 300052, PR China
| | - Shuyuan Yang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin 300052, PR China; Tianjin Neurological Institute, Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, 154 Anshan Road, Heping District, Tianjin 300052, PR China
| | - Jianning Zhang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin 300052, PR China; Tianjin Neurological Institute, Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, 154 Anshan Road, Heping District, Tianjin 300052, PR China
| | - Xinliang Ren
- Department of Neurosurgery, Heji Hospital affiliated Changzhi Medical College, 271 Taihang East Road, Changzhi 046000, PR China
| | - Xinyu Yang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin 300052, PR China; Tianjin Neurological Institute, Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, 154 Anshan Road, Heping District, Tianjin 300052, PR China.
| |
Collapse
|
14
|
Whittington N, Cunningham D, Le TK, De Maria D, Silva EM. Sox21 regulates the progression of neuronal differentiation in a dose-dependent manner. Dev Biol 2015; 397:237-47. [PMID: 25448693 PMCID: PMC4325979 DOI: 10.1016/j.ydbio.2014.11.012] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2014] [Accepted: 11/12/2014] [Indexed: 12/27/2022]
Abstract
Members of the SoxB transcription factor family play critical roles in the regulation of neurogenesis. The SoxB1 proteins are required for the induction and maintenance of a proliferating neural progenitor population in numerous vertebrates, however the role of the SoxB2 protein, Sox21, is less clear due to conflicting results. To clarify the role of Sox21 in neurogenesis, we examined its function in the Xenopus neural plate. Here we report that misexpression of Sox21 expands the neural progenitor domain, and represses neuron formation by binding to Neurogenin (Ngn2) and blocking its function. Conversely, we found that Sox21 is also required for neuron formation, as cells lacking Sox21 undergo cell death and thus are unable to differentiate. Together our data indicate that Sox21 plays more than one role in neurogenesis, where a threshold level is required for cell viability and normal differentiation of neurons, but a higher concentration of Sox21 inhibits neuron formation and instead promotes progenitor maintenance.
Collapse
Affiliation(s)
- Niteace Whittington
- Department of Biology, Georgetown University, 37th and O Streets NW, Regents Hall 408, Washington, DC 20057, USA.
| | - Doreen Cunningham
- Department of Biology, Georgetown University, 37th and O Streets NW, Regents Hall 408, Washington, DC 20057, USA.
| | - Thien-Kim Le
- Department of Biology, Georgetown University, 37th and O Streets NW, Regents Hall 408, Washington, DC 20057, USA.
| | - David De Maria
- Department of Biology, Georgetown University, 37th and O Streets NW, Regents Hall 408, Washington, DC 20057, USA.
| | - Elena M Silva
- Department of Biology, Georgetown University, 37th and O Streets NW, Regents Hall 408, Washington, DC 20057, USA.
| |
Collapse
|
15
|
Dirian L, Galant S, Coolen M, Chen W, Bedu S, Houart C, Bally-Cuif L, Foucher I. Spatial regionalization and heterochrony in the formation of adult pallial neural stem cells. Dev Cell 2014; 30:123-36. [PMID: 25017692 DOI: 10.1016/j.devcel.2014.05.012] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2013] [Revised: 03/25/2014] [Accepted: 05/14/2014] [Indexed: 01/10/2023]
Abstract
Little is known on the embryonic origin and related heterogeneity of adult neural stem cells (aNSCs). We use conditional genetic tracing, activated in a global or mosaic fashion by cell type-specific promoters or focal laser uncaging, coupled with gene expression analyses and Notch invalidations, to address this issue in the zebrafish adult telencephalon. We report that the germinal zone of the adult pallium originates from two distinct subtypes of embryonic progenitors and integrates two modes of aNSC formation. Dorsomedial aNSCs derive from the amplification of actively neurogenic radial glia of the embryonic telencephalon. On the contrary, the lateral aNSC population is formed by stepwise addition at the pallial edge from a discrete neuroepithelial progenitor pool of the posterior telencephalic roof, activated at postembryonic stages and persisting lifelong. This dual origin of the pallial germinal zone allows the temporally organized building of pallial territories as a patchwork of juxtaposed compartments.
Collapse
Affiliation(s)
- Lara Dirian
- Institute of Neurobiology A. Fessard, Laboratory of Neurobiology and Development, CNRS UPR3294, Team Zebrafish Neurogenetics, Avenue de la Terrasse, Building 5, 91198 Gif-sur-Yvette, France
| | - Sonya Galant
- Institute of Neurobiology A. Fessard, Laboratory of Neurobiology and Development, CNRS UPR3294, Team Zebrafish Neurogenetics, Avenue de la Terrasse, Building 5, 91198 Gif-sur-Yvette, France
| | - Marion Coolen
- Institute of Neurobiology A. Fessard, Laboratory of Neurobiology and Development, CNRS UPR3294, Team Zebrafish Neurogenetics, Avenue de la Terrasse, Building 5, 91198 Gif-sur-Yvette, France
| | - Wenbiao Chen
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, 2213 Garland Ave, Nashville, TN 37232, USA
| | - Sébastien Bedu
- Institute of Neurobiology A. Fessard, Laboratory of Neurobiology and Development, CNRS UPR3294, Team Zebrafish Neurogenetics, Avenue de la Terrasse, Building 5, 91198 Gif-sur-Yvette, France
| | - Corinne Houart
- Medical Research Council Centre for Developmental Neurobiology, King's College London, London SE1 1UL, UK
| | - Laure Bally-Cuif
- Institute of Neurobiology A. Fessard, Laboratory of Neurobiology and Development, CNRS UPR3294, Team Zebrafish Neurogenetics, Avenue de la Terrasse, Building 5, 91198 Gif-sur-Yvette, France.
| | - Isabelle Foucher
- Institute of Neurobiology A. Fessard, Laboratory of Neurobiology and Development, CNRS UPR3294, Team Zebrafish Neurogenetics, Avenue de la Terrasse, Building 5, 91198 Gif-sur-Yvette, France.
| |
Collapse
|
16
|
Coolen M, Katz S, Bally-Cuif L. miR-9: a versatile regulator of neurogenesis. Front Cell Neurosci 2013; 7:220. [PMID: 24312010 PMCID: PMC3834235 DOI: 10.3389/fncel.2013.00220] [Citation(s) in RCA: 213] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2013] [Accepted: 10/31/2013] [Indexed: 12/21/2022] Open
Abstract
Soon after its discovery, microRNA-9 (miR-9) attracted the attention of neurobiologists, since it is one of the most highly expressed microRNAs in the developing and adult vertebrate brain. Functional analyses in different vertebrate species have revealed a prominent role of this microRNA in balancing proliferation in embryonic neural progenitor populations. Key transcriptional regulators such as FoxG1, Hes1 or Tlx, were identified as direct targets of miR-9, placing it at the core of the gene network controlling the progenitor state. Recent data also suggest that this function could extend to adult neural stem cells. Other studies point to a role of miR-9 in differentiated neurons. Moreover miR-9 has been implicated in human brain pathologies, either displaying a protective role, such as in Progeria, or participating in disease progression in brain cancers. Altogether functional studies highlight a prominent feature of this highly conserved microRNA, its functional versatility, both along its evolutionary history and across cellular contexts.
Collapse
Affiliation(s)
- Marion Coolen
- Zebrafish Neurogenetics Team, Laboratory of Neurobiology and Development, Institute of Neurobiology Alfred Fessard, CNRS Gif-sur-Yvette, France
| | | | | |
Collapse
|
17
|
El Yakoubi W, Borday C, Hamdache J, Parain K, Tran HT, Vleminckx K, Perron M, Locker M. Hes4 controls proliferative properties of neural stem cells during retinal ontogenesis. Stem Cells 2013; 30:2784-95. [PMID: 22969013 PMCID: PMC3549485 DOI: 10.1002/stem.1231] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2012] [Accepted: 08/08/2012] [Indexed: 11/22/2022]
Abstract
The retina of fish and amphibian contains genuine neural stem cells located at the most peripheral edge of the ciliary marginal zone (CMZ). However, their cell-of-origin as well as the mechanisms that sustain their maintenance during development are presently unknown. We identified Hes4 (previously named XHairy2), a gene encoding a bHLH-O transcriptional repressor, as a stem cell-specific marker of the Xenopus CMZ that is positively regulated by the canonical Wnt pathway and negatively by Hedgehog signaling. We found that during retinogenesis, Hes4 labels a small territory, located first at the pigmented epithelium (RPE)/neural retina (NR) border and later in the retinal margin, that likely gives rise to adult retinal stem cells. We next addressed whether Hes4 might impart this cell subpopulation with retinal stem cell features: inhibited RPE or NR differentiation programs, continuous proliferation, and slow cell cycle speed. We could indeed show that Hes4 overexpression cell autonomously prevents retinal precursor cells from commitment toward retinal fates and maintains them in a proliferative state. Besides, our data highlight for the first time that Hes4 may also constitute a crucial regulator of cell cycle kinetics. Hes4 gain of function indeed significantly slows down cell division, mainly through the lengthening of G1 phase. As a whole, we propose that Hes4 maintains particular stemness features in a cellular cohort dedicated to constitute the adult retinal stem cell pool, by keeping it in an undifferentiated and slowly proliferative state along embryonic retinogenesis. Stem Cells 2012;30:2784–2795
Collapse
|
18
|
Schmidt R, Strähle U, Scholpp S. Neurogenesis in zebrafish - from embryo to adult. Neural Dev 2013; 8:3. [PMID: 23433260 PMCID: PMC3598338 DOI: 10.1186/1749-8104-8-3] [Citation(s) in RCA: 216] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2012] [Accepted: 01/17/2013] [Indexed: 01/19/2023] Open
Abstract
Neurogenesis in the developing central nervous system consists of the induction and proliferation of neural progenitor cells and their subsequent differentiation into mature neurons. External as well as internal cues orchestrate neurogenesis in a precise temporal and spatial way. In the last 20 years, the zebrafish has proven to be an excellent model organism to study neurogenesis in the embryo. Recently, this vertebrate has also become a model for the investigation of adult neurogenesis and neural regeneration. Here, we summarize the contributions of zebrafish in neural development and adult neurogenesis.
Collapse
Affiliation(s)
- Rebecca Schmidt
- Karlsruhe Institute of Technology, Institute of Toxicology and Genetics, 76021, Karlsruhe, Germany
| | | | | |
Collapse
|
19
|
Parain K, Mazurier N, Bronchain O, Borday C, Cabochette P, Chesneau A, Colozza G, El Yakoubi W, Hamdache J, Locker M, Gilchrist MJ, Pollet N, Perron M. A large scale screen for neural stem cell markers in Xenopus retina. Dev Neurobiol 2012; 72:491-506. [PMID: 22275214 DOI: 10.1002/dneu.20973] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Neural stem cell research suffers from a lack of molecular markers to specifically assess stem or progenitor cell properties. The organization of the Xenopus ciliary marginal zone (CMZ) in the retina allows the spatial distinction of these two cell types: stem cells are confined to the most peripheral region, while progenitors are more central. Despite this clear advantage, very few genes specifically expressed in retinal stem cells have been discovered so far in this model. To gain insight into the molecular signature of these cells, we performed a large-scale expression screen in the Xenopus CMZ, establishing it as a model system for stem cell gene profiling. Eighteen genes expressed specifically in the CMZ stem cell compartment were retrieved and are discussed here. These encode various types of proteins, including factors associated with proliferation, mitotic spindle organization, DNA/RNA processing, and cell adhesion. In addition, the publication of this work in a special issue on Xenopus prompted us to give a more general illustration of the value of large-scale screens in this model species. Thus, beyond neural stem cell specific genes, we give a broader highlight of our screen outcome, describing in particular other retinal cell markers that we found. Finally, we present how these can all be easily retrieved through a novel module we developed in the web-based annotation tool XenMARK, and illustrate the potential of this powerful searchable database in the context of the retina.
Collapse
Affiliation(s)
- Karine Parain
- Neurobiology and Development Laboratory, CNRS UPR 3294, Univ Paris-Sud, Orsay, France
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Kizil C, Dudczig S, Kyritsis N, Machate A, Blaesche J, Kroehne V, Brand M. The chemokine receptor cxcr5 regulates the regenerative neurogenesis response in the adult zebrafish brain. Neural Dev 2012; 7:27. [PMID: 22824261 PMCID: PMC3441421 DOI: 10.1186/1749-8104-7-27] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2012] [Accepted: 06/20/2012] [Indexed: 02/08/2023] Open
Abstract
Background Unlike mammals, zebrafish exhibits extensive neural regeneration after injury in adult stages of its lifetime due to the neurogenic activity of the radial glial cells. However, the genes involved in the regenerative neurogenesis response of the zebrafish brain are largely unknown. Thus, understanding the underlying principles of this regeneration capacity of the zebrafish brain is an interesting research realm that may offer vast clinical ramifications. Results In this paper, we characterized the expression pattern of cxcr5 and analyzed the function of this gene during adult neurogenesis and regeneration of the zebrafish telencephalon. We found that cxcr5 was upregulated transiently in the RGCs and neurons, and the expression in the immune cells such as leukocytes was negligible during both adult neurogenesis and regeneration. We observed that the transgenic misexpression of cxcr5 in the ventricular cells using dominant negative and full-length variants of the gene resulted in altered proliferation and neurogenesis response of the RGCs. When we knocked down cxcr5 using antisense morpholinos and cerebroventricular microinjection, we observed outcomes similar to the overexpression of the dominant negative cxcr5 variant. Conclusions Thus, based on our results, we propose that cxcr5 imposes a proliferative permissiveness to the radial glial cells and is required for differentiation of the RGCs to neurons, highlighting novel roles of cxcr5 in the nervous system of vertebrates. We therefore suggest that cxcr5 is an important cue for ventricular cell proliferation and regenerative neurogenesis in the adult zebrafish telencephalon. Further studies on the role of cxcr5 in mediating neuronal replenishment have the potential to produce clinical ramifications in efforts for regenerative therapeutic applications for human neurological disorders or acute injuries.
Collapse
Affiliation(s)
- Caghan Kizil
- DFG-Center for Regenerative Therapies Dresden - Cluster of Excellence (CRTD), Technische Universität Dresden, Fetscherstr, 105, Dresden, 01307, Germany
| | | | | | | | | | | | | |
Collapse
|
21
|
Chapouton P, Webb KJ, Stigloher C, Alunni A, Adolf B, Hesl B, Topp S, Kremmer E, Bally-Cuif L. Expression of hairy/enhancer of split genes in neural progenitors and neurogenesis domains of the adult zebrafish brain. J Comp Neurol 2012; 519:1748-69. [PMID: 21452233 DOI: 10.1002/cne.22599] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
All subdivisions of the adult zebrafish brain maintain niches of constitutive neurogenesis, sustained by quiescent and multipotent progenitor populations. In the telencephalon, the latter potential neural stem cells take the shape of radial glia aligned along the ventricle and are controlled by Notch signalling. With the aim of identifying new markers of this cell type and of comparing the effectors of embryonic and adult neurogenesis, we focused on the family of hairy/enhancer of split [E(spl)] genes. We report the expression of seven hairy/E(spl) (her) genes and the new helt gene in three neurogenic areas of the adult zebrafish brain (telencephalon, hypothalamus, and midbrain) in relation to radial glia, proliferation, and neurogenesis. We show that the expression of most her genes in the adult brain characterizes quiescent radial glia, whereas only few are expressed in progenitor domains engaged in active proliferation or neurogenesis. The low proliferation status of most her-positive progenitors contrasts with the embryonic nervous system, in which her genes are expressed in actively dividing progenitors. Likewise, we demonstrate largely overlapping expression domains of a set of her genes in the adult brain, which is in striking contrast to their distinct embryonic expression profiles. Overall, our data provide a consolidated map of her expression, quiescent glia, proliferation, and neurogenesis in these various subdivisions of the adult brain and suggest distinct regulation and function of Her factors in the embryonic and adult contexts.
Collapse
Affiliation(s)
- Prisca Chapouton
- Zebrafish Neurogenetics Department, Helmholtz Zentrum München, German Research Center for Environmental Health, D-85764 Neuherberg, Germany.
| | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Kizil C, Kaslin J, Kroehne V, Brand M. Adult neurogenesis and brain regeneration in zebrafish. Dev Neurobiol 2012; 72:429-61. [DOI: 10.1002/dneu.20918] [Citation(s) in RCA: 249] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
23
|
Kim HT, Lee MS, Choi JH, Jung JY, Ahn DG, Yeo SY, Choi DK, Kim CH. The microcephaly gene aspm is involved in brain development in zebrafish. Biochem Biophys Res Commun 2011; 409:640-4. [PMID: 21620798 DOI: 10.1016/j.bbrc.2011.05.056] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2011] [Accepted: 05/10/2011] [Indexed: 10/18/2022]
Abstract
MCPH is a neurodevelopmental disorder characterized by a global reduction in cerebral cortical volume. Homozygous mutation of the MCPH5 gene, also known as ASPM, is the most common cause of the MCPH phenotype. To elucidate the roles of ASPM during embryonic development, the zebrafish aspm was identified, which is specifically expressed in proliferating cells in the CNS. Morpholino-mediated knock-down of aspm resulted in a significant reduction in head size. Furthermore, aspm-deficient embryos exhibited a mitotic arrest during early development. These findings suggest that the reduction in brain size in MCPH might be caused by lack of aspm function in the mitotic cell cycle and demonstrate that the zebrafish can provide a model system for congenital diseases of the human nervous system.
Collapse
Affiliation(s)
- Hyun-Taek Kim
- Department of Biology and GRAST, Chungnam National University, Daejeon 305-764, Republic of Korea
| | | | | | | | | | | | | | | |
Collapse
|
24
|
Webb KJ, Coolen M, Gloeckner CJ, Stigloher C, Bahn B, Topp S, Ueffing M, Bally-Cuif L. The Enhancer of split transcription factor Her8a is a novel dimerisation partner for Her3 that controls anterior hindbrain neurogenesis in zebrafish. BMC DEVELOPMENTAL BIOLOGY 2011; 11:27. [PMID: 21586122 PMCID: PMC3125270 DOI: 10.1186/1471-213x-11-27] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/04/2010] [Accepted: 05/17/2011] [Indexed: 12/31/2022]
Abstract
Background Neurogenesis control and the prevention of premature differentiation in the vertebrate embryo are crucial processes, allowing the formation of late-born cell types and ensuring the correct shape and cytoarchitecture of the brain. Members of the Hairy/Enhancer of Split (Hairy/E(spl)) family of bHLH-Orange transcription factors, such as zebrafish Her3, 5, 9 and 11, are implicated in the local inhibition of neurogenesis to maintain progenitor pools within the early neural plate. To better understand how these factors exert their inhibitory function, we aimed to isolate some of their functional interactors. Results We used a yeast two-hybrid screen with Her5 as bait and recovered a novel zebrafish Hairy/E(spl) factor - Her8a. Using phylogenetic and synteny analyses, we demonstrate that her8a evolved from an ancient duplicate of Hes6 that was recently lost in the mammalian lineage. We show that her8a is expressed across the mid- and anterior hindbrain from the start of segmentation. Through knockdown and misexpression experiments, we demonstrate that Her8a is a negative regulator of neurogenesis and plays an essential role in generating progenitor pools within rhombomeres 2 and 4 - a role resembling that of Her3. Her8a co-purifies with Her3, suggesting that Her8a-Her3 heterodimers may be relevant in this domain of the neural plate, where both proteins are co-expressed. Finally, we demonstrate that her8a expression is independent of Notch signaling at the early neural plate stage but that SoxB factors play a role in its expression, linking patterning information to neurogenesis control. Overall, the regulation and function of Her8a differ strikingly from those of its closest relative in other vertebrates - the Hes6-like proteins. Conclusions Our results characterize the phylogeny, expression and functional interactions involving a new Her factor, Her8a, and highlight the complex interplay of E(spl) proteins that generates the neurogenesis pattern of the zebrafish early neural plate.
Collapse
Affiliation(s)
- Katharine J Webb
- Zebrafish Neurogenetics Department, Helmholtz Zentrum München, German Research Center for Environmental Health, Ingolstädter Landstr, 1, D-85764 Neuherberg, Germany.
| | | | | | | | | | | | | | | |
Collapse
|
25
|
Radosevic M, Robert-Moreno À, Coolen M, Bally-Cuif L, Alsina B. Her9 represses neurogenic fate downstream of Tbx1 and retinoic acid signaling in the inner ear. Development 2011; 138:397-408. [DOI: 10.1242/dev.056093] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Proper spatial control of neurogenesis in the inner ear ensures the precise innervation of mechanotransducing cells and the propagation of auditory and equilibrium stimuli to the brain. Members of the Hairy and enhancer of split (Hes) gene family regulate neurogenesis by inhibiting neuronal differentiation and maintaining neural stem cell pools in non-neurogenic zones. Remarkably, their role in the spatial control of neurogenesis in the ear is unknown. In this study, we identify her9, a zebrafish ortholog of Hes1, as a key gene in regulating otic neurogenesis through the definition of the posterolateral non-neurogenic field. First, her9 emerges as a novel otic patterning gene that represses proneural function and regulates the extent of the neurogenic domain. Second, we place Her9 downstream of Tbx1, linking these two families of transcription factors for the first time in the inner ear and suggesting that the reported role of Tbx1 in repressing neurogenesis is in part mediated by the bHLH transcriptional repressor Her9. Third, we have identified retinoic acid (RA) signaling as the upstream patterning signal of otic posterolateral genes such as tbx1 and her9. Finally, we show that at the level of the cranial otic field, opposing RA and Hedgehog signaling position the boundary between the neurogenic and non-neurogenic compartments. These findings permit modeling of the complex genetic cascade that underlies neural patterning of the otic vesicle.
Collapse
Affiliation(s)
- Marija Radosevic
- Developmental Biology Laboratory, Dept. Ciències Experimentals i de la Salut, Universitat Pompeu Fabra-Parc de Recerca Biomèdica de Barcelona, Dr Aiguader 88, 08003 Barcelona, Spain
| | - Àlex Robert-Moreno
- Developmental Biology Laboratory, Dept. Ciències Experimentals i de la Salut, Universitat Pompeu Fabra-Parc de Recerca Biomèdica de Barcelona, Dr Aiguader 88, 08003 Barcelona, Spain
| | - Marion Coolen
- Laboratory of Neurobiology and Development, Institute of Neurobiology Alfred Fessard, CNRS, Avenue de Terrasse, 91198 cedex, Gif-sur-Yvette, France
| | - Laure Bally-Cuif
- Laboratory of Neurobiology and Development, Institute of Neurobiology Alfred Fessard, CNRS, Avenue de Terrasse, 91198 cedex, Gif-sur-Yvette, France
| | - Berta Alsina
- Developmental Biology Laboratory, Dept. Ciències Experimentals i de la Salut, Universitat Pompeu Fabra-Parc de Recerca Biomèdica de Barcelona, Dr Aiguader 88, 08003 Barcelona, Spain
| |
Collapse
|
26
|
Abstract
Compared with other vertebrate animal models, zebrafish (Danio rerio) has its superior advantages for studying stem cell migration. Zebrafish have similar tissues and organs as mammals, where tissue-specific stem cells reside in. Zebrafish eggs are externally fertilized and remain transparent until most of the organs are fully developed. This allows imaging stem cells in vivo very easily. Recently, a zebrafish double pigmentation mutant, casper, became a new popular imaging model in the zebrafish field due to its completely transparent bodies in adulthood. It has been used as an excellent model to study adult hematopoietic stem cell (HSC) in the transplantation setting. The unparalleled imaging power of zebrafish provides great opportunities of tracing stem cells in vivo in the developmental and regenerative context. In this chapter, we use HSC as an example and combine the powerful imaging techniques in zebrafish, to provide protocols for in vivo imaging fluorescence-labeled stem cell migration, stem cell fate tracing in zebrafish embryos, HSC transplantation, and in vivo imaging in both zebrafish embryos and adults. These techniques can also be applied to other types of stem cells in zebrafish embryos and adults.
Collapse
|
27
|
Tallafuss A, Trepman A, Eisen JS. DeltaA mRNA and protein distribution in the zebrafish nervous system. Dev Dyn 2010; 238:3226-36. [PMID: 19924821 DOI: 10.1002/dvdy.22136] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Abstract
Physical interaction between the transmembrane proteins Delta and Notch allows only a subset of neural precursors to become neurons, as well as regulating other aspects of neural development. To examine the localization of Delta protein during neural development, we generated an antibody specific to zebrafish Delta A (Dla). Here, we describe for the first time the subcellular localization of Dla protein in distinct puncta at cell cortex and/or membrane, supporting the function of Dla in direct cell-cell communication. In situ RNA hybridization and immunohistochemistry revealed dynamic, coordinated expression patterns of dla mRNA and Dla protein in the developing and adult zebrafish nervous system. Dla expression is mostly excluded from differentiated neurons and is maintained in putative precursor cells at least until larval stages. In the adult brain, dla mRNA and Dla protein are expressed in proliferative zones normally associated with stem cells.
Collapse
|
28
|
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
|
29
|
Coolen M, Bally-Cuif L. [A microRNA as a gatekeeper of the midbrain-hindbrain boundary]. Med Sci (Paris) 2009; 24:787-9. [PMID: 18950566 DOI: 10.1051/medsci/20082410787] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
|
30
|
Ninkovic J, Stigloher C, Lillesaar C, Bally-Cuif L. Gsk3beta/PKA and Gli1 regulate the maintenance of neural progenitors at the midbrain-hindbrain boundary in concert with E(Spl) factor activity. Development 2008; 135:3137-48. [PMID: 18725518 DOI: 10.1242/dev.020479] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Neuronal production in the midbrain-hindbrain domain (MH) of the vertebrate embryonic neural tube depends on a progenitor pool called the ;intervening zone' (IZ), located at the midbrain-hindbrain boundary. The progressive recruitment of IZ progenitors along the mediolateral (future dorsoventral) axis prefigures the earlier maturation of the MH basal plate. It also correlates with a lower sensitivity of medial versus lateral IZ progenitors to the neurogenesis inhibition process that maintains the IZ pool. This role is performed in zebrafish by the E(Spl) factors Her5 and Her11, but the molecular cascades cooperating with Her5/11, and those accounting for their reduced effect in the medial IZ, remain unknown. We demonstrate here that the kinases Gsk3beta and cAMP-dependent protein kinase A (PKA) are novel determinants of IZ formation and cooperate with E(Spl) activity in a dose-dependent manner. Similar to E(Spl), we show that the activity of Gsk3beta/PKA is sensed differently by medial versus lateral IZ progenitors. Furthermore, we identify the transcription factor Gli1, expressed in medial IZ cells, as an antagonist of E(Spl) and Gsk3beta/PKA, and demonstrate that the neurogenesis-promoting activity of Gli1 accounts for the reduced sensitivity of medial IZ progenitors to neurogenesis inhibitors and their increased propensity to differentiate. We also show that the expression and activity of Gli1 in this process are, surprisingly, independent of Hedgehog signaling. Together, our results suggest a model in which the modulation of E(Spl) and Gsk3beta/PKA activities by Gli1 underlies the dynamic properties of IZ maintenance and recruitment.
Collapse
Affiliation(s)
- Jovica Ninkovic
- Helmholtz Zentrum Muenchen, German Research Center for Environmental Health, Department of Zebrafish Neurogenetics, Institute of Developmental Genetics, Ingolstaedter Landstrasse 1, D-85764 Neuherberg, Germany
| | | | | | | |
Collapse
|
31
|
Leucht C, Stigloher C, Wizenmann A, Klafke R, Folchert A, Bally-Cuif L. MicroRNA-9 directs late organizer activity of the midbrain-hindbrain boundary. Nat Neurosci 2008; 11:641-8. [PMID: 18454145 DOI: 10.1038/nn.2115] [Citation(s) in RCA: 240] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2008] [Accepted: 03/24/2008] [Indexed: 12/22/2022]
Abstract
The midbrain-hindbrain boundary (MHB) is a long-lasting organizing center in the vertebrate neural tube that is both necessary and sufficient for the ordered development of midbrain and anterior hindbrain (midbrain-hindbrain domain, MH). The MHB also coincides with a pool of progenitor cells that contributes neurons to the entire MH. Here we show that the organizing activity and progenitor state of the MHB are co-regulated by a single microRNA, miR-9, during late embryonic development in zebrafish. Endogenous miR-9 expression, initiated at late stages, selectively spares the MHB. Gain- and loss-of-function studies, in silico predictions and sensor assays in vivo demonstrate that miR-9 targets several components of the Fgf signaling pathway, thereby delimiting the organizing activity of the MHB. In addition, miR-9 promotes progression of neurogenesis in the MH, defining the MHB progenitor pool. Together, these findings highlight a previously unknown mechanism by which a single microRNA fine-tunes late MHB coherence via its co-regulation of patterning activities and neurogenesis.
Collapse
Affiliation(s)
- Christoph Leucht
- Department of Zebrafish Neurogenetics, Institute of Developmental Genetics, Helmholtz-Zentrum München, German Research Center for Environmental Health, Ingolstädter Landstrasse 1, D-85764 Neuherberg, Germany
| | | | | | | | | | | |
Collapse
|