1
|
Vassalli QA, Fasano G, Nittoli V, Gagliardi E, Sepe RM, Donizetti A, Aniello F, Sordino P, Kelsh R, Locascio A. The Zebrafish Retina and the Evolution of the Onecut-Mediated Pathway in Cell Type Differentiation. Cells 2024; 13:2071. [PMID: 39768162 PMCID: PMC11675081 DOI: 10.3390/cells13242071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2024] [Revised: 12/06/2024] [Accepted: 12/09/2024] [Indexed: 01/11/2025] Open
Abstract
Onecut/Hnf6 (Oc) genes play an important role in the proper formation of retinal cells in vertebrates, in particular horizontal, retinal ganglion and amacrine cells. However, it is not fully known how the unique and combined action of multiple Oc gene copies leads to the induction and differentiation of specific retinal cell types. To gain new insights on how Oc genes influence retina formation, we have examined the developmental role of oc1, oc2 and oc-like genes during eye formation in the non-mammalian vertebrate zebrafish Danio rerio. By using single and multiple morpholino knockdown of three zebrafish Oc genes we provide evidence for the independent and redundant role of each gene in the formation of photoreceptors and other retinal tissues. Through comparison of Oc genetic pathways in photoreceptor differentiation among chordates we demonstrate their mechanism of action through a series of conserved target genes involved in neural transmission.
Collapse
Affiliation(s)
- Quirino Attilio Vassalli
- Department of Biology and Evolution of Marine Organisms, Stazione Zoologica Anton Dohrn, 80121 Napoli, Italy; (Q.A.V.); (G.F.); (V.N.); (E.G.); (R.M.S.)
| | - Giulia Fasano
- Department of Biology and Evolution of Marine Organisms, Stazione Zoologica Anton Dohrn, 80121 Napoli, Italy; (Q.A.V.); (G.F.); (V.N.); (E.G.); (R.M.S.)
| | - Valeria Nittoli
- Department of Biology and Evolution of Marine Organisms, Stazione Zoologica Anton Dohrn, 80121 Napoli, Italy; (Q.A.V.); (G.F.); (V.N.); (E.G.); (R.M.S.)
| | - Eleonora Gagliardi
- Department of Biology and Evolution of Marine Organisms, Stazione Zoologica Anton Dohrn, 80121 Napoli, Italy; (Q.A.V.); (G.F.); (V.N.); (E.G.); (R.M.S.)
| | - Rosa Maria Sepe
- Department of Biology and Evolution of Marine Organisms, Stazione Zoologica Anton Dohrn, 80121 Napoli, Italy; (Q.A.V.); (G.F.); (V.N.); (E.G.); (R.M.S.)
| | - Aldo Donizetti
- Department of Biology, University of Naples Federico II, 80126 Naples, Italy; (A.D.); (F.A.)
| | - Francesco Aniello
- Department of Biology, University of Naples Federico II, 80126 Naples, Italy; (A.D.); (F.A.)
| | - Paolo Sordino
- Department of Biology and Evolution of Marine Organisms, Sicily Marine Centre, Stazione Zoologica Anton Dohrn, 98167 Messina, Italy;
| | - Robert Kelsh
- Department of Life Sciences, University of Bath, Bath BA2 7AY, UK;
| | - Annamaria Locascio
- Department of Biology and Evolution of Marine Organisms, Stazione Zoologica Anton Dohrn, 80121 Napoli, Italy; (Q.A.V.); (G.F.); (V.N.); (E.G.); (R.M.S.)
| |
Collapse
|
2
|
Zheng N, Liao T, Zhang C, Zhang Z, Yan S, Xi X, Ruan F, Yang C, Zhao Q, Deng W, Huang J, Huang Z, Chen Z, Wang X, Qu Q, Zuo Z, He C. Quantum Dots-caused Retinal Degeneration in Zebrafish Regulated by Ferroptosis and Mitophagy in Retinal Pigment Epithelial Cells through Inhibiting Spliceosome. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2406343. [PMID: 39420512 PMCID: PMC11633537 DOI: 10.1002/advs.202406343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/08/2024] [Revised: 08/23/2024] [Indexed: 10/19/2024]
Abstract
Quantum dots (QDs) are widely used, but their health impact on the visual system is little known. This study aims to elucidate the effects and mechanisms of typical metallic QDs on retinas using zebrafish. Comprehensive histology, imaging, and bulk RNA sequencing reveal that InP/ZnS QDs cause retinal degeneration. Furthermore, single-cell RNA-seq reveals a reduction in the number of retinal pigment epithelial cells (RPE) and short-wave cone UV photoreceptor cells (PR(UV)), accompanied by an increase in middle- and long-wave cone red, green, and blue photoreceptor cells [PR(RGB)]. Mechanistically, after endocytosis by RPE, InP/ZnS QDs inhibit the expression of splicing factor prpf8, resulting in gpx4b mRNA unsplicing, which finally decrease glutathione and induce ferroptosis and mitophagy. The decrease of RPE fails to engulf the damaged outer segments of PR, possibly promoting the differentiation of PR(UV) to PR(RGB). Knockout prpf8 or gpx4b with CRISPR/Cas9 system, the retinal damage is also observed. Whereas, overexpression of prpf8 or gpx4b, or supplement of glutathione can rescue the retinal degenerative damage caused by InP/ZnS QDs. In conclusion, this study illustrates the potential health risks of InP/ZnS QDs on eye development and provides valuable insights into the underlying mechanisms of InP/ZnS QDs-caused retinal degeneration.
Collapse
Affiliation(s)
- Naying Zheng
- Department of Ophthalmology in Xiang'an Hospital of Xiamen UniversityState Key Laboratory of Cellular Stress BiologySchool of Life SciencesFaculty of Medicine and Life SciencesXiamen UniversityXiamenFujian361102China
| | - Tingting Liao
- Department of Ophthalmology in Xiang'an Hospital of Xiamen UniversityState Key Laboratory of Cellular Stress BiologySchool of Life SciencesFaculty of Medicine and Life SciencesXiamen UniversityXiamenFujian361102China
| | - Chuchu Zhang
- Department of Ophthalmology in Xiang'an Hospital of Xiamen UniversityState Key Laboratory of Cellular Stress BiologySchool of Life SciencesFaculty of Medicine and Life SciencesXiamen UniversityXiamenFujian361102China
| | - Zheyang Zhang
- Department of Ophthalmology in Xiang'an Hospital of Xiamen UniversityState Key Laboratory of Cellular Stress BiologySchool of Life SciencesFaculty of Medicine and Life SciencesXiamen UniversityXiamenFujian361102China
| | - Sen Yan
- Department of ChemistryState Key Laboratory of Physical Chemistry of Solid SurfacesCollaborative Innovation Center of Chemistry for Energy Materials (i‐ChEM)Innovation Laboratory for Sciences and Technologies of Energy Materials of Fujian Province (IKKEM)College of Chemistry and Chemical EngineeringXiamen UniversityXiamen361005China
| | - Xiaohan Xi
- Department of ChemistryState Key Laboratory of Physical Chemistry of Solid SurfacesCollaborative Innovation Center of Chemistry for Energy Materials (i‐ChEM)Innovation Laboratory for Sciences and Technologies of Energy Materials of Fujian Province (IKKEM)College of Chemistry and Chemical EngineeringXiamen UniversityXiamen361005China
| | - Fengkai Ruan
- Department of Ophthalmology in Xiang'an Hospital of Xiamen UniversityState Key Laboratory of Cellular Stress BiologySchool of Life SciencesFaculty of Medicine and Life SciencesXiamen UniversityXiamenFujian361102China
| | - Chunyan Yang
- Department of Ophthalmology in Xiang'an Hospital of Xiamen UniversityState Key Laboratory of Cellular Stress BiologySchool of Life SciencesFaculty of Medicine and Life SciencesXiamen UniversityXiamenFujian361102China
| | - Qingliang Zhao
- State Key Laboratory of Vaccines for Infectious DiseasesCenter for Molecular Imaging and Translational MedicineXiang An Biomedicine LaboratorySchool of Public HealthXiamen UniversityXiamenFujian361005China
| | - Wenbo Deng
- Key Laboratory of Reproductive Health ResearchFujian Province UniversitySchool of MedicineXiamen UniversityXiamenFujian361005China
| | - Jialiang Huang
- Department of Ophthalmology in Xiang'an Hospital of Xiamen UniversityState Key Laboratory of Cellular Stress BiologySchool of Life SciencesFaculty of Medicine and Life SciencesXiamen UniversityXiamenFujian361102China
| | - Zi‐Tao Huang
- Guangdong Key Laboratory of Environmental Catalysis and Health Risk ControlGuangdong‐Hong Kong‐Macao Joint Laboratory for Contaminants Exposure and HealthSchool of Environmental Science and EngineeringGuangdong University of TechnologyGuangzhou510006China
| | - Zhi‐Feng Chen
- Guangdong Key Laboratory of Environmental Catalysis and Health Risk ControlGuangdong‐Hong Kong‐Macao Joint Laboratory for Contaminants Exposure and HealthSchool of Environmental Science and EngineeringGuangdong University of TechnologyGuangzhou510006China
| | - Xiang Wang
- Department of ChemistryState Key Laboratory of Physical Chemistry of Solid SurfacesCollaborative Innovation Center of Chemistry for Energy Materials (i‐ChEM)Innovation Laboratory for Sciences and Technologies of Energy Materials of Fujian Province (IKKEM)College of Chemistry and Chemical EngineeringXiamen UniversityXiamen361005China
| | - Qingming Qu
- Department of Ophthalmology in Xiang'an Hospital of Xiamen UniversityState Key Laboratory of Cellular Stress BiologySchool of Life SciencesFaculty of Medicine and Life SciencesXiamen UniversityXiamenFujian361102China
| | - Zhenghong Zuo
- Department of Ophthalmology in Xiang'an Hospital of Xiamen UniversityState Key Laboratory of Cellular Stress BiologySchool of Life SciencesFaculty of Medicine and Life SciencesXiamen UniversityXiamenFujian361102China
| | - Chengyong He
- Department of Ophthalmology in Xiang'an Hospital of Xiamen UniversityState Key Laboratory of Cellular Stress BiologySchool of Life SciencesFaculty of Medicine and Life SciencesXiamen UniversityXiamenFujian361102China
| |
Collapse
|
3
|
Dumas CM, St. Clair RM, Lasseigne AM, Ballif BA, Ebert AM. The intracellular domain of Sema6A is essential for development of the zebrafish retina. J Cell Sci 2024; 137:jcs261469. [PMID: 38963001 PMCID: PMC11795297 DOI: 10.1242/jcs.261469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Accepted: 06/24/2024] [Indexed: 07/05/2024] Open
Abstract
Semaphorin6A (Sema6A) is a repulsive guidance molecule that plays many roles in central nervous system, heart and bone development, as well as immune system responses and cell signaling in cancer. Loss of Sema6A or its receptor PlexinA2 in zebrafish leads to smaller eyes and improper retinal patterning. Here, we investigate a potential role for the Sema6A intracellular domain in zebrafish eye development and dissect which phenotypes rely on forward signaling and which rely on reverse signaling. We performed rescue experiments on zebrafish Sema6A morphants with either full-length Sema6A (Sema6A-FL) or Sema6A lacking its intracellular domain (Sema6A-ΔC). We identified that the intracellular domain is not required for eye size and retinal patterning, however it is required for retinal integrity, the number and end feet strength of Müller glia and protecting against retinal cell death. This novel function for the intracellular domain suggests a role for Sema6A reverse signaling in zebrafish eye development.
Collapse
Affiliation(s)
- Caroline M. Dumas
- Department of Biology, University of Vermont, Burlington, VT 05405, USA
| | | | | | - Bryan A. Ballif
- Department of Biology, University of Vermont, Burlington, VT 05405, USA
| | - Alicia M. Ebert
- Department of Biology, University of Vermont, Burlington, VT 05405, USA
| |
Collapse
|
4
|
Li Y, Yu S, Jia X, Qiu X, He J. Defining morphologically and genetically distinct GABAergic/cholinergic amacrine cell subtypes in the vertebrate retina. PLoS Biol 2024; 22:e3002506. [PMID: 38363811 PMCID: PMC10914270 DOI: 10.1371/journal.pbio.3002506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2023] [Revised: 03/05/2024] [Accepted: 01/18/2024] [Indexed: 02/18/2024] Open
Abstract
In mammals, retinal direction selectivity originates from GABAergic/cholinergic amacrine cells (ACs) specifically expressing the sox2 gene. However, the cellular diversity of GABAergic/cholinergic ACs of other vertebrate species remains largely unexplored. Here, we identified 2 morphologically and genetically distinct GABAergic/cholinergic AC types in zebrafish, a previously undescribed bhlhe22+ type and a mammalian counterpart sox2+ type. Notably, while sole sox2 disruption removed sox2+ type, the codisruption of bhlhe22 and bhlhe23 was required to remove bhlhe22+ type. Also, both types significantly differed in dendritic arbors, lamination, and soma position. Furthermore, in vivo two-photon calcium imaging and the behavior assay suggested the direction selectivity of both AC types. Nevertheless, the 2 types showed preferential responses to moving bars of different sizes. Thus, our findings provide new cellular diversity and functional characteristics of GABAergic/cholinergic ACs in the vertebrate retina.
Collapse
Affiliation(s)
- Yan Li
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Shuguang Yu
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Xinling Jia
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Xiaoying Qiu
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Jie He
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| |
Collapse
|
5
|
Nerli E, Kretzschmar J, Bianucci T, Rocha‐Martins M, Zechner C, Norden C. Deterministic and probabilistic fate decisions co-exist in a single retinal lineage. EMBO J 2023; 42:e112657. [PMID: 37184124 PMCID: PMC10350840 DOI: 10.15252/embj.2022112657] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 04/04/2023] [Accepted: 04/20/2023] [Indexed: 05/16/2023] Open
Abstract
Correct nervous system development depends on the timely differentiation of progenitor cells into neurons. While the output of progenitor differentiation is well investigated at the population and clonal level, how stereotypic or variable fate decisions are during development is still more elusive. To fill this gap, we here follow the fate outcome of single neurogenic progenitors in the zebrafish retina over time using live imaging. We find that neurogenic progenitor divisions produce two daughter cells, one of deterministic and one of probabilistic fate. Interference with the deterministic branch of the lineage affects lineage progression. In contrast, interference with fate probabilities of the probabilistic branch results in a broader range of fate possibilities than in wild-type and involves the production of any neuronal cell type even at non-canonical developmental stages. Combining the interference data with stochastic modelling of fate probabilities revealed that a simple gene regulatory network is able to predict the observed fate decision probabilities during wild-type development. These findings unveil unexpected lineage flexibility that could ensure robust development of the retina and other tissues.
Collapse
Affiliation(s)
- Elisa Nerli
- Instituto Gulbenkian de CiênciaOeirasPortugal
- Max Planck Institute of Molecular Cell Biology and GeneticsDresdenGermany
- Center for Systems Biology DresdenDresdenGermany
| | | | - Tommaso Bianucci
- Max Planck Institute of Molecular Cell Biology and GeneticsDresdenGermany
- Center for Systems Biology DresdenDresdenGermany
- Physics of Life, Cluster of ExcellenceTU DresdenDresdenGermany
| | - Mauricio Rocha‐Martins
- Instituto Gulbenkian de CiênciaOeirasPortugal
- Max Planck Institute of Molecular Cell Biology and GeneticsDresdenGermany
| | - Christoph Zechner
- Max Planck Institute of Molecular Cell Biology and GeneticsDresdenGermany
- Center for Systems Biology DresdenDresdenGermany
- Physics of Life, Cluster of ExcellenceTU DresdenDresdenGermany
| | - Caren Norden
- Instituto Gulbenkian de CiênciaOeirasPortugal
- Max Planck Institute of Molecular Cell Biology and GeneticsDresdenGermany
| |
Collapse
|
6
|
Li Y, Xu B, Jin M, Zhang H, Ren N, Hu J, He J. Homophilic interaction of cell adhesion molecule 3 coordinates retina neuroepithelial cell proliferation. J Cell Biol 2023; 222:e202204098. [PMID: 37022761 PMCID: PMC10082328 DOI: 10.1083/jcb.202204098] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 01/07/2023] [Accepted: 03/07/2023] [Indexed: 04/07/2023] Open
Abstract
Correct cell number generation is central to tissue development. However, in vivo roles of coordinated proliferation of individual neural progenitors in regulating cell numbers of developing neural tissues and the underlying molecular mechanism remain mostly elusive. Here, we showed that wild-type (WT) donor retinal progenitor cells (RPCs) generated significantly expanded clones in host retinae with G1-lengthening by p15 (cdkn2a/b) overexpression (p15+) in zebrafish. Further analysis showed that cell adhesion molecule 3 (cadm3) was reduced in p15+ host retinae, and overexpression of either full-length or ectodomains of Cadm3 in p15+ host retinae markedly suppressed the clonal expansion of WT donor RPCs. Notably, WT donor RPCs in retinae with cadm3 disruption recapitulated expanded clones that were found in p15+ retinae. More strikingly, overexpression of Cadm3 without extracellular ig1 domain in RPCs resulted in expanded clones and increased retinal total cell number. Thus, homophilic interaction of Cadm3 provides an intercellular mechanism underlying coordinated cell proliferation to ensure cell number homeostasis of the developing neuroepithelia.
Collapse
Affiliation(s)
- Yanan Li
- State Key Laboratory of Neuroscience, Institute of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Baijie Xu
- State Key Laboratory of Neuroscience, Institute of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Mengmeng Jin
- State Key Laboratory of Neuroscience, Institute of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Hui Zhang
- State Key Laboratory of Neuroscience, Institute of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Ningxin Ren
- State Key Laboratory of Neuroscience, Institute of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Jinhui Hu
- State Key Laboratory of Neuroscience, Institute of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Jie He
- State Key Laboratory of Neuroscience, Institute of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| |
Collapse
|
7
|
Jin M, Zhang H, Xu B, Li Y, Qin H, Yu S, He J. Jag2b-Notch3/1b-mediated neuron-to-glia crosstalk controls retinal gliogenesis. EMBO Rep 2022; 23:e54922. [PMID: 36047082 PMCID: PMC9535768 DOI: 10.15252/embr.202254922] [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: 02/23/2022] [Revised: 08/03/2022] [Accepted: 08/18/2022] [Indexed: 11/09/2022] Open
Abstract
In the developing central nervous systems (CNS), neural progenitor cells generate neurons and glia in sequential order. However, the influence of neurons on glia generation remains elusive. Here, we report that photoreceptor cell-derived Jag2b is required for Notch-dependent Müller glia (MG) generation in the developing zebrafish retina. In jab2b-/- mutants, differentiating MGs are re-specified into lineage-related bipolar neuron fate at the expense of mature MG. Single-cell transcriptome analysis and knock-in animals reveal that jab2b is specifically expressed in crx+ -photoreceptor cells during MG generation. Crx promoter-driven jag2b, but not other Notch ligands, is sufficient to rescue the loss of MGs observed in jag2b-/- mutants. Furthermore, we observe a severe and moderate decrease in the number of MGs in notch3-/- and notch1b-/- mutants, respectively, and the activation of Notch3 or Notch1b rescues the MG loss in jag2b-/- mutants. Together, our findings reveal that the interaction of Jag2b and Notch3/Notch1b mediates the crosstalk between neurons and glial cells to ensure the irreversible differentiation of MG, providing novel mechanistic insights into the temporal specification of glial cell fate in a developing vertebrate CNS structure.
Collapse
Affiliation(s)
- Mengmeng Jin
- State Key Laboratory of NeuroscienceInstitute of NeuroscienceCenter for Excellence in Brain Science and Intelligence TechnologyChinese Academy of SciencesShanghaiChina
- University of Chinese Academy of SciencesBeijingChina
| | - Hui Zhang
- State Key Laboratory of NeuroscienceInstitute of NeuroscienceCenter for Excellence in Brain Science and Intelligence TechnologyChinese Academy of SciencesShanghaiChina
- University of Chinese Academy of SciencesBeijingChina
| | - Baijie Xu
- State Key Laboratory of NeuroscienceInstitute of NeuroscienceCenter for Excellence in Brain Science and Intelligence TechnologyChinese Academy of SciencesShanghaiChina
- University of Chinese Academy of SciencesBeijingChina
| | - Yanan Li
- State Key Laboratory of NeuroscienceInstitute of NeuroscienceCenter for Excellence in Brain Science and Intelligence TechnologyChinese Academy of SciencesShanghaiChina
- University of Chinese Academy of SciencesBeijingChina
| | - Huiwen Qin
- State Key Laboratory of NeuroscienceInstitute of NeuroscienceCenter for Excellence in Brain Science and Intelligence TechnologyChinese Academy of SciencesShanghaiChina
- University of Chinese Academy of SciencesBeijingChina
| | - Shuguang Yu
- State Key Laboratory of NeuroscienceInstitute of NeuroscienceCenter for Excellence in Brain Science and Intelligence TechnologyChinese Academy of SciencesShanghaiChina
- University of Chinese Academy of SciencesBeijingChina
| | - Jie He
- State Key Laboratory of NeuroscienceInstitute of NeuroscienceCenter for Excellence in Brain Science and Intelligence TechnologyChinese Academy of SciencesShanghaiChina
- Shanghai Center for Brain Science and Brain‐Inspired Intelligence TechnologyShanghaiChina
| |
Collapse
|
8
|
Grigoryan EN. Self-Organization of the Retina during Eye Development, Retinal Regeneration In Vivo, and in Retinal 3D Organoids In Vitro. Biomedicines 2022; 10:1458. [PMID: 35740479 PMCID: PMC9221005 DOI: 10.3390/biomedicines10061458] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 06/16/2022] [Accepted: 06/18/2022] [Indexed: 11/23/2022] Open
Abstract
Self-organization is a process that ensures histogenesis of the eye retina. This highly intricate phenomenon is not sufficiently studied due to its biological complexity and genetic heterogeneity. The review aims to summarize the existing central theories and ideas for a better understanding of retinal self-organization, as well as to address various practical problems of retinal biomedicine. The phenomenon of self-organization is discussed in the spatiotemporal context and illustrated by key findings during vertebrate retina development in vivo and retinal regeneration in amphibians in situ. Described also are histotypic 3D structures obtained from the disaggregated retinal progenitor cells of birds and retinal 3D organoids derived from the mouse and human pluripotent stem cells. The review highlights integral parts of retinal development in these conditions. On the cellular level, these include competence, differentiation, proliferation, apoptosis, cooperative movements, and migration. On the physical level, the focus is on the mechanical properties of cell- and cell layer-derived forces and on the molecular level on factors responsible for gene regulation, such as transcription factors, signaling molecules, and epigenetic changes. Finally, the self-organization phenomenon is discussed as a basis for the production of retinal organoids, a promising model for a wide range of basic scientific and medical applications.
Collapse
Affiliation(s)
- Eleonora N Grigoryan
- Koltzov Institute of Developmental Biology, Russian Academy of Sciences, 119334 Moscow, Russia
| |
Collapse
|
9
|
Stergas HR, Kalbag Z, St Clair RM, Talbot JC, Ballif BA, Ebert AM. Crk adaptor proteins are necessary for the development of the zebrafish retina. Dev Dyn 2021; 251:362-376. [PMID: 34268820 DOI: 10.1002/dvdy.402] [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: 02/05/2021] [Revised: 05/18/2021] [Accepted: 07/09/2021] [Indexed: 11/07/2022] Open
Abstract
BACKGROUND The development of the central nervous system (CNS) requires critical cell signaling molecules to coordinate cell proliferation and migration in order to structure the adult tissue. Chicken tumor virus #10 Regulator of Kinase (CRK) and CRK-like (CRKL) are adaptor proteins with pre-metazoan ancestry and are known to be required for patterning laminated structures downstream of Reelin (RELN), such as the cerebral cortex, cerebellum, and hippocampus. CRK and CRKL also play crucial roles in a variety of other growth factor and extracellular matrix signaling cascades. The neuronal retina is another highly laminated structure within the CNS that is dependent on migration for proper development, but the cell signaling mechanisms behind neuronal positioning in the retina are only partly understood. RESULTS We find that crk and crkl have largely overlapping expression within the developing zebrafish nervous system. We find that their disruption results in smaller eye size and loss of retinal lamination. CONCLUSIONS Our data indicate that Crk adaptors are critical for proper development of the zebrafish neural retina in a crk/crkl dose-dependent manner.
Collapse
Affiliation(s)
- Helaina R Stergas
- Department of Biology, The University of Vermont, Burlington, Vermont, USA
| | - Zoë Kalbag
- Department of Biology, The University of Vermont, Burlington, Vermont, USA
| | - Riley M St Clair
- Department of Biology, The University of Vermont, Burlington, Vermont, USA
| | - Jared C Talbot
- School of Biology and Ecology, The University of Maine, Orono, Maine, USA
| | - Bryan A Ballif
- Department of Biology, The University of Vermont, Burlington, Vermont, USA
| | - Alicia M Ebert
- Department of Biology, The University of Vermont, Burlington, Vermont, USA
| |
Collapse
|
10
|
Hedde PN, Cinco R, Malacrida L, Kamaid A, Gratton E. Phasor-based hyperspectral snapshot microscopy allows fast imaging of live, three-dimensional tissues for biomedical applications. Commun Biol 2021; 4:721. [PMID: 34117344 PMCID: PMC8195998 DOI: 10.1038/s42003-021-02266-z] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Accepted: 05/26/2021] [Indexed: 01/31/2023] Open
Abstract
Hyperspectral imaging is highly sought after in many fields including mineralogy and geology, environment and agriculture, astronomy and, importantly, biomedical imaging and biological fluorescence. We developed ultrafast phasor-based hyperspectral snapshot microscopy based on sine/cosine interference filters for biomedical imaging not feasible with conventional hyperspectral detection methods. Current approaches rely on slow spatial or spectral scanning limiting their application in living biological tissues, while faster snapshot methods such as image mapping spectrometry and multispectral interferometry are limited in spatial and/or spectral resolution, are computationally demanding, and imaging devices are very expensive to manufacture. Leveraging light sheet microscopy, phasor-based hyperspectral snapshot microscopy improved imaging speed 10-100 fold which, combined with minimal light exposure and high detection efficiency, enabled hyperspectral metabolic imaging of live, three-dimensional mouse tissues not feasible with other methods. As a fit-free method that does not require any a priori information often unavailable in complex and evolving biological systems, the rule of linear combinations of the phasor could spectrally resolve subtle differences between cell types in the developing zebrafish retina and spectrally separate and track multiple organelles in 3D cultured cells over time. The sine/cosine snapshot method is adaptable to any microscope or imaging device thus making hyperspectral imaging and fit-free analysis based on linear combinations broadly available to researchers and the public.
Collapse
Affiliation(s)
- Per Niklas Hedde
- Laboratory for Fluorescence Dynamics, University of California, Irvine, CA, USA.
- Department of Pharmaceutical Sciences, University of California, Irvine, CA, USA.
- Beckman Laser Institute & Medical Clinic, University of California, Irvine, CA, USA.
| | - Rachel Cinco
- Laboratory for Fluorescence Dynamics, University of California, Irvine, CA, USA
| | - Leonel Malacrida
- Departamento de Fisiopatología, Hospital de Clínicas, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
- Advanced Bioimaging Unit, Institut Pasteur of Montevideo and Universidad de la República, Montevideo, Uruguay
| | - Andrés Kamaid
- Advanced Bioimaging Unit, Institut Pasteur of Montevideo and Universidad de la República, Montevideo, Uruguay
| | - Enrico Gratton
- Laboratory for Fluorescence Dynamics, University of California, Irvine, CA, USA.
- Beckman Laser Institute & Medical Clinic, University of California, Irvine, CA, USA.
| |
Collapse
|
11
|
Lusk S, Casey MA, Kwan KM. 4-Dimensional Imaging of Zebrafish Optic Cup Morphogenesis. J Vis Exp 2021:10.3791/62155. [PMID: 34125104 PMCID: PMC8848516 DOI: 10.3791/62155] [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] [Indexed: 10/31/2022] Open
Abstract
Visual system function requires the establishment of precise tissue and organ structures. In the vertebrate eye, structural defects are a common cause of visual impairment, yet mechanisms of eye morphogenesis are still poorly understood. The basic organization of the embryonic eye is conserved throughout vertebrates, thus live imaging of zebrafish embryos has become a powerful approach to directly observe eye development at real time under normal and pathological conditions. Dynamic cell processes including movements, morphologies, interactions, division, and death can be visualized in the embryo. We have developed methods for uniform labeling of subcellular structures and timelapse confocal microscopy of early eye development in zebrafish. This protocol outlines the method of generating capped mRNA for injection into the 1-cell zebrafish embryo, mounting embryos at optic vesicle stage (~12 hours post fertilization, hpf), and performing multi-dimensional timelapse imaging of optic cup morphogenesis on a laser scanning confocal microscope, such that multiple datasets are acquired sequentially in the same imaging session. Such an approach yields data that can be used for a variety of purposes, including cell tracking, volume measurements, three-dimensional (3D) rendering, and visualization. Our approaches allow us to pinpoint the cellular and molecular mechanisms driving optic cup development, in both wild type and genetic mutant conditions. These methods can be employed directly by other groups or adapted to visualize many additional aspects of zebrafish eye development.
Collapse
Affiliation(s)
- Sarah Lusk
- Department of Human Genetics, University of Utah
| | | | | |
Collapse
|
12
|
Slit2 is necessary for optic axon organization in the zebrafish ventral midline. Cells Dev 2021; 166:203677. [PMID: 33994352 DOI: 10.1016/j.cdev.2021.203677] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 03/25/2021] [Accepted: 03/26/2021] [Indexed: 02/07/2023]
Abstract
Slit-Robo signaling has been implicated in regulating several steps of retinal ganglion cell axon guidance, with a central role assigned to Slit2. We report here the phenotypical characterization of a CRISPR-Cas9-generated zebrafish null mutant for this gene, along with a detailed analysis of its expression pattern by WM-FISH. All evident defects in the optic axons in slit2-/- mutants were detected outside the retina, coincident with the major sites of expression at the ventral forebrain, around the developing optic nerve and anterior to the optic chiasm/proximal tract. Anterograde axon tracing experiments in zygotic and maternal-zygotic mutants, as well as morphants, showed the occurrence of axon sorting defects, which appeared mild at the optic nerve level, but more severe in the optic chiasm and the proximal tract. A remarkable sorting defect was the usual splitting of one of the optic nerves in two branches that surrounded the contralateral nerve at the chiasm. Although all axons eventually crossed the midline, the retinotopic order appeared lost at the proximal optic tract, to eventually correct distally. Time-lapse analysis demonstrated the sporadic occurrence of axon misrouting at the chiasm level, which could be responsible for the sorting errors. Our results support previous evidence of a channeling role for Slit molecules in retinal ganglion cell axons at the optic nerve, in addition to a function in the segregation of axons coming from each nerve and from different retinal regions at the medio-ventral area of the forebrain.
Collapse
|
13
|
Cyclin A2/cyclin-dependent kinase 1-dependent phosphorylation of Top2a is required for S phase entry during retinal development in zebrafish. J Genet Genomics 2021; 48:63-74. [PMID: 33832859 DOI: 10.1016/j.jgg.2021.01.001] [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: 11/11/2020] [Revised: 12/31/2020] [Accepted: 01/05/2021] [Indexed: 12/26/2022]
Abstract
Cyclin-dependent kinase 1 (CDK1) plays an essential role in cell cycle regulation. However, as mouse Cdk1 embryos die early, the role of CDK1 in regulating the cell cycle and embryo development remains unclear. Here, we showed that zebrafish cdk1-/- embryos exhibit severe microphthalmia accompanied by multiple defects in S phase entry, M phase progression, and cell differentiation but not in interkinetic nuclear migration. We identified Top2a as a potential downstream target and cyclin A2 and cyclin B1 as partners of Cdk1 in cell cycle regulation via an in silico analysis. While depletion of either cyclin A2 or Top2a led to the decreased S phase entry in zebrafish retinal cells, the depletion of cyclin B1 led to M phase arrest. Moreover, phosphorylation of Top2a at serine 1213 (S1213) was nearly abolished in both cdk1 and ccna2 mutants, but not in ccnb1 mutants. Furthermore, overexpression of TOP2AS1213D, the phosphomimetic form of human TOP2A, rescued S phase entry and alleviated the microphthalmia defects in both cdk1-/- and ccna2-/- embryos. Taken together, our data suggest that Cdk1 interacts with cyclin A2 to regulate S phase entry partially through Top2a phosphorylation and interacts with cyclin B1 to regulate M phase progression.
Collapse
|
14
|
Lahne M, Brecker M, Jones SE, Hyde DR. The Regenerating Adult Zebrafish Retina Recapitulates Developmental Fate Specification Programs. Front Cell Dev Biol 2021; 8:617923. [PMID: 33598455 PMCID: PMC7882614 DOI: 10.3389/fcell.2020.617923] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Accepted: 12/31/2020] [Indexed: 12/20/2022] Open
Abstract
Adult zebrafish possess the remarkable capacity to regenerate neurons. In the damaged zebrafish retina, Müller glia reprogram and divide to produce neuronal progenitor cells (NPCs) that proliferate and differentiate into both lost neuronal cell types and those unaffected by the damage stimulus, which suggests that developmental specification/differentiation programs might be recapitulated during regeneration. Quantitative real-time polymerase chain reaction revealed that developmental competence factors are expressed following photoreceptor damage induced by intense light or in a genetic rod photoreceptor cell ablation model. In both light- and N-Methyl-D-aspartic acid (NMDA)-damaged adult zebrafish retinas, NPCs, but not proliferating Müller glia, expressed fluorescent reporters controlled by promoters of ganglion (atoh7), amacrine (ptf1a), bipolar (vsx1), or red cone photoreceptor cell competence factors (thrb) in a temporal expression sequence. In both damage paradigms, atoh7:GFP was expressed first, followed by ptf1a:EGFP and lastly, vsx1:GFP, whereas thrb:Tomato was observed in NPCs at the same time as ptf1a:GFP following light damage but shifted alongside vsx1:GFP in the NMDA-damaged retina. Moreover, HuC/D, indicative of ganglion and amacrine cell differentiation, colocalized with atoh7:GFP prior to ptf1a:GFP expression in the ganglion cell layer, which was followed by Zpr-1 expression (red/green cone photoreceptors) in thrb:Tomato-positive cells in the outer nuclear layer in both damage paradigms, mimicking the developmental differentiation sequence. However, comparing NMDA- to light-damaged retinas, the fraction of PCNA-positive cells expressing atoh7:GFP increased, that of thrb:Tomato and vsx1:GFP decreased, and that of ptf1a:GFP remained similar. To summarize, developmental cell specification programs were recapitulated during retinal regeneration, which adapted to account for the cell type lost.
Collapse
Affiliation(s)
- Manuela Lahne
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN, United States.,Center for Stem Cells and Regenerative Medicine, University of Notre Dame, Notre Dame, IN, United States.,Center for Zebrafish Research, University of Notre Dame, Notre Dame, IN, United States
| | - Margaret Brecker
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN, United States.,Center for Stem Cells and Regenerative Medicine, University of Notre Dame, Notre Dame, IN, United States.,Center for Zebrafish Research, University of Notre Dame, Notre Dame, IN, United States
| | - Stuart E Jones
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN, United States
| | - David R Hyde
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN, United States.,Center for Stem Cells and Regenerative Medicine, University of Notre Dame, Notre Dame, IN, United States.,Center for Zebrafish Research, University of Notre Dame, Notre Dame, IN, United States
| |
Collapse
|
15
|
Xu B, Tang X, Jin M, Zhang H, Du L, Yu S, He J. Unifying developmental programs for embryonic and postembryonic neurogenesis in the zebrafish retina. Development 2020; 147:dev.185660. [PMID: 32467236 DOI: 10.1242/dev.185660] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2019] [Accepted: 05/13/2020] [Indexed: 01/14/2023]
Abstract
The zebrafish retina grows for a lifetime. Whether embryonic and postembryonic retinogenesis conform to the same developmental program is an outstanding question that remains under debate. Using single-cell RNA sequencing of ∼20,000 cells of the developing zebrafish retina at four different stages, we identified seven distinct developmental states. Each state explicitly expresses a gene set. Disruption of individual state-specific marker genes results in various defects ranging from small eyes to the loss of distinct retinal cell types. Using a similar approach, we further characterized the developmental states of postembryonic retinal stem cells (RSCs) and their progeny in the ciliary marginal zone. Expression pattern analysis of state-specific marker genes showed that the developmental states of postembryonic RSCs largely recapitulated those of their embryonic counterparts, except for some differences in rod photoreceptor genesis. Thus, our findings reveal the unifying developmental program used by the embryonic and postembryonic retinogenesis in zebrafish.
Collapse
Affiliation(s)
- Baijie Xu
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China.,University of Chinese Academy of Sciences, Beijing 100049, China.,Shanghai Center for Brain Science and Brain-Inspired Intelligence Technology, Shanghai 201210, China
| | - Xia Tang
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China .,Shanghai Center for Brain Science and Brain-Inspired Intelligence Technology, Shanghai 201210, China
| | - Mengmeng Jin
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China.,University of Chinese Academy of Sciences, Beijing 100049, China.,Shanghai Center for Brain Science and Brain-Inspired Intelligence Technology, Shanghai 201210, China
| | - Hui Zhang
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China.,University of Chinese Academy of Sciences, Beijing 100049, China.,Shanghai Center for Brain Science and Brain-Inspired Intelligence Technology, Shanghai 201210, China
| | - Lei Du
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China.,University of Chinese Academy of Sciences, Beijing 100049, China.,Shanghai Center for Brain Science and Brain-Inspired Intelligence Technology, Shanghai 201210, China
| | - Shuguang Yu
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China.,University of Chinese Academy of Sciences, Beijing 100049, China.,Shanghai Center for Brain Science and Brain-Inspired Intelligence Technology, Shanghai 201210, China
| | - Jie He
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China .,Shanghai Center for Brain Science and Brain-Inspired Intelligence Technology, Shanghai 201210, China
| |
Collapse
|
16
|
Hailstone M, Waithe D, Samuels TJ, Yang L, Costello I, Arava Y, Robertson E, Parton RM, Davis I. CytoCensus, mapping cell identity and division in tissues and organs using machine learning. eLife 2020; 9:e51085. [PMID: 32423529 PMCID: PMC7237217 DOI: 10.7554/elife.51085] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Accepted: 03/17/2020] [Indexed: 01/16/2023] Open
Abstract
A major challenge in cell and developmental biology is the automated identification and quantitation of cells in complex multilayered tissues. We developed CytoCensus: an easily deployed implementation of supervised machine learning that extends convenient 2D 'point-and-click' user training to 3D detection of cells in challenging datasets with ill-defined cell boundaries. In tests on such datasets, CytoCensus outperforms other freely available image analysis software in accuracy and speed of cell detection. We used CytoCensus to count stem cells and their progeny, and to quantify individual cell divisions from time-lapse movies of explanted Drosophila larval brains, comparing wild-type and mutant phenotypes. We further illustrate the general utility and future potential of CytoCensus by analysing the 3D organisation of multiple cell classes in Zebrafish retinal organoids and cell distributions in mouse embryos. CytoCensus opens the possibility of straightforward and robust automated analysis of developmental phenotypes in complex tissues.
Collapse
Affiliation(s)
- Martin Hailstone
- Department of Biochemistry, University of OxfordOxfordUnited Kingdom
| | - Dominic Waithe
- Wolfson Imaging Center & MRC WIMM Centre for Computational Biology MRC Weather all Institute of Molecular Medicine University of OxfordOxfordUnited Kingdom
| | - Tamsin J Samuels
- Department of Biochemistry, University of OxfordOxfordUnited Kingdom
| | - Lu Yang
- Department of Biochemistry, University of OxfordOxfordUnited Kingdom
| | - Ita Costello
- The Dunn School of Pathology,University of OxfordOxfordUnited Kingdom
| | - Yoav Arava
- Department of Biology, Technion - Israel Institute of TechnologyHaifaIsrael
| | | | - Richard M Parton
- Department of Biochemistry, University of OxfordOxfordUnited Kingdom
- Micron Advanced Bioimaging Unit, Department of Biochemistry, University of OxfordOxfordUnited Kingdom
| | - Ilan Davis
- Department of Biochemistry, University of OxfordOxfordUnited Kingdom
- Micron Advanced Bioimaging Unit, Department of Biochemistry, University of OxfordOxfordUnited Kingdom
| |
Collapse
|
17
|
Emerson SE, Stergas HR, Bupp-Chickering SO, Ebert AM. Shootin-1 is required for nervous system development in zebrafish. Dev Dyn 2020; 249:1285-1295. [PMID: 32406957 DOI: 10.1002/dvdy.194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Revised: 04/09/2020] [Accepted: 05/05/2020] [Indexed: 11/08/2022] Open
Abstract
BACKGROUND Semaphorin6A (Sema6A) and its PlexinA2 (PlxnA2) receptor canonically function as repulsive axon guidance cues. To understand downstream signaling mechanisms, we performed a microarray screen and identified the "clutch molecule" shootin-1 (shtn-1) as a transcriptionally repressed target. Shtn-1 is a key proponent of cell migration and neuronal polarization and must be regulated during nervous system development. The mechanisms of Shtn-1 regulation and the phenotypic consequences of loss of repression are poorly understood. RESULTS We demonstrate shtn-1 overexpression results in impaired migration of the optic vesicles, lack of retinal pigmented epithelium, and pathfinding errors of retinotectal projections. We also observed patterning defects in the peripheral nervous system. Importantly, these phenotypes were rescued by overexpressing PlxnA2. CONCLUSIONS We demonstrate a functional role for repression of shtn-1 by PlxnA2 in development of the eyes and peripheral nervous system in zebrafish. These results demonstrate that careful regulation of shtn-1 is critical for development of the nervous system.
Collapse
Affiliation(s)
- Sarah E Emerson
- Department of Biology, University of Vermont, Burlington, Vermont, USA
| | - Helaina R Stergas
- Department of Biology, University of Vermont, Burlington, Vermont, USA
| | | | - Alicia M Ebert
- Department of Biology, University of Vermont, Burlington, Vermont, USA
| |
Collapse
|
18
|
Carrillo-Rosas S, Weber C, Fievet L, Messaddeq N, Karam A, Trottier Y. Loss of zebrafish Ataxin-7, a SAGA subunit responsible for SCA7 retinopathy, causes ocular coloboma and malformation of photoreceptors. Hum Mol Genet 2020; 28:912-927. [PMID: 30445451 DOI: 10.1093/hmg/ddy401] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Revised: 10/31/2018] [Accepted: 11/10/2018] [Indexed: 12/26/2022] Open
Abstract
Polyglutamine (polyQ) expansion in Ataxin-7 (ATXN7) results in spinocerebellar ataxia type 7 (SCA7) and causes visual impairment. SCA7 photoreceptors progressively lose their outer segments (OSs), a structure essential for their visual function. ATXN7 is a subunit of the transcriptional coactivator Spt-Ada-Gcn5 Acetyltransferase complex, implicated in the development of the visual system in flies. To determine the function of ATXN7 in the vertebrate eye, we have inactivated ATXN7 in zebrafish. While ATXN7 depletion in flies led to gross retinal degeneration, in zebrafish, it primarily results in ocular coloboma, a structural malformation responsible for pediatric visual impairment in humans. ATXN7 inactivation leads to elevated Hedgehog signaling in the forebrain, causing an alteration of proximo-distal patterning of the optic vesicle during early eye development and coloboma. At later developmental stages, malformations of photoreceptors due to incomplete formation of their OSs are observed and correlate with altered expression of crx, a key transcription factor involved in the formation of photoreceptor OS. Therefore, we propose that a primary toxic effect of polyQ expansion is the alteration of ATXN7 function in the daily renewal of OS in SCA7. Together, our data indicate that ATXN7 plays an essential role in vertebrate eye morphogenesis and photoreceptor differentiation, and its loss of function may contribute to the development of human coloboma.
Collapse
Affiliation(s)
- Samantha Carrillo-Rosas
- Institute of Genetics and Molecular and Cellular Biology (IGBMC).,Centre National de la Recherche Scientifique, UMR7104.,Institut National de la Santé et de la Recherche Médicale, U1254.,University of Strasbourg, Illkirch, 67000, France
| | - Chantal Weber
- Institute of Genetics and Molecular and Cellular Biology (IGBMC).,Centre National de la Recherche Scientifique, UMR7104.,Institut National de la Santé et de la Recherche Médicale, U1254.,University of Strasbourg, Illkirch, 67000, France
| | - Lorraine Fievet
- Institute of Genetics and Molecular and Cellular Biology (IGBMC).,Centre National de la Recherche Scientifique, UMR7104.,Institut National de la Santé et de la Recherche Médicale, U1254.,University of Strasbourg, Illkirch, 67000, France
| | - Nadia Messaddeq
- Institute of Genetics and Molecular and Cellular Biology (IGBMC).,Centre National de la Recherche Scientifique, UMR7104.,Institut National de la Santé et de la Recherche Médicale, U1254.,University of Strasbourg, Illkirch, 67000, France
| | - Alice Karam
- Institute of Genetics and Molecular and Cellular Biology (IGBMC).,Centre National de la Recherche Scientifique, UMR7104.,Institut National de la Santé et de la Recherche Médicale, U1254.,University of Strasbourg, Illkirch, 67000, France
| | - Yvon Trottier
- Institute of Genetics and Molecular and Cellular Biology (IGBMC).,Centre National de la Recherche Scientifique, UMR7104.,Institut National de la Santé et de la Recherche Médicale, U1254.,University of Strasbourg, Illkirch, 67000, France
| |
Collapse
|
19
|
Chouket R, Pellissier-Tanon A, Lemarchand A, Espagne A, Le Saux T, Jullien L. Dynamic contrast with reversibly photoswitchable fluorescent labels for imaging living cells. Chem Sci 2020; 11:2882-2887. [PMID: 34122788 PMCID: PMC8157520 DOI: 10.1039/d0sc00182a] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Accepted: 02/23/2020] [Indexed: 12/20/2022] Open
Abstract
Interrogating living cells requires sensitive imaging of a large number of components in real time. The state-of-the-art of multiplexed imaging is usually limited to a few components. This review reports on the promise and the challenges of dynamic contrast to overcome this limitation.
Collapse
Affiliation(s)
- Raja Chouket
- PASTEUR, Département de Chimie, École Normale Supérieure, PSL University, Sorbonne Université, CNRS 24, rue Lhomond 75005 Paris France +33 4432 3333
| | - Agnès Pellissier-Tanon
- PASTEUR, Département de Chimie, École Normale Supérieure, PSL University, Sorbonne Université, CNRS 24, rue Lhomond 75005 Paris France +33 4432 3333
| | - Annie Lemarchand
- Sorbonne Université, Centre National de la Recherche Scientifique (CNRS), Laboratoire de Physique Théorique de la Matière Condensée (LPTMC) 4 Place Jussieu, Case Courrier 121 75252 Paris Cedex 05 France
| | - Agathe Espagne
- PASTEUR, Département de Chimie, École Normale Supérieure, PSL University, Sorbonne Université, CNRS 24, rue Lhomond 75005 Paris France +33 4432 3333
| | - Thomas Le Saux
- PASTEUR, Département de Chimie, École Normale Supérieure, PSL University, Sorbonne Université, CNRS 24, rue Lhomond 75005 Paris France +33 4432 3333
| | - Ludovic Jullien
- PASTEUR, Département de Chimie, École Normale Supérieure, PSL University, Sorbonne Université, CNRS 24, rue Lhomond 75005 Paris France +33 4432 3333
| |
Collapse
|
20
|
Seritrakul P, Gross JM. Genetic and epigenetic control of retinal development in zebrafish. Curr Opin Neurobiol 2019; 59:120-127. [PMID: 31255843 PMCID: PMC6888853 DOI: 10.1016/j.conb.2019.05.008] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Revised: 04/23/2019] [Accepted: 05/28/2019] [Indexed: 12/24/2022]
Abstract
The vertebrate retina is a complex structure composed of seven cell types (six neuron and one glia), and all of which originate from a seemingly homogeneous population of proliferative multipotent retinal progenitor cells (RPCs) that exit the cell cycle and differentiate in a spatio-temporally regulated and stereotyped fashion. This neurogenesis process requires intricate genetic regulation involving a combination of cell intrinsic transcription factors and extrinsic signaling molecules, and many critical factors have been identified that influence the timing and composition of the developing retina. Adding complexity to the process, over the past decade, a variety of epigenetic regulatory mechanisms have been shown to influence neurogenesis, and these include changes in histone modifications and the chromatin landscape and changes in DNA methylation and hydroxymethylation patterns. This review summarizes recent findings in the genetic and epigenetic regulation of retinal development, with an emphasis on the zebrafish model system, and it outlines future areas of investigation that will continue to push the field forward into the epigenomics era.
Collapse
Affiliation(s)
- Pawat Seritrakul
- Faculty of Animal Sciences and Agricultural Technology, Silpakorn University, Phetchaburi, 76120, Thailand.
| | - Jeffrey M Gross
- Departments of Ophthalmology, and Developmental Biology, The Louis J. Fox Center for Vision Restoration, The McGowan Institute for Regenerative Medicine, The University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, United States.
| |
Collapse
|
21
|
Baden T, Euler T, Berens P. Understanding the retinal basis of vision across species. Nat Rev Neurosci 2019; 21:5-20. [PMID: 31780820 DOI: 10.1038/s41583-019-0242-1] [Citation(s) in RCA: 178] [Impact Index Per Article: 29.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/22/2019] [Indexed: 12/12/2022]
Abstract
The vertebrate retina first evolved some 500 million years ago in ancestral marine chordates. Since then, the eyes of different species have been tuned to best support their unique visuoecological lifestyles. Visual specializations in eye designs, large-scale inhomogeneities across the retinal surface and local circuit motifs mean that all species' retinas are unique. Computational theories, such as the efficient coding hypothesis, have come a long way towards an explanation of the basic features of retinal organization and function; however, they cannot explain the full extent of retinal diversity within and across species. To build a truly general understanding of vertebrate vision and the retina's computational purpose, it is therefore important to more quantitatively relate different species' retinal functions to their specific natural environments and behavioural requirements. Ultimately, the goal of such efforts should be to build up to a more general theory of vision.
Collapse
Affiliation(s)
- Tom Baden
- Sussex Neuroscience, School of Life Sciences, University of Sussex, Brighton, UK. .,Institute for Ophthalmic Research, University of Tübingen, Tübingen, Germany.
| | - Thomas Euler
- Institute for Ophthalmic Research, University of Tübingen, Tübingen, Germany.,Werner Reichardt Centre for Integrative Neuroscience, University of Tübingen, Tübingen, Germany
| | - Philipp Berens
- Institute for Ophthalmic Research, University of Tübingen, Tübingen, Germany.,Werner Reichardt Centre for Integrative Neuroscience, University of Tübingen, Tübingen, Germany.,Institute for Bioinformatics and Medical Informatics, University of Tübingen, Tübingen, Germany.,Bernstein Centre for Computational Neuroscience, University of Tübingen, Tübingen, Germany
| |
Collapse
|
22
|
Abdeladim L, Matho KS, Clavreul S, Mahou P, Sintes JM, Solinas X, Arganda-Carreras I, Turney SG, Lichtman JW, Chessel A, Bemelmans AP, Loulier K, Supatto W, Livet J, Beaurepaire E. Multicolor multiscale brain imaging with chromatic multiphoton serial microscopy. Nat Commun 2019; 10:1662. [PMID: 30971684 PMCID: PMC6458155 DOI: 10.1038/s41467-019-09552-9] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Accepted: 03/12/2019] [Indexed: 11/20/2022] Open
Abstract
Large-scale microscopy approaches are transforming brain imaging, but currently lack efficient multicolor contrast modalities. We introduce chromatic multiphoton serial (ChroMS) microscopy, a method integrating one-shot multicolor multiphoton excitation through wavelength mixing and serial block-face image acquisition. This approach provides organ-scale micrometric imaging of spectrally distinct fluorescent proteins and label-free nonlinear signals with constant micrometer-scale resolution and sub-micron channel registration over the entire imaged volume. We demonstrate tridimensional (3D) multicolor imaging over several cubic millimeters as well as brain-wide serial 2D multichannel imaging. We illustrate the strengths of this method through color-based 3D analysis of astrocyte morphology and contacts in the mouse cerebral cortex, tracing of individual pyramidal neurons within densely Brainbow-labeled tissue, and multiplexed whole-brain mapping of axonal projections labeled with spectrally distinct tracers. ChroMS will be an asset for multiscale and system-level studies in neuroscience and beyond.
Collapse
Affiliation(s)
- Lamiae Abdeladim
- Laboratory for Optics and Biosciences, Ecole polytechnique, CNRS, INSERM, IP Paris, Palaiseau, 91128, France
| | - Katherine S Matho
- Laboratory for Optics and Biosciences, Ecole polytechnique, CNRS, INSERM, IP Paris, Palaiseau, 91128, France
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, 17 rue Moreau, Paris, 75012, France
- Cold Spring Harbor Laboratory, Cold Spring Harbor, 11724, NY, USA
| | - Solène Clavreul
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, 17 rue Moreau, Paris, 75012, France
| | - Pierre Mahou
- Laboratory for Optics and Biosciences, Ecole polytechnique, CNRS, INSERM, IP Paris, Palaiseau, 91128, France
| | - Jean-Marc Sintes
- Laboratory for Optics and Biosciences, Ecole polytechnique, CNRS, INSERM, IP Paris, Palaiseau, 91128, France
| | - Xavier Solinas
- Laboratory for Optics and Biosciences, Ecole polytechnique, CNRS, INSERM, IP Paris, Palaiseau, 91128, France
| | - Ignacio Arganda-Carreras
- Department of Computer Science and Artificial Intelligence, University of the Basque Country, San Sebastian, 20018, Spain
- IKERBASQUE, Basque Foundation for Science, Bilbao, 48013, Spain
- Donostia International Physics Center (DIPC), San Sebastian, 20018, Spain
| | - Stephen G Turney
- Center for Brain Science and Department of Molecular and Cellular Biology, Harvard University, Cambridge, 02138, MA, USA
| | - Jeff W Lichtman
- Center for Brain Science and Department of Molecular and Cellular Biology, Harvard University, Cambridge, 02138, MA, USA
| | - Anatole Chessel
- Laboratory for Optics and Biosciences, Ecole polytechnique, CNRS, INSERM, IP Paris, Palaiseau, 91128, France
| | - Alexis-Pierre Bemelmans
- Neurodegenerative Diseases Laboratory, Molecular Imaging Research Center, Institut de Biologie François Jacob, CEA, CNRS, Université Paris-Sud, Fontenay-aux-Roses, 92265, France
| | - Karine Loulier
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, 17 rue Moreau, Paris, 75012, France
| | - Willy Supatto
- Laboratory for Optics and Biosciences, Ecole polytechnique, CNRS, INSERM, IP Paris, Palaiseau, 91128, France
| | - Jean Livet
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, 17 rue Moreau, Paris, 75012, France.
| | - Emmanuel Beaurepaire
- Laboratory for Optics and Biosciences, Ecole polytechnique, CNRS, INSERM, IP Paris, Palaiseau, 91128, France.
| |
Collapse
|
23
|
Eldred MK, Muresan L, Harris WA. Disaggregation and Reaggregation of Zebrafish Retinal Cells for the Analysis of Neuronal Layering. Methods Mol Biol 2019; 1576:255-271. [PMID: 28710687 DOI: 10.1007/7651_2017_46] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/17/2023]
Abstract
The reaggregation of dissociated cells to form organotypic structures provides an in vitro system for the analysis of the cellular interactions and molecular mechanisms involved in the formation of tissue architecture. The retina, an outgrowth of the forebrain, is a precisely layered neural tissue, yet the mechanisms underlying layer formation are largely unexplored. Here we describe the protocol to dissociate, re-aggregate, and culture zebrafish retinal cells from a transgenic, Spectrum of Fates, line where all main cell types are labelled with a combination of fluorescent proteins driven by fate-specific promoters. These cells re-aggregate and self-organize in just 48 h in minimal culture conditions. We also describe how the patterning in these aggregates can be analyzed using isocontour profiling to compare whether different conditions affect their self-organization.
Collapse
Affiliation(s)
- Megan K Eldred
- Department of Physiology, Development and Neuroscience, Cambridge University, Cambridge, UK.
| | - Leila Muresan
- Department of Physiology, Development and Neuroscience, Cambridge University, Cambridge, UK
| | - William A Harris
- Department of Physiology, Development and Neuroscience, Cambridge University, Cambridge, UK
| |
Collapse
|
24
|
Crespo C, Knust E. Characterisation of maturation of photoreceptor cell subtypes during zebrafish retinal development. Biol Open 2018; 7:bio.036632. [PMID: 30237290 PMCID: PMC6262866 DOI: 10.1242/bio.036632] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Photoreceptor cells (PRCs) mature from simple epithelial cells, a process characterised by growth and compartmentalisation of the apical membrane into an inner and an outer segment. So far, a PRC subtype-specific description of morphological and cellular changes in the developing zebrafish retina is missing. Here, we performed an in-depth characterisation of four of the five PRC subtypes of the zebrafish retina between 51 and 120 h post fertilisation, including quantification of the size of different compartments, localisation of polarity proteins and positioning of organelles. One of the major findings was the anisotropic and subtype-specific growth of the different PRC compartments. In addition, a transient accumulation of endoplasmic reticulum in rod PRCs, changes in chromatin organisation in UV sensitive cones and differential expression of polarity proteins during the initial stages of PRC maturation were observed. The results obtained provide a developmental timeline that can be used as a platform for future studies on PRC maturation and function. This platform was applied to document that increased exposure to light leads to smaller apical domains of PRCs. Summary: We characterised subtype-specific growth of the different photoreceptor compartments, organelle distribution and the influence of light on the growth of the apical membrane.
Collapse
Affiliation(s)
- Cátia Crespo
- Max-Planck-Institute of Molecular Cell Biology and Genetics, Pfotenhauerstrasse 108, 01307, Dresden, Germany
| | - Elisabeth Knust
- Max-Planck-Institute of Molecular Cell Biology and Genetics, Pfotenhauerstrasse 108, 01307, Dresden, Germany
| |
Collapse
|
25
|
A non-cell-autonomous actin redistribution enables isotropic retinal growth. PLoS Biol 2018. [PMID: 30096143 DOI: 10.1371/journal.pbio.2006018.] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Tissue shape is often established early in development and needs to be scaled isotropically during growth. However, the cellular contributors and ways by which cells interact tissue-wide to enable coordinated isotropic tissue scaling are not yet understood. Here, we follow cell and tissue shape changes in the zebrafish retinal neuroepithelium, which forms a cup with a smooth surface early in development and maintains this architecture as it grows. By combining 3D analysis and theory, we show how a global increase in cell height can maintain tissue shape during growth. Timely cell height increase occurs concurrently with a non-cell-autonomous actin redistribution. Blocking actin redistribution and cell height increase perturbs isotropic scaling and leads to disturbed, folded tissue shape. Taken together, our data show how global changes in cell shape enable isotropic growth of the developing retinal neuroepithelium, a concept that could also apply to other systems.
Collapse
|
26
|
Matejčić M, Salbreux G, Norden C. A non-cell-autonomous actin redistribution enables isotropic retinal growth. PLoS Biol 2018; 16:e2006018. [PMID: 30096143 PMCID: PMC6117063 DOI: 10.1371/journal.pbio.2006018] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Revised: 08/30/2018] [Accepted: 07/31/2018] [Indexed: 11/26/2022] Open
Abstract
Tissue shape is often established early in development and needs to be scaled isotropically during growth. However, the cellular contributors and ways by which cells interact tissue-wide to enable coordinated isotropic tissue scaling are not yet understood. Here, we follow cell and tissue shape changes in the zebrafish retinal neuroepithelium, which forms a cup with a smooth surface early in development and maintains this architecture as it grows. By combining 3D analysis and theory, we show how a global increase in cell height can maintain tissue shape during growth. Timely cell height increase occurs concurrently with a non-cell-autonomous actin redistribution. Blocking actin redistribution and cell height increase perturbs isotropic scaling and leads to disturbed, folded tissue shape. Taken together, our data show how global changes in cell shape enable isotropic growth of the developing retinal neuroepithelium, a concept that could also apply to other systems.
Collapse
Affiliation(s)
- Marija Matejčić
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | | | - Caren Norden
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| |
Collapse
|
27
|
Amini R, Rocha-Martins M, Norden C. Neuronal Migration and Lamination in the Vertebrate Retina. Front Neurosci 2018; 11:742. [PMID: 29375289 PMCID: PMC5767219 DOI: 10.3389/fnins.2017.00742] [Citation(s) in RCA: 71] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2017] [Accepted: 12/20/2017] [Indexed: 01/04/2023] Open
Abstract
In the retina, like in most other brain regions, developing neurons are arranged into distinct layers giving the mature tissue its stratified appearance. This process needs to be highly controlled and orchestrated, as neuronal layering defects lead to impaired retinal function. To achieve successful neuronal layering and lamination in the retina and beyond, three main developmental steps need to be executed: First, the correct type of neuron has to be generated at a precise developmental time. Second, as most retinal neurons are born away from the position at which they later function, newborn neurons have to move to their final layer within the developing tissue, a process also termed neuronal lamination. Third, these neurons need to connect to their correct synaptic partners. Here, we discuss neuronal migration and lamination in the vertebrate retina and summarize our knowledge on these aspects of retinal development. We give an overview of how lamination emerges and discuss the different modes of neuronal translocation that occur during retinogenesis and what we know about the cell biological machineries driving them. In addition, retinal mosaics and their importance for correct retinal function are examined. We close by stating the open questions and future directions in this exciting field.
Collapse
Affiliation(s)
- Rana Amini
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | | | - Caren Norden
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| |
Collapse
|
28
|
Abstract
The zebrafish (Danio rerio) possesses a vertebrate-type retina that is extraordinarily conserved in evolution. This well-organized and anatomically easily accessible part of the central nervous system has been widely investigated in zebrafish, promoting general understanding of retinal development, morphology, function and associated diseases. Over the recent years, genome and protein engineering as well as imaging techniques have experienced revolutionary advances and innovations, creating new possibilities and methods to study zebrafish development and function. In this review, we focus on some of these emerging technologies and how they may impact retinal research in the future. We place an emphasis on genetic techniques, such as transgenic approaches and the revolutionizing new possibilities in genome editing.
Collapse
Affiliation(s)
- Stephanie Niklaus
- a Institute of Molecular Life Sciences , University of Zurich , Zurich , Switzerland.,b Life Science Zurich Graduate Program - Neuroscience , Zurich , Switzerland
| | - Stephan C F Neuhauss
- a Institute of Molecular Life Sciences , University of Zurich , Zurich , Switzerland
| |
Collapse
|
29
|
Icha J, Kunath C, Rocha-Martins M, Norden C. Independent modes of ganglion cell translocation ensure correct lamination of the zebrafish retina. J Cell Biol 2017; 215:259-275. [PMID: 27810916 PMCID: PMC5084647 DOI: 10.1083/jcb.201604095] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2016] [Accepted: 09/19/2016] [Indexed: 11/22/2022] Open
Abstract
Icha et al. show that retinal ganglion cells (RGCs) can move by two different modes across the embryonic zebrafish retina and that correct RGC translocation is crucial for neuronal lamination and retinal development. The arrangement of neurons into distinct layers is critical for neuronal connectivity and function. During development, most neurons move from their birthplace to the appropriate layer, where they polarize. However, kinetics and modes of many neuronal translocation events still await exploration. In this study, we investigate retinal ganglion cell (RGC) translocation across the embryonic zebrafish retina. After completing their translocation, RGCs establish the most basal retinal layer where they form the optic nerve. Using in toto light sheet microscopy, we show that somal translocation of RGCs is a fast and directed event. It depends on basal process attachment and stabilized microtubules. Interestingly, interference with somal translocation induces a switch to multipolar migration. This multipolar mode is less efficient but still leads to successful RGC layer formation. When both modes are inhibited though, RGCs fail to translocate and induce lamination defects. This indicates that correct RGC translocation is crucial for subsequent retinal lamination.
Collapse
Affiliation(s)
- Jaroslav Icha
- Max Planck Institute of Molecular Cell Biology and Genetics, 01307 Dresden, Germany
| | - Christiane Kunath
- Max Planck Institute of Molecular Cell Biology and Genetics, 01307 Dresden, Germany
| | - Mauricio Rocha-Martins
- Max Planck Institute of Molecular Cell Biology and Genetics, 01307 Dresden, Germany.,Instituto de Biofísica Carlos Chagas Filho, 21941-902 Rio de Janeiro, Brazil
| | - Caren Norden
- Max Planck Institute of Molecular Cell Biology and Genetics, 01307 Dresden, Germany
| |
Collapse
|
30
|
Tang X, Gao J, Jia X, Zhao W, Zhang Y, Pan W, He J. Bipotent progenitors as embryonic origin of retinal stem cells. J Cell Biol 2017; 216:1833-1847. [PMID: 28465291 PMCID: PMC5461025 DOI: 10.1083/jcb.201611057] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2016] [Revised: 03/01/2017] [Accepted: 04/03/2017] [Indexed: 01/24/2023] Open
Abstract
In lower vertebrates, retinal stem cells (RSCs) capable of producing all retinal cell types are a resource for retinal tissue growth throughout life. However, the embryonic origin of RSCs remains largely elusive. Using a Zebrabow-based clonal analysis, we characterized the RSC niche in the ciliary marginal zone of zebrafish retina and illustrate that blood vessels associated with RSCs are required for the maintenance of actively proliferating RSCs. Full lineage analysis of RSC progenitors reveals lineage patterns of RSC production. Moreover, in vivo lineage analysis demonstrates that these RSC progenitors are the direct descendants of a set of bipotent progenitors in the medial epithelial layer of developing optic vesicles, suggesting the involvement of the mixed-lineage states in the RSC lineage specification.
Collapse
Affiliation(s)
- Xia Tang
- State Key Laboratory of Neuroscience, Institute of Neuroscience, Shanghai Institutes for Biological Sciences, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Jianan Gao
- State Key Laboratory of Neuroscience, Institute of Neuroscience, Shanghai Institutes for Biological Sciences, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Xinling Jia
- State Key Laboratory of Neuroscience, Institute of Neuroscience, Shanghai Institutes for Biological Sciences, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Wencao Zhao
- Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences and Shanghai Jiao Tong University School of Medicine, Shanghai 200031, China
| | - Yijie Zhang
- State Key Laboratory of Neuroscience, Institute of Neuroscience, Shanghai Institutes for Biological Sciences, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Weijun Pan
- Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences and Shanghai Jiao Tong University School of Medicine, Shanghai 200031, China
| | - Jie He
- State Key Laboratory of Neuroscience, Institute of Neuroscience, Shanghai Institutes for Biological Sciences, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
| |
Collapse
|
31
|
Eldred MK, Charlton-Perkins M, Muresan L, Harris WA. Self-organising aggregates of zebrafish retinal cells for investigating mechanisms of neural lamination. Development 2017; 144:1097-1106. [PMID: 28174240 PMCID: PMC5358108 DOI: 10.1242/dev.142760] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Accepted: 01/17/2017] [Indexed: 02/04/2023]
Abstract
To investigate the cell-cell interactions necessary for the formation of retinal layers, we cultured dissociated zebrafish retinal progenitors in agarose microwells. Within these wells, the cells re-aggregated within hours, forming tight retinal organoids. Using a Spectrum of Fates zebrafish line, in which all different types of retinal neurons show distinct fluorescent spectra, we found that by 48 h in culture, the retinal organoids acquire a distinct spatial organisation, i.e. they became coarsely but clearly laminated. Retinal pigment epithelium cells were in the centre, photoreceptors and bipolar cells were next most central and amacrine cells and retinal ganglion cells were on the outside. Image analysis allowed us to derive quantitative measures of lamination, which we then used to find that Müller glia, but not RPE cells, are essential for this process.
Collapse
Affiliation(s)
- Megan K Eldred
- Department of Physiology, Development and Neuroscience, Cambridge University, Cambridge CB2 3DY, UK
| | - Mark Charlton-Perkins
- Department of Physiology, Development and Neuroscience, Cambridge University, Cambridge CB2 3DY, UK
| | - Leila Muresan
- Department of Physiology, Development and Neuroscience, Cambridge University, Cambridge CB2 3DY, UK
| | - William A Harris
- Department of Physiology, Development and Neuroscience, Cambridge University, Cambridge CB2 3DY, UK
| |
Collapse
|
32
|
Lepanto P, Davison C, Casanova G, Badano JL, Zolessi FR. Characterization of primary cilia during the differentiation of retinal ganglion cells in the zebrafish. Neural Dev 2016; 11:10. [PMID: 27053191 PMCID: PMC4823885 DOI: 10.1186/s13064-016-0064-z] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2015] [Accepted: 03/29/2016] [Indexed: 01/05/2023] Open
Abstract
Background Retinal ganglion cell (RGC) differentiation in vivo is a highly stereotyped process, likely resulting from the interaction of cell type-specific transcription factors and tissue-derived signaling factors. The primary cilium, as a signaling hub in the cell, may have a role during this process but its presence and localization during RGC generation, and its contribution to the process of cell differentiation, have not been previously assessed in vivo. Methods In this work we analyzed the distribution of primary cilia in vivo using laser scanning confocal microscopy, as well as their main ultrastructural features by transmission electron microscopy, in the early stages of retinal histogenesis in the zebrafish, around the time of RGC generation and initial differentiation. In addition, we knocked-down ift88 and elipsa, two genes with an essential role in cilia generation and maintenance, a treatment that caused a general reduction in organelle size. The effect on retinal development and RGC differentiation was assessed by confocal microscopy of transgenic or immunolabeled embryos. Results Our results show that retinal neuroepithelial cells have an apically-localized primary cilium usually protruding from the apical membrane. We also found a small proportion of sub-apical cilia, before and during the neurogenic period. This organelle was also present in an apical position in neuroblasts during apical process retraction and dendritogenesis, although between these stages cilia appeared highly dynamic regarding both presence and position. Disruption of cilia caused a decrease in the proliferation of retinal progenitors and a reduction of neural retina volume. In addition, retinal histogenesis was globally delayed albeit RGC layer formation was preferentially reduced with respect to the amacrine and photoreceptor cell layers. Conclusions These results indicate that primary cilia exhibit a highly dynamic behavior during early retinal differentiation, and that they are required for the proliferation and survival of retinal progenitors, as well as for neuronal generation, particularly of RGCs. Electronic supplementary material The online version of this article (doi:10.1186/s13064-016-0064-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Paola Lepanto
- Human Molecular Genetics Laboratory, Institut Pasteur de Montevideo, Mataojo 2020, Montevideo, 11400, Uruguay
| | - Camila Davison
- Cell Biology of Neural Development Laboratory, Institut Pasteur de Montevideo, Mataojo 2020, Montevideo, 11400, Uruguay.,Sección Biología Celular, Departamento de Biología Celular y Molecular, Facultad de Ciencias, Universidad de la República, Iguá 4225, Montevideo, 11400, Uruguay
| | - Gabriela Casanova
- Unidad de Microscopía Electrónica, Facultad de Ciencias, Universidad de la República, Iguá 4225, Montevideo, 11400, Uruguay
| | - Jose L Badano
- Human Molecular Genetics Laboratory, Institut Pasteur de Montevideo, Mataojo 2020, Montevideo, 11400, Uruguay.
| | - Flavio R Zolessi
- Cell Biology of Neural Development Laboratory, Institut Pasteur de Montevideo, Mataojo 2020, Montevideo, 11400, Uruguay. .,Sección Biología Celular, Departamento de Biología Celular y Molecular, Facultad de Ciencias, Universidad de la República, Iguá 4225, Montevideo, 11400, Uruguay.
| |
Collapse
|
33
|
Kei JNC, Dudczig S, Currie PD, Jusuf PR. Feedback from each retinal neuron population drives expression of subsequent fate determinant genes without influencing the cell cycle exit timing. J Comp Neurol 2016; 524:2553-66. [PMID: 26850379 DOI: 10.1002/cne.23976] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2015] [Revised: 01/26/2016] [Accepted: 01/29/2016] [Indexed: 11/10/2022]
Abstract
During neurogenesis, progenitors balance proliferation and cell cycle exit together with expression of fate determinant genes to ensure that the correct number of each of these neuron types is generated. Although intrinsic gene expression acting cell autonomously within each progenitor drives these processes, the final number of neurons generated is also influenced by extrinsic cues, representing a potential avenue to direct neurogenesis in developmental disorders or regenerative settings without the requirement to change intrinsic gene expression. Thus, it is important to understand which of these stages of neurogenesis are amenable to such extrinsic influences. Additionally, all types of neurons are specified in a highly conserved histogenic order, although its significance is unknown. This study makes use of conserved patterns of neurogenesis in the relatively simple yet highly organized zebrafish retina model, in which such histogenic birth order is well characterized. We directly visualize and quantify birth dates and cell fate determinant expression in WT vs. environments lacking different neuronal populations. This study shows that extrinsic feedback from developing retinal neurons is important for the temporal expression of intrinsic fate determinants but not for the timing of birth dates. We found no changes in cell cycle exit timing but did find a significant delay in the expression of genes driving the generation only of later- but not earlier-born cells, suggesting that the robustness of this process depends on continuous feedback from earlier-formed cell types. Thus, extrinsic cues selectively influence cell fate determinant progression, which may explain the function of the retinal histogenic order observed. J. Comp. Neurol. 524:2553-2566, 2016. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Jeremy Ng Chi Kei
- Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria, 3800, Australia
| | - Stefanie Dudczig
- Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria, 3800, Australia
| | - Peter D Currie
- Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria, 3800, Australia
| | - Patricia R Jusuf
- Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria, 3800, Australia
| |
Collapse
|
34
|
|
35
|
MacDonald RB, Randlett O, Oswald J, Yoshimatsu T, Franze K, Harris WA. Müller glia provide essential tensile strength to the developing retina. J Cell Biol 2015; 210:1075-83. [PMID: 26416961 PMCID: PMC4586739 DOI: 10.1083/jcb.201503115] [Citation(s) in RCA: 82] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
When the formation of Müller glia is inhibited in the zebrafish retina, a major consequence is that the retina begins to rip apart due to a loss of the mechanical resilience that these glial cells provide to the neural tissue. To investigate the cellular basis of tissue integrity in a vertebrate central nervous system (CNS) tissue, we eliminated Müller glial cells (MG) from the zebrafish retina. For well over a century, glial cells have been ascribed a mechanical role in the support of neural tissues, yet this idea has not been specifically tested in vivo. We report here that retinas devoid of MG rip apart, a defect known as retinoschisis. Using atomic force microscopy, we show that retinas without MG have decreased resistance to tensile stress and are softer than controls. Laser ablation of MG processes showed that these cells are under tension in the tissue. Thus, we propose that MG act like springs that hold the neural retina together, finally confirming an active mechanical role of glial cells in the CNS.
Collapse
Affiliation(s)
- Ryan B MacDonald
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge CB2 3DY, England, UK
| | - Owen Randlett
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge CB2 3DY, England, UK
| | - Julia Oswald
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge CB2 3DY, England, UK
| | - Takeshi Yoshimatsu
- Department of Biological Structure, University of Washington, Seattle, WA 98195
| | - Kristian Franze
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge CB2 3DY, England, UK
| | - William A Harris
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge CB2 3DY, England, UK
| |
Collapse
|
36
|
Boije H, Rulands S, Dudczig S, Simons BD, Harris WA. The Independent Probabilistic Firing of Transcription Factors: A Paradigm for Clonal Variability in the Zebrafish Retina. Dev Cell 2015; 34:532-43. [PMID: 26343455 PMCID: PMC4572358 DOI: 10.1016/j.devcel.2015.08.011] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2014] [Revised: 06/29/2015] [Accepted: 08/13/2015] [Indexed: 01/31/2023]
Abstract
Early retinal progenitor cells (RPCs) in vertebrates produce lineages that vary greatly both in terms of cell number and fate composition, yet how this variability is achieved remains unknown. One possibility is that these RPCs are individually distinct and that each gives rise to a unique lineage. Another is that stochastic mechanisms play upon the determinative machinery of equipotent early RPCs to drive clonal variability. Here we show that a simple model, based on the independent firing of key fate-influencing transcription factors, can quantitatively account for the intrinsic clonal variance in the zebrafish retina and predict the distributions of neuronal cell types in clones where one or more of these fates are made unavailable. A simple quantitative model can explain clonal variability in the retina This model is based on the firing probabilities of key transcription factors These probabilities are shown to be largely independent of each other The environment has only a minor effect on these probabilities
Collapse
Affiliation(s)
- Henrik Boije
- Department of Physiology, Development and Neuroscience, Cambridge University, Cambridge CB2 3DY, UK; Department of Neuroscience, Uppsala University, 751 24 Uppsala, Sweden
| | - Steffen Rulands
- Department of Physics, Cambridge University, Cambridge CB3 0HE, UK
| | - Stefanie Dudczig
- Department of Physiology, Development and Neuroscience, Cambridge University, Cambridge CB2 3DY, UK
| | | | - William A Harris
- Department of Physiology, Development and Neuroscience, Cambridge University, Cambridge CB2 3DY, UK.
| |
Collapse
|
37
|
Chow RW, Almeida AD, Randlett O, Norden C, Harris WA. Inhibitory neuron migration and IPL formation in the developing zebrafish retina. Development 2015; 142:2665-77. [PMID: 26116662 PMCID: PMC4529032 DOI: 10.1242/dev.122473] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2015] [Accepted: 06/11/2015] [Indexed: 12/22/2022]
Abstract
The mature vertebrate retina is a highly ordered neuronal network of cell bodies and synaptic neuropils arranged in distinct layers. Little, however, is known about the emergence of this spatial arrangement. Here, we investigate how the three main types of retinal inhibitory neuron (RIN) – horizontal cells (HCs), inner nuclear layer amacrine cells (iACs) and displaced amacrine cells (dACs) – reach their specific laminar positions during development. Using in vivo time-lapse imaging of zebrafish retinas, we show that RINs undergo distinct phases of migration. The first phase, common to all RINs, is bipolar migration directed towards the apicobasal centre of the retina. All RINs then transition to a less directionally persistent multipolar phase of migration. Finally, HCs, iACs and dACs each undergo cell type-specific migration. In contrast to current hypotheses, we find that most dACs send processes into the forming inner plexiform layer (IPL) before migrating through it and inverting their polarity. By imaging and quantifying the dynamics of HCs, iACs and dACs from birth to final position, this study thus provides evidence for distinct and new migration patterns during retinal lamination and insights into the initiation of IPL formation. Highlighted article: The quantification of cellular behaviour in real time provides new insights into interneuron migration and inner plexiform layer formation during the lamination of the zebrafish retina.
Collapse
Affiliation(s)
- Renee W Chow
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge CB2 3DY, UK
| | - Alexandra D Almeida
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge CB2 3DY, UK
| | - Owen Randlett
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge CB2 3DY, UK
| | - Caren Norden
- MPI of Molecular Cell Biology and Genetics, Pfotenhauerstraße 108, Dresden 01307, Germany
| | - William A Harris
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge CB2 3DY, UK
| |
Collapse
|
38
|
Whalley K. A colourful concept. Nat Rev Neurosci 2014. [DOI: 10.1038/nrn3751] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|