1
|
Bassi I, Grunspan M, Hen G, Ravichandran KA, Moshe N, Gutierrez-Miranda L, Safriel SR, Kostina D, Shen A, Ruiz de Almodovar C, Yaniv K. Endolysosomal dysfunction in radial glia progenitor cells leads to defective cerebral angiogenesis and compromised blood-brain barrier integrity. Nat Commun 2024; 15:8158. [PMID: 39289367 PMCID: PMC11408700 DOI: 10.1038/s41467-024-52365-8] [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: 11/07/2023] [Accepted: 09/04/2024] [Indexed: 09/19/2024] Open
Abstract
The neurovascular unit (NVU) is a complex multicellular structure that helps maintain cerebral homeostasis and blood-brain barrier (BBB) integrity. While extensive evidence links NVU alterations to cerebrovascular diseases and neurodegeneration, the underlying molecular mechanisms remain unclear. Here, we use zebrafish embryos carrying a mutation in Scavenger Receptor B2, a highly conserved endolysosomal protein expressed predominantly in Radial Glia Cells (RGCs), to investigate the interplay among different NVU components. Through live imaging and genetic manipulations, we demonstrate that compromised acidification of the endolysosomal compartment in mutant RGCs leads to impaired Notch3 signaling, thereby inducing excessive neurogenesis and reduced glial differentiation. We further demonstrate that alterations to the neuron/glia balance result in impaired VEGF and Wnt signaling, leading to severe vascular defects, hemorrhages, and a leaky BBB. Altogether, our findings provide insights into NVU formation and function and offer avenues for investigating diseases involving white matter defects and vascular abnormalities.
Collapse
Affiliation(s)
- Ivan Bassi
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Moshe Grunspan
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Gideon Hen
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Kishore A Ravichandran
- Institute for Neurovascular Cell Biology, Medical Faculty, University of Bonn, Bonn, Germany
| | - Noga Moshe
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Laura Gutierrez-Miranda
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Stav R Safriel
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Daria Kostina
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Amitay Shen
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Carmen Ruiz de Almodovar
- Institute for Neurovascular Cell Biology, Medical Faculty, University of Bonn, Bonn, Germany
- Schlegel Chair for Neurovascular Cell Biology, University of Bonn, Bonn, Germany
| | - Karina Yaniv
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot, Israel.
| |
Collapse
|
2
|
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
|
3
|
Uribe-Arias A, Rozenblat R, Vinepinsky E, Marachlian E, Kulkarni A, Zada D, Privat M, Topsakalian D, Charpy S, Candat V, Nourin S, Appelbaum L, Sumbre G. Radial astrocyte synchronization modulates the visual system during behavioral-state transitions. Neuron 2023; 111:4040-4057.e6. [PMID: 37863038 PMCID: PMC10783638 DOI: 10.1016/j.neuron.2023.09.022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 08/01/2023] [Accepted: 09/15/2023] [Indexed: 10/22/2023]
Abstract
Glial cells support the function of neurons. Recent evidence shows that astrocytes are also involved in brain computations. To explore whether and how their excitable nature affects brain computations and motor behaviors, we used two-photon Ca2+ imaging of zebrafish larvae expressing GCaMP in both neurons and radial astrocytes (RAs). We found that in the optic tectum, RAs synchronize their Ca2+ transients immediately after the end of an escape behavior. Using optogenetics, ablations, and a genetically encoded norepinephrine sensor, we observed that RA synchronous Ca2+ events are mediated by the locus coeruleus (LC)-norepinephrine circuit. RA synchronization did not induce direct excitation or inhibition of tectal neurons. Nevertheless, it modulated the direction selectivity and the long-distance functional correlations among neurons. This mechanism supports freezing behavior following a switch to an alerted state. These results show that LC-mediated neuro-glial interactions modulate the visual system during transitions between behavioral states.
Collapse
Affiliation(s)
- Alejandro Uribe-Arias
- Institut de Biologie de l'ENS (IBENS), Département de biologie, École normale supérieure, CNRS, INSERM, Université PSL, 75005 Paris, France
| | - Rotem Rozenblat
- The Faculty of Life Sciences and The Multidisciplinary Brain Research Center, Bar-Ilan University, Ramat-Gan, Israel
| | - Ehud Vinepinsky
- Institut de Biologie de l'ENS (IBENS), Département de biologie, École normale supérieure, CNRS, INSERM, Université PSL, 75005 Paris, France
| | - Emiliano Marachlian
- Institut de Biologie de l'ENS (IBENS), Département de biologie, École normale supérieure, CNRS, INSERM, Université PSL, 75005 Paris, France
| | - Anirudh Kulkarni
- Institut de Biologie de l'ENS (IBENS), Département de biologie, École normale supérieure, CNRS, INSERM, Université PSL, 75005 Paris, France
| | - David Zada
- The Faculty of Life Sciences and The Multidisciplinary Brain Research Center, Bar-Ilan University, Ramat-Gan, Israel
| | - Martin Privat
- Institut de Biologie de l'ENS (IBENS), Département de biologie, École normale supérieure, CNRS, INSERM, Université PSL, 75005 Paris, France
| | - Diego Topsakalian
- Institut de Biologie de l'ENS (IBENS), Département de biologie, École normale supérieure, CNRS, INSERM, Université PSL, 75005 Paris, France
| | - Sarah Charpy
- Institut de Biologie de l'ENS (IBENS), Département de biologie, École normale supérieure, CNRS, INSERM, Université PSL, 75005 Paris, France
| | - Virginie Candat
- Institut de Biologie de l'ENS (IBENS), Département de biologie, École normale supérieure, CNRS, INSERM, Université PSL, 75005 Paris, France
| | - Sarah Nourin
- Institut de Biologie de l'ENS (IBENS), Département de biologie, École normale supérieure, CNRS, INSERM, Université PSL, 75005 Paris, France
| | - Lior Appelbaum
- The Faculty of Life Sciences and The Multidisciplinary Brain Research Center, Bar-Ilan University, Ramat-Gan, Israel
| | - Germán Sumbre
- Institut de Biologie de l'ENS (IBENS), Département de biologie, École normale supérieure, CNRS, INSERM, Université PSL, 75005 Paris, France.
| |
Collapse
|
4
|
Krylov A, Yu S, Veen K, Newton A, Ye A, Qin H, He J, Jusuf PR. Heterogeneity in quiescent Müller glia in the uninjured zebrafish retina drive differential responses following photoreceptor ablation. Front Mol Neurosci 2023; 16:1087136. [PMID: 37575968 PMCID: PMC10413128 DOI: 10.3389/fnmol.2023.1087136] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Accepted: 06/23/2023] [Indexed: 08/15/2023] Open
Abstract
Introduction Loss of neurons in the neural retina is a leading cause of vision loss. While humans do not possess the capacity for retinal regeneration, zebrafish can achieve this through activation of resident Müller glia. Remarkably, despite the presence of Müller glia in humans and other mammalian vertebrates, these cells lack an intrinsic ability to contribute to regeneration. Upon activation, zebrafish Müller glia can adopt a stem cell-like state, undergo proliferation and generate new neurons. However, the underlying molecular mechanisms of this activation subsequent retinal regeneration remains unclear. Methods/Results To address this, we performed single-cell RNA sequencing (scRNA-seq) and report remarkable heterogeneity in gene expression within quiescent Müller glia across distinct dorsal, central and ventral retina pools of such cells. Next, we utilized a genetically driven, chemically inducible nitroreductase approach to study Müller glia activation following selective ablation of three distinct photoreceptor subtypes: long wavelength sensitive cones, short wavelength sensitive cones, and rods. There, our data revealed that a region-specific bias in activation of Müller glia exists in the zebrafish retina, and this is independent of the distribution of the ablated cell type across retinal regions. Notably, gene ontology analysis revealed that injury-responsive dorsal and central Müller glia express genes related to dorsal/ventral pattern formation, growth factor activity, and regulation of developmental process. Through scRNA-seq analysis, we identify a shared genetic program underlying initial Müller glia activation and cell cycle entry, followed by differences that drive the fate of regenerating neurons. We observed an initial expression of AP-1 and injury-responsive transcription factors, followed by genes involved in Notch signaling, ribosome biogenesis and gliogenesis, and finally expression of cell cycle, chromatin remodeling and microtubule-associated genes. Discussion Taken together, our findings document the regional specificity of gene expression within quiescent Müller glia and demonstrate unique Müller glia activation and regeneration features following neural ablation. These findings will improve our understanding of the molecular pathways relevant to neural regeneration in the retina.
Collapse
Affiliation(s)
- Aaron Krylov
- School of BioSciences, University of Melbourne, Parkville, VIC, Australia
| | - Shuguang Yu
- State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Institute of Neuroscience, Chinese Academy of Sciences, Shanghai, China
| | - Kellie Veen
- School of BioSciences, University of Melbourne, Parkville, VIC, Australia
| | - Axel Newton
- School of BioSciences, University of Melbourne, Parkville, VIC, Australia
| | - Aojun Ye
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Huiwen Qin
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Jie He
- State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Institute of Neuroscience, Chinese Academy of Sciences, Shanghai, China
| | - Patricia R. Jusuf
- School of BioSciences, University of Melbourne, Parkville, VIC, Australia
| |
Collapse
|
5
|
Bai C, Zheng Y, Tian L, Lin J, Song Y, Huang C, Dong Q, Chen J. Structure-based developmental toxicity and ASD-phenotypes of bisphenol A analogues in embryonic zebrafish. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2023; 253:114643. [PMID: 36805134 DOI: 10.1016/j.ecoenv.2023.114643] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 02/08/2023] [Accepted: 02/11/2023] [Indexed: 06/18/2023]
Abstract
Autism spectrum disorder (ASD) is a neurodevelopmental disorder that has become more prevalent in recent years. Environmental endocrine disruptor bisphenol A (BPA) has been linked to ASD. BPA analogues (BPs) are structure-modified substitutes widely used as safer alternatives in consumer products, yet few studies have explored the developmental neurotoxicity (DNT) of BPA analogues. In the present study, we used the larval zebrafish model to assess the DNT effects of BPA and its analogues. Our results showed that many BPA analogues are more toxic than BPA in the embryonic zebrafish assay regarding teratogenic effect and mortality, which may partially due to differences in lipophilicity and/or different substitutes of structural function groups such as CF3, benzene, or cyclohexane. At sublethal concentrations, zebrafish embryos exposed to BPA or BPs also displayed reduced prosocial behavior in later larval development, evidenced by increased nearest neighbor distance (NND) and the interindividual distance (IID) in shoaling, which appears to be structurally independent. An in-depth analysis of BPA, bisphenol F (BPF), and bisphenol S (BPS) revealed macrocephaly and ASD-like behavioral deficits resulting from exposures to sublethal concentrations of these chemicals. The ASD-like behavioral deficits were characterized by hyperactivity, increased anxiety-like behavior, and decreased social contact. Mechanistically, accelerated neurogenesis that manifested by increased cell proliferation, the proportion of newborn mature neurons, and the number of neural stem cells in proliferation, as well as upregulated genes related to the K+ channels, may have contributed to the observed ASD-like morphological and behavioral alterations. Our findings indicate that BPF and BPS may also pose significant risks to ASD development in humans and highlight the importance of a comprehensive assessment of DNT effects for all BPA analogues in the future.
Collapse
Affiliation(s)
- Chenglian Bai
- School of Public Health and Management, Wenzhou Medical University, Wenzhou 325035, PR China; The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou 325035, PR China
| | - Yi Zheng
- School of Laboratory Medicine and Life Science, Wenzhou Medical University, Wenzhou 325035, PR China
| | - Linjie Tian
- School of Public Health and Management, Wenzhou Medical University, Wenzhou 325035, PR China
| | - Jian Lin
- The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou 325035, PR China
| | - Yang Song
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, PR China
| | - Changjiang Huang
- School of Public Health and Management, Wenzhou Medical University, Wenzhou 325035, PR China
| | - Qiaoxiang Dong
- School of Public Health and Management, Wenzhou Medical University, Wenzhou 325035, PR China; The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou 325035, PR China.
| | - Jiangfei Chen
- School of Public Health and Management, Wenzhou Medical University, Wenzhou 325035, PR China.
| |
Collapse
|
6
|
Shoenhard H, Jain RA, Granato M. The calcium-sensing receptor (CaSR) regulates zebrafish sensorimotor decision making via a genetically defined cluster of hindbrain neurons. Cell Rep 2022; 41:111790. [PMID: 36476852 PMCID: PMC9813870 DOI: 10.1016/j.celrep.2022.111790] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 09/21/2022] [Accepted: 11/15/2022] [Indexed: 12/12/2022] Open
Abstract
Decision making is a fundamental nervous system function that ranges widely in complexity and speed of execution. We previously established larval zebrafish as a model for sensorimotor decision making and identified the G-protein-coupled calcium-sensing receptor (CaSR) to be critical for this process. Here, we report that CaSR functions in neurons to dynamically regulate the bias between two behavioral outcomes: escapes and reorientations. By employing a computational guided transgenic strategy, we identify a genetically defined neuronal cluster in the hindbrain as a key candidate site for CaSR function. Finally, we demonstrate that transgenic CaSR expression targeting this cluster consisting of a few hundred neurons shifts behavioral bias in wild-type animals and restores decision making deficits in CaSR mutants. Combined, our data provide a rare example of a G-protein-coupled receptor that biases vertebrate sensorimotor decision making via a defined neuronal cluster.
Collapse
Affiliation(s)
- Hannah Shoenhard
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Roshan A Jain
- Department of Biology, Haverford College, Haverford, PA 19041, USA
| | - Michael Granato
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
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
|
Shimizu Y, Kawasaki T, Deguchi T. Gfap transgenic medaka as a novel reporter line for neural stem cells. Gene X 2022; 820:146213. [PMID: 35104578 DOI: 10.1016/j.gene.2022.146213] [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: 06/07/2021] [Revised: 12/02/2021] [Accepted: 01/13/2022] [Indexed: 11/04/2022] Open
Abstract
Radial glial cells (RGCs) play an essential role in developing, maintaining, and repairing the central nervous system (CNS). However, a specific reporter line of RGCs is limited in medaka. Glial fibrillary acid protein (GFAP) is abundant in teleost CNS, including the brain and spinal cord, and is a possible candidate for a marker for RGCs in medaka CNS. We generated a transgenic medaka in which enhanced green fluorescent protein (EGFP) expression is regulated under putative medaka gfap regulatory elements. We observed EGFP expression in the CNS of live larval and juvenile medaka through the transparent body of the See-through medaka strain. Histological analysis for juvenile and adult Tg(gfap:EGFP) medaka showed that EGFP was expressed in GFAP-positive cells in the telencephalon, optic tectum, retina, and spinal cord. We further found another EGFP expressing cells in the optic tectum and retina. These cells are possibly neuroepithelial-like stem cells, deducing from the distribution of these EGFP-positive cells. We concluded that this reporter line would be valuable in the investigation of neural stem cell function during the development and regeneration of medaka CNS visualizing two types of neural stem cells, RGCs and neuroepithelial-like stem cells.
Collapse
Affiliation(s)
- Yuki Shimizu
- Functional Biomolecular Research Group, Biomedical Research Institute, National Institute of Advanced Industrial Science and Technology, Osaka, Japan; DBT-AIST International Laboratory for Advanced Biomedicine, National Institute of Advanced Industrial Science and Technology, Osaka, Japan.
| | - Takashi Kawasaki
- Functional Biomolecular Research Group, Biomedical Research Institute, National Institute of Advanced Industrial Science and Technology, Osaka, Japan
| | - Tomonori Deguchi
- Advanced Genome Design Research Group, Biomedical Research Institute, National Institute of Advanced Industrial Science and Technology, Osaka, Japan
| |
Collapse
|
9
|
Chowdhury K, Lin S, Lai SL. Comparative Study in Zebrafish and Medaka Unravels the Mechanisms of Tissue Regeneration. Front Ecol Evol 2022. [DOI: 10.3389/fevo.2022.783818] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Tissue regeneration has been in the spotlight of research for its fascinating nature and potential applications in human diseases. The trait of regenerative capacity occurs diversely across species and tissue contexts, while it seems to decline over evolution. Organisms with variable regenerative capacity are usually distinct in phylogeny, anatomy, and physiology. This phenomenon hinders the feasibility of studying tissue regeneration by directly comparing regenerative with non-regenerative animals, such as zebrafish (Danio rerio) and mice (Mus musculus). Medaka (Oryzias latipes) is a fish model with a complete reference genome and shares a common ancestor with zebrafish approximately 110–200 million years ago (compared to 650 million years with mice). Medaka shares similar features with zebrafish, including size, diet, organ system, gross anatomy, and living environment. However, while zebrafish regenerate almost every organ upon experimental injury, medaka shows uneven regenerative capacity. Their common and distinct biological features make them a unique platform for reciprocal analyses to understand the mechanisms of tissue regeneration. Here we summarize current knowledge about tissue regeneration in these fish models in terms of injured tissues, repairing mechanisms, available materials, and established technologies. We further highlight the concept of inter-species and inter-organ comparisons, which may reveal mechanistic insights and hint at therapeutic strategies for human diseases.
Collapse
|
10
|
Burrows DRW, Samarut É, Liu J, Baraban SC, Richardson MP, Meyer MP, Rosch RE. Imaging epilepsy in larval zebrafish. Eur J Paediatr Neurol 2020; 24:70-80. [PMID: 31982307 PMCID: PMC7035958 DOI: 10.1016/j.ejpn.2020.01.006] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Revised: 01/03/2020] [Accepted: 01/04/2020] [Indexed: 12/19/2022]
Abstract
Our understanding of the genetic aetiology of paediatric epilepsies has grown substantially over the last decade. However, in order to translate improved diagnostics to personalised treatments, there is an urgent need to link molecular pathophysiology in epilepsy to whole-brain dynamics in seizures. Zebrafish have emerged as a promising new animal model for epileptic seizure disorders, with particular relevance for genetic and developmental epilepsies. As a novel model organism for epilepsy research they combine key advantages: the small size of larval zebrafish allows high throughput in vivo experiments; the availability of advanced genetic tools allows targeted modification to model specific human genetic disorders (including genetic epilepsies) in a vertebrate system; and optical access to the entire central nervous system has provided the basis for advanced microscopy technologies to image structure and function in the intact larval zebrafish brain. There is a growing body of literature describing and characterising features of epileptic seizures and epilepsy in larval zebrafish. Recently genetically encoded calcium indicators have been used to investigate the neurobiological basis of these seizures with light microscopy. This approach offers a unique window into the multiscale dynamics of epileptic seizures, capturing both whole-brain dynamics and single-cell behaviour concurrently. At the same time, linking observations made using calcium imaging in the larval zebrafish brain back to an understanding of epileptic seizures largely derived from cortical electrophysiological recordings in human patients and mammalian animal models is non-trivial. In this review we briefly illustrate the state of the art of epilepsy research in zebrafish with particular focus on calcium imaging of epileptic seizures in the larval zebrafish. We illustrate the utility of a dynamic systems perspective on the epileptic brain for providing a principled approach to linking observations across species and identifying those features of brain dynamics that are most relevant to epilepsy. In the following section we survey the literature for imaging features associated with epilepsy and epileptic seizures and link these to observations made from humans and other more traditional animal models. We conclude by identifying the key challenges still facing epilepsy research in the larval zebrafish and indicate strategies for future research to address these and integrate more directly with the themes and questions that emerge from investigating epilepsy in other model systems and human patients.
Collapse
Affiliation(s)
- D R W Burrows
- MRC Centre for Neurodevelopmental Disorders, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - É Samarut
- Department of Neurosciences, Research Center of the University of Montreal Hospital Center, Montreal, Quebec, Canada
| | - J Liu
- Department of Neurological Surgery and Weill Institute for Neuroscience, University of California, San Francisco, San Francisco, CA, USA
| | - S C Baraban
- Department of Neurological Surgery and Weill Institute for Neuroscience, University of California, San Francisco, San Francisco, CA, USA
| | - M P Richardson
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - M P Meyer
- MRC Centre for Neurodevelopmental Disorders, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK; Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - R E Rosch
- MRC Centre for Neurodevelopmental Disorders, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK; Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA; Department of Paediatric Neurology, Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK.
| |
Collapse
|
11
|
Rosch R, Burrows DRW, Jones LB, Peters CH, Ruben P, Samarut É. Functional Genomics of Epilepsy and Associated Neurodevelopmental Disorders Using Simple Animal Models: From Genes, Molecules to Brain Networks. Front Cell Neurosci 2019; 13:556. [PMID: 31920556 PMCID: PMC6923670 DOI: 10.3389/fncel.2019.00556] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Accepted: 12/02/2019] [Indexed: 01/06/2023] Open
Abstract
The genetic diagnosis of patients with seizure disorders has been improved significantly by the development of affordable next-generation sequencing technologies. Indeed, in the last 20 years, dozens of causative genes and thousands of associated variants have been described and, for many patients, are now considered responsible for their disease. However, the functional consequences of these mutations are often not studied in vivo, despite such studies being central to understanding pathogenic mechanisms and identifying novel therapeutic avenues. One main roadblock to functionally characterizing pathogenic mutations is generating and characterizing in vivo mammalian models carrying clinically relevant variants in specific genes identified in patients. Although the emergence of new mutagenesis techniques facilitates the production of rodent mutants, the fact that early development occurs internally hampers the investigation of gene function during neurodevelopment. In this context, functional genomics studies using simple animal models such as flies or fish are advantageous since they open a dynamic window of investigation throughout embryonic development. In this review, we will summarize how the use of simple animal models can fill the gap between genetic diagnosis and functional and phenotypic correlates of gene function in vivo. In particular, we will discuss how these simple animals offer the possibility to study gene function at multiple scales, from molecular function (i.e., ion channel activity), to cellular circuit and brain network dynamics. As a result, simple model systems offer alternative avenues of investigation to model aspects of the disease phenotype not currently possible in rodents, which can help to unravel the pathogenic substratum in vivo.
Collapse
Affiliation(s)
- Richard Rosch
- MRC Centre for Neurodevelopmental Disorders, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, United Kingdom
- Department of Paediatric Neurology, Great Ormond Street Hospital, NHS Foundation Trust, London, United Kingdom
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, United States
| | - Dominic R. W. Burrows
- MRC Centre for Neurodevelopmental Disorders, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, United Kingdom
| | - Laura B. Jones
- Department of Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, BC, Canada
| | - Colin H. Peters
- Department of Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, BC, Canada
| | - Peter Ruben
- Department of Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, BC, Canada
| | - Éric Samarut
- Department of Neurosciences, Research Center of the University of Montreal Hospital Center (CRCHUM), Université de Montréal, Montreal, QC, Canada
- Modelis Inc., Montreal, QC, Canada
| |
Collapse
|
12
|
Glia-neuron interactions underlie state transitions to generalized seizures. Nat Commun 2019; 10:3830. [PMID: 31444362 PMCID: PMC6707163 DOI: 10.1038/s41467-019-11739-z] [Citation(s) in RCA: 79] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Accepted: 07/31/2019] [Indexed: 11/08/2022] Open
Abstract
Brain activity and connectivity alter drastically during epileptic seizures. The brain networks shift from a balanced resting state to a hyperactive and hypersynchronous state. It is, however, less clear which mechanisms underlie the state transitions. By studying neural and glial activity in zebrafish models of epileptic seizures, we observe striking differences between these networks. During the preictal period, neurons display a small increase in synchronous activity only locally, while the gap-junction-coupled glial network was highly active and strongly synchronized across large distances. The transition from a preictal state to a generalized seizure leads to an abrupt increase in neural activity and connectivity, which is accompanied by a strong alteration in glia-neuron interactions and a massive increase in extracellular glutamate. Optogenetic activation of glia excites nearby neurons through the action of glutamate and gap junctions, emphasizing a potential role for glia-glia and glia-neuron connections in the generation of epileptic seizures. During epileptic seizures, neural activity across the brain switches into a hyperactive and hypersynchronized state. Here, the authors report on the role of glia-glia and glia-neuron interactions in mediating the changes that result in the ictal state in a zebrafish model of epilepsy.
Collapse
|
13
|
Chen J, Lei L, Tian L, Hou F, Roper C, Ge X, Zhao Y, Chen Y, Dong Q, Tanguay RL, Huang C. Developmental and behavioral alterations in zebrafish embryonically exposed to valproic acid (VPA): An aquatic model for autism. Neurotoxicol Teratol 2018; 66:8-16. [PMID: 29309833 DOI: 10.1016/j.ntt.2018.01.002] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Revised: 12/28/2017] [Accepted: 01/02/2018] [Indexed: 01/20/2023]
Abstract
Autism spectrum disorder (ASD) has complex neurodevelopmental impairments and origins that are linked to both genetic and environmental factors. Hence, there is an urgency to establish animal models with ASD-like characteristics to understand the underlying mechanisms of ASD. Prenatal exposure to valproic acid (VPA) produced ASD-like symptoms in humans, rats, and recently zebrafish. The present study investigated the use of VPA exposure to generate an ASD model in zebrafish. Early life stage exposures produced ASD-like phenotypes in the developing brain development and behavioral changes in embryonic and larval zebrafish. Our findings revealed that treating zebrafish embryos with VPA starting at 8h post fertilization (hpf) resulted in significant: increase in the ASD macrocephalic phenotype; hyperactivity of embryo/larvae movement behaviors; and increases of ASD-like larval social behaviors. Further analysis showed increases in cell proliferation, the proportion of mature newborn neurons, and neural stem cell proliferation in the brain region, which may contribute to the brain overgrowth and macrocephaly observed following VPA exposure. Our study demonstrated that VPA exposure generates ASD-like phenotypes and behaviors, indicating that zebrafish is an alternative model to investigate underlying ASD mechanisms.
Collapse
Affiliation(s)
- Jiangfei Chen
- Zhejiang Provincial Key Laboratory for Technology and Application of Model Organisms, Institute of Environmental Safety and Human Health, Wenzhou Medical University, Wenzhou 325035, China
| | - Lei Lei
- Zhejiang Provincial Key Laboratory for Technology and Application of Model Organisms, Institute of Environmental Safety and Human Health, Wenzhou Medical University, Wenzhou 325035, China
| | - Linjie Tian
- Zhejiang Provincial Key Laboratory for Technology and Application of Model Organisms, Institute of Environmental Safety and Human Health, Wenzhou Medical University, Wenzhou 325035, China
| | - Fei Hou
- Zhejiang Provincial Key Laboratory for Technology and Application of Model Organisms, Institute of Environmental Safety and Human Health, Wenzhou Medical University, Wenzhou 325035, China
| | - Courtney Roper
- Environmental and Molecular Toxicology, The Sinnhuber Aquatic Research Laboratory and the Environmental Health Sciences Center, Oregon State University, Corvallis, OR 97333, USA
| | - Xiaoqing Ge
- Zhejiang Provincial Key Laboratory for Technology and Application of Model Organisms, Institute of Environmental Safety and Human Health, Wenzhou Medical University, Wenzhou 325035, China
| | - Yuxin Zhao
- Zhejiang Provincial Key Laboratory for Technology and Application of Model Organisms, Institute of Environmental Safety and Human Health, Wenzhou Medical University, Wenzhou 325035, China
| | - Yuanhong Chen
- Zhejiang Provincial Key Laboratory for Technology and Application of Model Organisms, Institute of Environmental Safety and Human Health, Wenzhou Medical University, Wenzhou 325035, China
| | - Qiaoxiang Dong
- Zhejiang Provincial Key Laboratory for Technology and Application of Model Organisms, Institute of Environmental Safety and Human Health, Wenzhou Medical University, Wenzhou 325035, China
| | - Robert L Tanguay
- Environmental and Molecular Toxicology, The Sinnhuber Aquatic Research Laboratory and the Environmental Health Sciences Center, Oregon State University, Corvallis, OR 97333, USA.
| | - Changjiang Huang
- Zhejiang Provincial Key Laboratory for Technology and Application of Model Organisms, Institute of Environmental Safety and Human Health, Wenzhou Medical University, Wenzhou 325035, China.
| |
Collapse
|
14
|
Tahara N, Brush M, Kawakami Y. Cell migration during heart regeneration in zebrafish. Dev Dyn 2016; 245:774-87. [PMID: 27085002 PMCID: PMC5839122 DOI: 10.1002/dvdy.24411] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2013] [Revised: 03/17/2016] [Accepted: 04/12/2016] [Indexed: 12/27/2022] Open
Abstract
Zebrafish possess the remarkable ability to regenerate injured hearts as adults, which contrasts the very limited ability in mammals. Although very limited, mammalian hearts do in fact have measurable levels of cardiomyocyte regeneration. Therefore, elucidating mechanisms of zebrafish heart regeneration would provide information of naturally occurring regeneration to potentially apply to mammalian studies, in addition to addressing this biologically interesting phenomenon in itself. Studies over the past 13 years have identified processes and mechanisms of heart regeneration in zebrafish. After heart injury, pre-existing cardiomyocytes dedifferentiate, enter the cell cycle, and repair the injured myocardium. This process requires interaction with epicardial cells, endocardial cells, and vascular endothelial cells. Epicardial cells envelope the heart, while endocardial cells make up the inner lining of the heart. They provide paracrine signals to cardiomyocytes to regenerate the injured myocardium, which is vascularized during heart regeneration. In addition, accumulating results suggest that local migration of these major cardiac cell types have roles in heart regeneration. In this review, we summarize the characteristics of various heart injury methods used in the research community and regeneration of the major cardiac cell types. Then, we discuss local migration of these cardiac cell types and immune cells during heart regeneration. Developmental Dynamics 245:774-787, 2016. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Naoyuki Tahara
- Department of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, Minnesota
- Stem Cell Institute, University of Minnesota, Minneapolis, Minnesota
| | - Michael Brush
- Department of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, Minnesota
| | - Yasuhiko Kawakami
- Department of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, Minnesota
- Stem Cell Institute, University of Minnesota, Minneapolis, Minnesota
| |
Collapse
|
15
|
Johnson K, Barragan J, Bashiruddin S, Smith CJ, Tyrrell C, Parsons MJ, Doris R, Kucenas S, Downes GB, Velez CM, Schneider C, Sakai C, Pathak N, Anderson K, Stein R, Devoto SH, Mumm JS, Barresi MJF. Gfap-positive radial glial cells are an essential progenitor population for later-born neurons and glia in the zebrafish spinal cord. Glia 2016; 64:1170-89. [PMID: 27100776 PMCID: PMC4918407 DOI: 10.1002/glia.22990] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2015] [Revised: 03/27/2016] [Accepted: 03/30/2016] [Indexed: 11/12/2022]
Abstract
Radial glial cells are presumptive neural stem cells (NSCs) in the developing nervous system. The direct requirement of radial glia for the generation of a diverse array of neuronal and glial subtypes, however, has not been tested. We employed two novel transgenic zebrafish lines and endogenous markers of NSCs and radial glia to show for the first time that radial glia are essential for neurogenesis during development. By using the gfap promoter to drive expression of nuclear localized mCherry we discerned two distinct radial glial-derived cell types: a major nestin+/Sox2+ subtype with strong gfap promoter activity and a minor Sox2+ subtype lacking this activity. Fate mapping studies in this line indicate that gfap+ radial glia generate later-born CoSA interneurons, secondary motorneurons, and oligodendroglia. In another transgenic line using the gfap promoter-driven expression of the nitroreductase enzyme, we induced cell autonomous ablation of gfap+ radial glia and observed a reduction in their specific derived lineages, but not Blbp+ and Sox2+/gfap-negative NSCs, which were retained and expanded at later larval stages. Moreover, we provide evidence supporting classical roles of radial glial in axon patterning, blood-brain barrier formation, and locomotion. Our results suggest that gfap+ radial glia represent the major NSC during late neurogenesis for specific lineages, and possess diverse roles to sustain the structure and function of the spinal cord. These new tools will both corroborate the predicted roles of astroglia and reveal novel roles related to development, physiology, and regeneration in the vertebrate nervous system. GLIA 2016;64:1170-1189.
Collapse
Affiliation(s)
- Kimberly Johnson
- Department of Biological Sciences, Smith College, Northampton, Massachusetts
- Molecular and Cellular Biology Program, University of Massachusetts, Amherst, Massachusetts
| | - Jessica Barragan
- Department of Biological Sciences, Smith College, Northampton, Massachusetts
| | - Sarah Bashiruddin
- Department of Biological Sciences, Smith College, Northampton, Massachusetts
| | - Cody J Smith
- Department of Biology, University of Virginia, Charlottesville, Virginia
| | - Chelsea Tyrrell
- Program in Neuroscience and Behavior, University of Massachusetts, Amherst, Massachusetts
| | - Michael J Parsons
- Department of Surgery, Johns Hopkins University, Baltimore, Maryland
| | - Rosemarie Doris
- Department of Biology, Wesleyan University, Middletown, Connecticut
| | - Sarah Kucenas
- Department of Biology, University of Virginia, Charlottesville, Virginia
| | - Gerald B Downes
- Department of Biology, University of Massachusetts, Amherst, Massachusetts
| | - Carla M Velez
- Department of Biological Sciences, Smith College, Northampton, Massachusetts
| | - Caitlin Schneider
- Department of Biological Sciences, Smith College, Northampton, Massachusetts
| | - Catalina Sakai
- Department of Biological Sciences, Smith College, Northampton, Massachusetts
| | - Narendra Pathak
- Department of Biological Sciences, Smith College, Northampton, Massachusetts
| | - Katrina Anderson
- Department of Biological Sciences, Smith College, Northampton, Massachusetts
| | - Rachael Stein
- Department of Biological Sciences, Smith College, Northampton, Massachusetts
| | - Stephen H Devoto
- Department of Biology, Wesleyan University, Middletown, Connecticut
| | - Jeff S Mumm
- Wilmer Eye Institute, Johns Hopkins University, Baltimore, Maryland
| | - Michael J F Barresi
- Department of Biological Sciences, Smith College, Northampton, Massachusetts
- Molecular and Cellular Biology Program, University of Massachusetts, Amherst, Massachusetts
- Program in Neuroscience and Behavior, University of Massachusetts, Amherst, Massachusetts
| |
Collapse
|