1
|
Massoz L, Bergemann D, Lavergne A, Reynders C, Désiront C, Goossens C, Flasse L, Peers B, Voz MM, Manfroid I. Negative cell cycle regulation by calcineurin is necessary for proper beta cell regeneration in zebrafish. eLife 2024; 12:RP88813. [PMID: 39383064 PMCID: PMC11464004 DOI: 10.7554/elife.88813] [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: 10/11/2024] Open
Abstract
Stimulation of pancreatic beta cell regeneration could be a therapeutic lead to treat diabetes. Unlike humans, the zebrafish can efficiently regenerate beta cells, notably from ductal pancreatic progenitors. To gain insight into the molecular pathways involved in this process, we established the transcriptomic profile of the ductal cells after beta cell ablation in the adult zebrafish. These data highlighted the protein phosphatase calcineurin (CaN) as a new potential modulator of beta cell regeneration. We showed that CaN overexpression abolished the regenerative response, leading to glycemia dysregulation. On the opposite, CaN inhibition increased ductal cell proliferation and subsequent beta cell regeneration. Interestingly, the enhanced proliferation of the progenitors was paradoxically coupled with their exhaustion. This suggests that the proliferating progenitors are next entering in differentiation. CaN appears as a guardian which prevents an excessive progenitor proliferation to preserve the pool of progenitors. Altogether, our findings reveal CaN as a key player in the balance between proliferation and differentiation to enable a proper beta cell regeneration.
Collapse
Affiliation(s)
- Laura Massoz
- Zebrafish Development and Disease Models Laboratory, GIGA-Stem Cells, University of LiègeLiègeBelgium
| | - David Bergemann
- Zebrafish Development and Disease Models Laboratory, GIGA-Stem Cells, University of LiègeLiègeBelgium
| | - Arnaud Lavergne
- Zebrafish Development and Disease Models Laboratory, GIGA-Stem Cells, University of LiègeLiègeBelgium
- GIGA-Genomics Core Facility, GIGA, University of LiègLiègeBelgium
| | - Célia Reynders
- Zebrafish Development and Disease Models Laboratory, GIGA-Stem Cells, University of LiègeLiègeBelgium
| | - Caroline Désiront
- Zebrafish Development and Disease Models Laboratory, GIGA-Stem Cells, University of LiègeLiègeBelgium
| | - Chiara Goossens
- Zebrafish Development and Disease Models Laboratory, GIGA-Stem Cells, University of LiègeLiègeBelgium
| | - Lydie Flasse
- Zebrafish Development and Disease Models Laboratory, GIGA-Stem Cells, University of LiègeLiègeBelgium
| | - Bernard Peers
- Zebrafish Development and Disease Models Laboratory, GIGA-Stem Cells, University of LiègeLiègeBelgium
| | - Marianne M Voz
- Zebrafish Development and Disease Models Laboratory, GIGA-Stem Cells, University of LiègeLiègeBelgium
| | - Isabelle Manfroid
- Zebrafish Development and Disease Models Laboratory, GIGA-Stem Cells, University of LiègeLiègeBelgium
| |
Collapse
|
2
|
Gusmão LA, Köster RW, Tedesco AC. Protective effect of nanoemulsions containing CdTe quantum dots with potential application as a diagnostic agent. Mikrochim Acta 2024; 191:610. [PMID: 39302532 DOI: 10.1007/s00604-024-06690-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Accepted: 09/05/2024] [Indexed: 09/22/2024]
Abstract
A nanoemulsion containing CdTe quantum dots (NE-CdTe-QD) was developed to shield cells from cadmium toxicity and shown to be a promising candidate for brain tumor diagnosis. CdTe-QD was characterized by X-ray diffraction (XRD), Fourier transform infrared spectroscopy (FTIR), and Raman spectroscopy. CdTe-QD exhibited high luminescence emission at 700 nm, and their stability was maintained when encapsulated in lipidic/polymeric nanoemulsions (198 ± 2.0 nm; PDI = 0.174; - 49.0 mV). The biological effects of free and nanoemulsified CdTe-QD were tested in normal cells (NHF) and glioblastoma cell lines (U87-MG and T98G). Membrane colocalization of NE-CdTe-QD by T98G cells was observed. Instead, intracellular endoplasmic reticulum localization of NE-CdTe-QD was verified in U87-MG cells. Cell viability was reduced only when NE-CdTe-QD permeated the membrane of GBM cells, as observed in U87-MG cells, whereas no cytotoxic effects were observed in normal fibroblasts. Incorporating quantum dots directly into the brain cells is difficult. However, the nanoemulsions reduced the toxicity of CdTe-QD in zebrafish larvae and increased their circulation time, and direct injection into the zebrafish brain did not affect neural cell viability. This validates the potential application of these nanomaterials as diagnostic agents and satisfies the necessary criteria for their use as photosensitizers in photodynamic therapy.
Collapse
Affiliation(s)
- Luiza Araújo Gusmão
- Department of Chemistry, Center of Nanotechnology and Tissue Engineering, Photobiology and Photomedicine Research Group, Faculty of Philosophy, Sciences and Letters of Ribeirão Preto, University of São Paulo, São Paulo, Brazil
| | - Reinhard Wolfgang Köster
- Zoological Institute, Cellular and Molecular Neurobiology, Technische Universität Braunschweig, Brunswick, Germany
| | - Antonio Claudio Tedesco
- Department of Chemistry, Center of Nanotechnology and Tissue Engineering, Photobiology and Photomedicine Research Group, Faculty of Philosophy, Sciences and Letters of Ribeirão Preto, University of São Paulo, São Paulo, Brazil.
| |
Collapse
|
3
|
Pose-Méndez S, Rehbock M, Wolf-Asseburg A, Köster RW. In Vivo Monitoring of Fabp7 Expression in Transgenic Zebrafish. Cells 2024; 13:1138. [PMID: 38994990 PMCID: PMC11240397 DOI: 10.3390/cells13131138] [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: 02/13/2024] [Revised: 06/20/2024] [Accepted: 06/29/2024] [Indexed: 07/13/2024] Open
Abstract
In zebrafish, like in mammals, radial glial cells (RGCs) can act as neural progenitors during development and regeneration in adults. However, the heterogeneity of glia subpopulations entails the need for different specific markers of zebrafish glia. Currently, fluorescent protein expression mediated by a regulatory element from the glial fibrillary acidic protein (gfap) gene is used as a prominent glia reporter. We now expand this tool by demonstrating that a regulatory element from the mouse Fatty acid binding protein 7 (Fabp7) gene drives reliable expression in fabp7-expressing zebrafish glial cells. By using three different Fabp7 regulatory element-mediated fluorescent protein reporter strains, we reveal in double transgenic zebrafish that progenitor cells expressing fluorescent proteins driven by the Fabp7 regulatory element give rise to radial glia, oligodendrocyte progenitors, and some neuronal precursors. Furthermore, Bergmann glia represent the almost only glial population of the zebrafish cerebellum (besides a few oligodendrocytes), and the radial glia also remain in the mature cerebellum. Fabp7 regulatory element-mediated reporter protein expression in Bergmann glia progenitors suggests their origin from the ventral cerebellar proliferation zone, the ventricular zone, but not from the dorsally positioned upper rhombic lip. These new Fabp7 reporters will be valuable for functional studies during development and regeneration.
Collapse
Affiliation(s)
- Sol Pose-Méndez
- Cellular and Molecular Neurobiology, Zoological Institut, Technische Universität Braunschweig, 38106 Braunschweig, Germany
| | - Michel Rehbock
- Cellular and Molecular Neurobiology, Zoological Institut, Technische Universität Braunschweig, 38106 Braunschweig, Germany
| | - Alexandra Wolf-Asseburg
- Cellular and Molecular Neurobiology, Zoological Institut, Technische Universität Braunschweig, 38106 Braunschweig, Germany
| | - Reinhard W Köster
- Cellular and Molecular Neurobiology, Zoological Institut, Technische Universität Braunschweig, 38106 Braunschweig, Germany
| |
Collapse
|
4
|
Kalvaitytė M, Gabrilavičiūtė S, Balciunas D. Rapid generation of single-insertion transgenics by Tol2 transposition in zebrafish. Dev Dyn 2024. [PMID: 38946125 DOI: 10.1002/dvdy.719] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 05/06/2024] [Accepted: 05/14/2024] [Indexed: 07/02/2024] Open
Abstract
BACKGROUND The Tol2 transposable element is the most widely used transgenesis tool in zebrafish. However, its high activity almost always leads to multiple unlinked integrations of the transgenic cassette in F1 fish. Each of these transgenes is susceptible to positional effects from the surrounding regulatory landscape, which can lead to altered expression and, consequently, activity. Scientists therefore must strike a balance between the need to maximize reproducibility by establishing single-insertion transgenic lines and the need to complete experiments within a reasonable timeframe. RESULTS In this article, we introduce a simple competitive dilution strategy for rapid generation of single-insertion transgenics. By using cry:BFP reporter plasmid as a competitor, we achieved a nearly fourfold reduction in the number of the transgene of interest integrations while simultaneously increasing the proportion of single-insertion F1 generation transgenics to over 50%. We also observed variations in transgene of interest expression among independent single-insertion transgenics, highlighting that the commonly used ubiquitous ubb promoter is susceptible to position effects. CONCLUSIONS Wide application of our competitive dilution strategy will save time, reduce animal usage, and improve reproducibility of zebrafish research.
Collapse
Affiliation(s)
- Miglė Kalvaitytė
- Institute of Biotechnology, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| | - Sofija Gabrilavičiūtė
- Institute of Biotechnology, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| | - Darius Balciunas
- Institute of Biotechnology, Life Sciences Center, Vilnius University, Vilnius, Lithuania
- Department of Biology, Temple University, Philadelphia, Pennsylvania, USA
| |
Collapse
|
5
|
Orlova SY, Ruzina MN, Emelianova OR, Sergeev AA, Chikurova EA, Orlov AM, Mugue NS. In Search of a Target Gene for a Desirable Phenotype in Aquaculture: Genome Editing of Cyprinidae and Salmonidae Species. Genes (Basel) 2024; 15:726. [PMID: 38927661 PMCID: PMC11202958 DOI: 10.3390/genes15060726] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 05/28/2024] [Accepted: 05/29/2024] [Indexed: 06/28/2024] Open
Abstract
Aquaculture supplies the world food market with a significant amount of valuable protein. Highly productive aquaculture fishes can be derived by utilizing genome-editing methods, and the main problem is to choose a target gene to obtain the desirable phenotype. This paper presents a review of the studies of genome editing for genes controlling body development, growth, pigmentation and sex determination in five key aquaculture Salmonidae and Cyprinidae species, such as rainbow trout (Onchorhynchus mykiss), Atlantic salmon (Salmo salar), common carp (Cyprinus carpio), goldfish (Carassius auratus), Gibel carp (Carassius gibelio) and the model fish zebrafish (Danio rerio). Among the genes studied, the most applicable for aquaculture are mstnba, pomc, and acvr2, the knockout of which leads to enhanced muscle growth; runx2b, mutants of which do not form bones in myoseptae; lepr, whose lack of function makes fish fast-growing; fads2, Δ6abc/5Mt, and Δ6bcMt, affecting the composition of fatty acids in fish meat; dnd mettl3, and wnt4a, mutants of which are sterile; and disease-susceptibility genes prmt7, gab3, gcJAM-A, and cxcr3.2. Schemes for obtaining common carp populations consisting of only large females are promising for use in aquaculture. The immobilized and uncolored zebrafish line is of interest for laboratory use.
Collapse
Affiliation(s)
- Svetlana Yu. Orlova
- Laboratory of Molecular Genetics, Russian Federal Research Institute of Fisheries and Oceanography, 105187 Moscow, Russia; (S.Y.O.)
| | - Maria N. Ruzina
- Laboratory of Molecular Genetics, Russian Federal Research Institute of Fisheries and Oceanography, 105187 Moscow, Russia; (S.Y.O.)
| | - Olga R. Emelianova
- Laboratory of Molecular Genetics, Russian Federal Research Institute of Fisheries and Oceanography, 105187 Moscow, Russia; (S.Y.O.)
- Department of Biological Evolution, Faculty of Biology, Lomonosov Moscow State University, 119234 Moscow, Russia
| | - Alexey A. Sergeev
- Laboratory of Molecular Genetics, Russian Federal Research Institute of Fisheries and Oceanography, 105187 Moscow, Russia; (S.Y.O.)
| | - Evgeniya A. Chikurova
- Laboratory of Molecular Genetics, Russian Federal Research Institute of Fisheries and Oceanography, 105187 Moscow, Russia; (S.Y.O.)
| | - Alexei M. Orlov
- Laboratory of Oceanic Ichthyofauna, Shirshov Institute of Oceanology, Russian Academy of Sciences, 117218 Moscow, Russia
- Laboratory of Behavior of Lower Vertebrates, Severtsov Institute of Ecology and Evolution, Russian Academy of Sciences, 119071 Moscow, Russia
- Department of Ichthyology, Dagestan State University, 367000 Makhachkala, Russia
| | - Nikolai S. Mugue
- Laboratory of Molecular Genetics, Russian Federal Research Institute of Fisheries and Oceanography, 105187 Moscow, Russia; (S.Y.O.)
- Laboratory of Genome Evolution and Speciation, Institute of Developmental Biology Russian Academy of Sciences, 117808 Moscow, Russia
| |
Collapse
|
6
|
Palieri V, Paoli E, Wu YK, Haesemeyer M, Grunwald Kadow IC, Portugues R. The preoptic area and dorsal habenula jointly support homeostatic navigation in larval zebrafish. Curr Biol 2024; 34:489-504.e7. [PMID: 38211586 PMCID: PMC10849091 DOI: 10.1016/j.cub.2023.12.030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 11/22/2023] [Accepted: 12/11/2023] [Indexed: 01/13/2024]
Abstract
Animals must maintain physiological processes within an optimal temperature range despite changes in their environment. Through behavioral assays, whole-brain functional imaging, and neural ablations, we show that larval zebrafish, an ectothermic vertebrate, achieves thermoregulation through homeostatic navigation-non-directional and directional movements toward the temperature closest to its physiological setpoint. A brain-wide circuit encompassing several brain regions enables this behavior. We identified the preoptic area of the hypothalamus (PoA) as a key brain structure in triggering non-directional reorientation when thermal conditions are worsening. This result shows an evolutionary conserved role of the PoA as principal thermoregulator of the brain also in ectotherms. We further show that the habenula (Hb)-interpeduncular nucleus (IPN) circuit retains a short-term memory of the sensory history to support the generation of coherent directed movements even in the absence of continuous sensory cues. We finally provide evidence that this circuit may not be exclusive for temperature but may convey a more abstract representation of relative valence of physiologically meaningful stimuli regardless of their specific identity to enable homeostatic navigation.
Collapse
Affiliation(s)
- Virginia Palieri
- Institute of Neuroscience, Technical University of Munich, Biedersteiner Strasse 29, 80802 Munich, Germany; School of Life Sciences, Technical University of Munich, Freising, Germany
| | - Emanuele Paoli
- Institute of Neuroscience, Technical University of Munich, Biedersteiner Strasse 29, 80802 Munich, Germany
| | - You Kure Wu
- Institute of Neuroscience, Technical University of Munich, Biedersteiner Strasse 29, 80802 Munich, Germany
| | - Martin Haesemeyer
- Department of Neuroscience, The Ohio State University College of Medicine, Columbus, OH 43210, USA
| | - Ilona C Grunwald Kadow
- School of Life Sciences, Technical University of Munich, Freising, Germany; Institute of Physiology II, University of Bonn, Medical Faculty (UKB), Nussallee 11, 53115 Bonn, Germany.
| | - Ruben Portugues
- Institute of Neuroscience, Technical University of Munich, Biedersteiner Strasse 29, 80802 Munich, Germany; Munich Cluster of Systems Neurology (SyNergy), Feodor-Lynen-Str. 17, 81377 Munich, Germany.
| |
Collapse
|
7
|
He J, Blazeski A, Nilanthi U, Menéndez J, Pirani SC, Levic DS, Bagnat M, Singh MK, Raya JG, García-Cardeña G, Torres-Vázquez J. Plxnd1-mediated mechanosensing of blood flow controls the caliber of the Dorsal Aorta via the transcription factor Klf2. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.24.576555. [PMID: 38328196 PMCID: PMC10849625 DOI: 10.1101/2024.01.24.576555] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/09/2024]
Abstract
The cardiovascular system generates and responds to mechanical forces. The heartbeat pumps blood through a network of vascular tubes, which adjust their caliber in response to the hemodynamic environment. However, how endothelial cells in the developing vascular system integrate inputs from circulatory forces into signaling pathways to define vessel caliber is poorly understood. Using vertebrate embryos and in vitro-assembled microvascular networks of human endothelial cells as models, flow and genetic manipulations, and custom software, we reveal that Plexin-D1, an endothelial Semaphorin receptor critical for angiogenic guidance, employs its mechanosensing activity to serve as a crucial positive regulator of the Dorsal Aorta's (DA) caliber. We also uncover that the flow-responsive transcription factor KLF2 acts as a paramount mechanosensitive effector of Plexin-D1 that enlarges endothelial cells to widen the vessel. These findings illuminate the molecular and cellular mechanisms orchestrating the interplay between cardiovascular development and hemodynamic forces.
Collapse
Affiliation(s)
- Jia He
- Department of Cell Biology, NYU Grossman School of Medicine, New York, NY 10016, USA
| | - Adriana Blazeski
- Center for Excellence in Vascular Biology, Department of Pathology, Brigham and Women’s Hospital, Boston, MA, USA and Harvard Medical School, Boston, MA, USA
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Uthayanan Nilanthi
- Programme in Cardiovascular and Metabolic Disorders, Duke-NUS Medical School, 8 College Road, Singapore, 169857
| | - Javier Menéndez
- Department of Cell Biology, NYU Grossman School of Medicine, New York, NY 10016, USA
| | - Samuel C. Pirani
- Department of Cell Biology, NYU Grossman School of Medicine, New York, NY 10016, USA
| | - Daniel S. Levic
- Department of Cell Biology, Duke University, Durham, NC 27710, USA
| | - Michel Bagnat
- Department of Cell Biology, Duke University, Durham, NC 27710, USA
| | - Manvendra K. Singh
- Programme in Cardiovascular and Metabolic Disorders, Duke-NUS Medical School, 8 College Road, Singapore, 169857
- National Heart Research Institute Singapore, National Heart Centre Singapore, 5 Hospital Drive, Singapore, 169609
| | - José G Raya
- Department of Radiology, New York University School of Medicine, New York, NY 10016, USA
| | - Guillermo García-Cardeña
- Center for Excellence in Vascular Biology, Department of Pathology, Brigham and Women’s Hospital, Boston, MA, USA and Harvard Medical School, Boston, MA, USA
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Jesús Torres-Vázquez
- Department of Cell Biology, NYU Grossman School of Medicine, New York, NY 10016, USA
| |
Collapse
|
8
|
Mulligan TS, Mumm JS. Selective Cell Ablation Using an Improved Prodrug-Converting Nitroreductase. Methods Mol Biol 2024; 2707:223-234. [PMID: 37668916 DOI: 10.1007/978-1-0716-3401-1_15] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/06/2023]
Abstract
Selective cell ablation is an invaluable tool to investigate the function of cell types, the regeneration of cells, and the modeling of diseases associated with cell loss. The nitroreductase (NTR)-mediated cell ablation system is a simple method enabling the elimination of targeted cells through the expression of a nitroreductase enzyme and the application of a prodrug (such as metronidazole). The prodrug is reduced to a cytotoxic product by nitroreductase, thereby leading to DNA damage-induced cell death. In species with elevated regenerative capacity such as zebrafish, removing the prodrug allows endogenous tissue to replace the lost cells. Herein, we describe a method for the use of a markedly improved nitroreductase enzyme for spatially and temporally controlled targeted cell ablation in the zebrafish. Recently, we identified an NTR variant (NTR 2.0) that achieves effective targeted cell ablation at concentrations of metronidazole well below those causing toxic side effects. NTR 2.0 thereby enables the ablation of "resistant" cell types and novel cell ablation paradigms. These advances simplify investigations of cell function, enable interrogations of the effects of chronic inflammation on regenerative processes and facilitate modeling of degenerative diseases associated with chronic cell loss. Techniques for transgenic nitroreductase expression and prodrug application are discussed.
Collapse
Affiliation(s)
- Timothy S Mulligan
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University, Baltimore, MD, USA
| | - Jeff S Mumm
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University, Baltimore, MD, USA.
| |
Collapse
|
9
|
Burton AH, Jiao B, Bai Q, Van Laar VS, Wheeler TB, Watkins SC, Bruchez MP, Burton EA. Full-field exposure of larval zebrafish to narrow waveband LED light sources at defined power and energy for optogenetic applications. J Neurosci Methods 2024; 401:110001. [PMID: 37914002 PMCID: PMC10843659 DOI: 10.1016/j.jneumeth.2023.110001] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 10/15/2023] [Accepted: 10/27/2023] [Indexed: 11/03/2023]
Abstract
BACKGROUND Optogenetic approaches in transparent zebrafish models have provided numerous insights into vertebrate neurobiology. The purpose of this study was to develop methods to activate light-sensitive transgene products simultaneously throughout an entire larval zebrafish. NEW METHOD We developed a LED illumination stand and microcontroller unit to expose zebrafish larvae reproducibly to full field illumination at defined wavelength, power, and energy. RESULTS The LED stand generated a sufficiently flat illumination field to expose multiple larval zebrafish to high power light stimuli uniformly, while avoiding sample bath warming. The controller unit allowed precise automated delivery of predetermined amounts of light energy at calibrated power. We demonstrated the utility of the approach by driving photoconversion of Kaede (398 nm), photodimerization of GAVPO (450 nm), and photoactivation of dL5**/MG2I (661 nm) in neurons throughout the CNS of larval zebrafish. Observed outcomes were influenced by both total light energy and its rate of delivery, highlighting the importance of controlling these variables to obtain reproducible results. COMPARISON WITH EXISTING METHODS Our approach employs inexpensive LED chip arrays to deliver narrow-waveband light with a sufficiently flat illumination field to span multiple larval zebrafish simultaneously. Calibration of light power and energy are built into the workflow. CONCLUSIONS The LED illuminator and controller can be constructed from widely available materials using the drawings, instructions, and software provided. This approach will be useful for multiple optogenetic applications in zebrafish and other models.
Collapse
Affiliation(s)
- Alexander H Burton
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA, USA; Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh, Pittsburgh, PA, USA; Undergraduate Program in Chemical and Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA, USA
| | - Binxuan Jiao
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA, USA; Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh, Pittsburgh, PA, USA; Tsinghua University Medical School, Beijing, China
| | - Qing Bai
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA, USA; Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh, Pittsburgh, PA, USA
| | - Victor S Van Laar
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA, USA; Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh, Pittsburgh, PA, USA
| | - Travis B Wheeler
- Department of Cell Biology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Simon C Watkins
- Department of Cell Biology, University of Pittsburgh, Pittsburgh, PA, USA; Center for Biologic Imaging, University of Pittsburgh, Pittsburgh, PA, USA
| | - Marcel P Bruchez
- Department of Chemistry, Carnegie Mellon University, Pittsburgh, PA, USA; Molecular Biosensors and Imaging Center, Carnegie Mellon University, Pittsburgh, PA, USA
| | - Edward A Burton
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA, USA; Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh, Pittsburgh, PA, USA; Geriatric Research Education and Clinical Center, Pittsburgh VA Healthcare System, Pittsburgh, PA, USA.
| |
Collapse
|
10
|
Liu J, Li W, Jin X, Lin F, Han J, Zhang Y. Optimal tagging strategies for illuminating expression profiles of genes with different abundance in zebrafish. Commun Biol 2023; 6:1300. [PMID: 38129658 PMCID: PMC10739737 DOI: 10.1038/s42003-023-05686-1] [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: 01/18/2023] [Accepted: 12/07/2023] [Indexed: 12/23/2023] Open
Abstract
CRISPR-mediated knock-in (KI) technology opens a new era of fluorescent-protein labeling in zebrafish, a preferred model organism for in vivo imaging. We described here an optimized zebrafish gene-tagging strategy, which enables easy and high-efficiency KI, ensures high odds of obtaining seamless KI germlines and is suitable for wide applications. Plasmid donors for 3'-labeling were optimized by shortening the microhomologous arms and by reducing the number and reversing the sequence of the consensus Cas9/sgRNA binding sites. To allow for scar-less KI across the genome, linearized dsDNA donors with 5'-chemical modifications were generated and successfully incorporated into our method. To refine the germline screen workflow and expedite the screen process, we combined fluorescence enrichment and caudal-fin junction-PCR. Furthermore, to trace proteins expressed at a low abundance, we developed a fluorescent signal amplifier using the transcriptional activation strategy. Together, our strategies enable efficient gene-tagging and sensitive expression detection for almost every gene in zebrafish.
Collapse
Affiliation(s)
- Jiannan Liu
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, 361102, Xiamen, Fujian, China
| | - Wenyuan Li
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, 361102, Xiamen, Fujian, China
| | - Xuepu Jin
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, 361102, Xiamen, Fujian, China
| | - Fanjia Lin
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, 361102, Xiamen, Fujian, China
| | - Jiahuai Han
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, 361102, Xiamen, Fujian, China.
- Laboratory Animal Center, Xiamen University, 361102, Xiamen, Fujian, China.
- Research Unit of Cellular Stress of CAMS, Cancer Research Center of Xiamen University, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, 361102, Xiamen, Fujian, China.
| | - Yingying Zhang
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, 361102, Xiamen, Fujian, China.
| |
Collapse
|
11
|
Belmonte-Mateos C, Meister L, Pujades C. Hindbrain rhombomere centers harbor a heterogenous population of dividing progenitors which rely on Notch signaling. Front Cell Dev Biol 2023; 11:1268631. [PMID: 38020924 PMCID: PMC10652760 DOI: 10.3389/fcell.2023.1268631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Accepted: 10/11/2023] [Indexed: 12/01/2023] Open
Abstract
Tissue growth and morphogenesis are interrelated processes, whose tight coordination is essential for the production of different cell fates and the timely precise allocation of stem cell capacities. The zebrafish embryonic brainstem, the hindbrain, exemplifies such coupling between spatiotemporal cell diversity acquisition and tissue growth as the neurogenic commitment is differentially distributed over time. Here, we combined cell lineage and in vivo imaging approaches to reveal the emergence of specific cell population properties within the rhombomeres. We studied the molecular identity of hindbrain rhombomere centers and showed that they harbor different progenitor capacities that change over time. By clonal analysis, we revealed that cells within the center of rhombomeres decrease the proliferative capacity to remain mainly in the G1 phase. Proliferating progenitors give rise to neurons by asymmetric and symmetric neurogenic divisions while maintaining the pool of progenitors. The proliferative capacity of these cells differs from their neighbors, and they are delayed in the onset of Notch activity. Through functional studies, we demonstrated that they rely on Notch3 signaling to be maintained as non-committed progenitors. In this study, we show that cells in rhombomere centers, despite the neurogenic asynchrony, might share steps of a similar program with the rhombomere counterparts, to ensure proper tissue growth.
Collapse
|
12
|
Sahai-Hernandez P, Pouget C, Eyal S, Svoboda O, Chacon J, Grimm L, Gjøen T, Traver D. Dermomyotome-derived endothelial cells migrate to the dorsal aorta to support hematopoietic stem cell emergence. eLife 2023; 12:e58300. [PMID: 37695317 PMCID: PMC10495111 DOI: 10.7554/elife.58300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Accepted: 09/03/2023] [Indexed: 09/12/2023] Open
Abstract
Development of the dorsal aorta is a key step in the establishment of the adult blood-forming system, since hematopoietic stem and progenitor cells (HSPCs) arise from ventral aortic endothelium in all vertebrate animals studied. Work in zebrafish has demonstrated that arterial and venous endothelial precursors arise from distinct subsets of lateral plate mesoderm. Here, we profile the transcriptome of the earliest detectable endothelial cells (ECs) during zebrafish embryogenesis to demonstrate that tissue-specific EC programs initiate much earlier than previously appreciated, by the end of gastrulation. Classic studies in the chick embryo showed that paraxial mesoderm generates a subset of somite-derived endothelial cells (SDECs) that incorporate into the dorsal aorta to replace HSPCs as they exit the aorta and enter circulation. We describe a conserved program in the zebrafish, where a rare population of endothelial precursors delaminates from the dermomyotome to incorporate exclusively into the developing dorsal aorta. Although SDECs lack hematopoietic potential, they act as a local niche to support the emergence of HSPCs from neighboring hemogenic endothelium. Thus, at least three subsets of ECs contribute to the developing dorsal aorta: vascular ECs, hemogenic ECs, and SDECs. Taken together, our findings indicate that the distinct spatial origins of endothelial precursors dictate different cellular potentials within the developing dorsal aorta.
Collapse
Affiliation(s)
- Pankaj Sahai-Hernandez
- Department of Cell and Developmental Biology, University of California, San Diego, La Jolla, United States
| | - Claire Pouget
- Department of Cell and Developmental Biology, University of California, San Diego, La Jolla, United States
| | - Shai Eyal
- Department of Cell and Developmental Biology, University of California, San Diego, La Jolla, United States
| | - Ondrej Svoboda
- Department of Cell and Developmental Biology, University of California, San Diego, La Jolla, United States
- Department of Cell Differentiation, Institute of Molecular Genetics, Academy of Sciences of the Czech Republic v.v.i, Prague, Czech Republic
| | - Jose Chacon
- Department of Cell and Developmental Biology, University of California, San Diego, La Jolla, United States
| | - Lin Grimm
- Department of Cell and Developmental Biology, University of California, San Diego, La Jolla, United States
| | - Tor Gjøen
- Department of Pharmacy, University of Oslo, Oslo, Norway
| | - David Traver
- Department of Cell and Developmental Biology, University of California, San Diego, La Jolla, United States
| |
Collapse
|
13
|
Podobnik M, Singh AP, Fu Z, Dooley CM, Frohnhöfer HG, Firlej M, Stednitz SJ, Elhabashy H, Weyand S, Weir JR, Lu J, Nüsslein-Volhard C, Irion U. kcnj13 regulates pigment cell shapes in zebrafish and has diverged by cis-regulatory evolution between Danio species. Development 2023; 150:dev201627. [PMID: 37530080 PMCID: PMC10482006 DOI: 10.1242/dev.201627] [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: 01/17/2023] [Accepted: 07/21/2023] [Indexed: 08/03/2023]
Abstract
Teleost fish of the genus Danio are excellent models to study the genetic and cellular bases of pigment pattern variation in vertebrates. The two sister species Danio rerio and Danio aesculapii show divergent patterns of horizontal stripes and vertical bars that are partly caused by the divergence of the potassium channel gene kcnj13. Here, we show that kcnj13 is required only in melanophores for interactions with xanthophores and iridophores, which cause location-specific pigment cell shapes and thereby influence colour pattern and contrast in D. rerio. Cis-regulatory rather than protein coding changes underlie kcnj13 divergence between the two Danio species. Our results suggest that homotypic and heterotypic interactions between the pigment cells and their shapes diverged between species by quantitative changes in kcnj13 expression during pigment pattern diversification.
Collapse
Affiliation(s)
- Marco Podobnik
- Max Planck Institute for Biology, 72076 Tübingen, Germany
| | - Ajeet P. Singh
- Chemical Biology and Therapeutics, Novartis Institutes for BioMedical Research, Cambridge, MA 02139, USA
| | - Zhenqiang Fu
- School of Marine Sciences, Sun Yat-sen University, Zhuhai 519082, China
| | - Christopher M. Dooley
- Department of Genetics, Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
| | | | - Magdalena Firlej
- Friedrich Miescher Laboratory of the Max Planck Society, 72076 Tübingen, Germany
| | - Sarah J. Stednitz
- Department of Anatomy & Physiology, University of Melbourne, Victoria, 3010, Melbourne, Australia
| | - Hadeer Elhabashy
- Department of Protein Evolution, Max Planck Institute for Biology, 72076 Tübingen, Germany
- Institute for Bioinformatics and Medical Informatics, University of Tübingen, 72076 Tübingen, Germany
- Department of Computer Science, University of Tübingen, 72076 Tübingen, Germany
| | - Simone Weyand
- Department of Biochemistry, University of Cambridge, Cambridge, CB2 1QW, UK
| | - John R. Weir
- Friedrich Miescher Laboratory of the Max Planck Society, 72076 Tübingen, Germany
| | - Jianguo Lu
- School of Marine Sciences, Sun Yat-sen University, Zhuhai 519082, China
| | | | - Uwe Irion
- Max Planck Institute for Biology, 72076 Tübingen, Germany
| |
Collapse
|
14
|
Du XF, Li FN, Peng XL, Xu B, Zhang Y, Li G, Liu T, Li Y, Wang H, Yan J, Du JL. Circadian regulation of developmental synaptogenesis via the hypocretinergic system. Nat Commun 2023; 14:3195. [PMID: 37268623 DOI: 10.1038/s41467-023-38973-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 05/24/2023] [Indexed: 06/04/2023] Open
Abstract
The circadian clock orchestrates a wide variety of physiological and behavioral processes, enabling animals to adapt to daily environmental changes, particularly the day-night cycle. However, the circadian clock's role in the developmental processes remains unclear. Here, we employ the in vivo long-term time-lapse imaging of retinotectal synapses in the optic tectum of larval zebrafish and reveal that synaptogenesis, a fundamental developmental process for neural circuit formation, exhibits circadian rhythm. This rhythmicity arises primarily from the synapse formation rather than elimination and requires the hypocretinergic neural system. Disruption of this synaptogenic rhythm, by impairing either the circadian clock or the hypocretinergic system, affects the arrangement of the retinotectal synapses on axon arbors and the refinement of the postsynaptic tectal neuron's receptive field. Thus, our findings demonstrate that the developmental synaptogenesis is under hypocretin-dependent circadian regulation, suggesting an important role of the circadian clock in neural development.
Collapse
Affiliation(s)
- Xu-Fei Du
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, 320 Yue-Yang Road, 200031, Shanghai, China.
- University of Chinese Academy of Sciences, 19A Yu-Quan Road, 100049, Beijing, China.
| | - Fu-Ning Li
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, 320 Yue-Yang Road, 200031, Shanghai, China
- University of Chinese Academy of Sciences, 19A Yu-Quan Road, 100049, Beijing, China
| | - Xiao-Lan Peng
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, 320 Yue-Yang Road, 200031, Shanghai, China
| | - Bing Xu
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, 320 Yue-Yang Road, 200031, Shanghai, China
| | - Yu Zhang
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, 320 Yue-Yang Road, 200031, Shanghai, China
| | - Guang Li
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, 320 Yue-Yang Road, 200031, Shanghai, China
| | - Taole Liu
- Center for Circadian Clocks, Soochow University, 215123, Suzhou, Jiangsu, China
- School of Biology and Basic Medical Sciences, Suzhou Medical College, Soochow University, 215123, Suzhou, Jiangsu, China
| | - Ying Li
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, 320 Yue-Yang Road, 200031, Shanghai, China
| | - Han Wang
- Center for Circadian Clocks, Soochow University, 215123, Suzhou, Jiangsu, China
- School of Biology and Basic Medical Sciences, Suzhou Medical College, Soochow University, 215123, Suzhou, Jiangsu, China
| | - Jun Yan
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, 320 Yue-Yang Road, 200031, Shanghai, China
- University of Chinese Academy of Sciences, 19A Yu-Quan Road, 100049, Beijing, China
| | - Jiu-Lin Du
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, 320 Yue-Yang Road, 200031, Shanghai, China.
- University of Chinese Academy of Sciences, 19A Yu-Quan Road, 100049, Beijing, China.
- School of Life Science and Technology, ShanghaiTech University, 319 Yue-Yang Road, 200031, Shanghai, China.
| |
Collapse
|
15
|
Xia J, Liu M, Zhu C, Liu S, Ai L, Ma D, Zhu P, Wang L, Liu F. Activation of lineage competence in hemogenic endothelium precedes the formation of hematopoietic stem cell heterogeneity. Cell Res 2023; 33:448-463. [PMID: 37016019 PMCID: PMC10235423 DOI: 10.1038/s41422-023-00797-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Accepted: 03/01/2023] [Indexed: 04/06/2023] Open
Abstract
Hematopoietic stem and progenitor cells (HSPCs) are considered as a heterogeneous population, but precisely when, where and how HSPC heterogeneity arises remain largely unclear. Here, using a combination of single-cell multi-omics, lineage tracing and functional assays, we show that embryonic HSPCs originate from heterogeneous hemogenic endothelial cells (HECs) during zebrafish embryogenesis. Integrated single-cell transcriptome and chromatin accessibility analysis demonstrates transcriptional heterogeneity and regulatory programs that prime lymphoid/myeloid fates at the HEC level. Importantly, spi2+ HECs give rise to lymphoid/myeloid-primed HSPCs (L/M-HSPCs) and display a stress-responsive function under acute inflammation. Moreover, we uncover that Spi2 is required for the formation of L/M-HSPCs through tightly controlling the endothelial-to-hematopoietic transition program. Finally, single-cell transcriptional comparison of zebrafish and human HECs and human induced pluripotent stem cell-based hematopoietic differentiation results support the evolutionary conservation of L/M-HECs and a conserved role of SPI1 (spi2 homolog in mammals) in humans. These results unveil the lineage origin, biological function and molecular determinant of HSPC heterogeneity and lay the foundation for new strategies for induction of transplantable lineage-primed HSPCs in vitro.
Collapse
Affiliation(s)
- Jun Xia
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Mengyao Liu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Caiying Zhu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Shicheng Liu
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Lanlan Ai
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Dongyuan Ma
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Ping Zhu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Lu Wang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China.
| | - Feng Liu
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China.
- University of Chinese Academy of Sciences, Beijing, China.
- School of Life Sciences, Shandong University, Qingdao, Shandong, China.
| |
Collapse
|
16
|
Kemmler CL, Moran HR, Murray BF, Scoresby A, Klem JR, Eckert RL, Lepovsky E, Bertho S, Nieuwenhuize S, Burger S, D'Agati G, Betz C, Puller AC, Felker A, Ditrychova K, Bötschi S, Affolter M, Rohner N, Lovely CB, Kwan KM, Burger A, Mosimann C. Next-generation plasmids for transgenesis in zebrafish and beyond. Development 2023; 150:dev201531. [PMID: 36975217 PMCID: PMC10263156 DOI: 10.1242/dev.201531] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Accepted: 03/10/2023] [Indexed: 03/29/2023]
Abstract
Transgenesis is an essential technique for any genetic model. Tol2-based transgenesis paired with Gateway-compatible vector collections has transformed zebrafish transgenesis with an accessible modular system. Here, we establish several next-generation transgenesis tools for zebrafish and other species to expand and enhance transgenic applications. To facilitate gene regulatory element testing, we generated Gateway middle entry vectors harboring the small mouse beta-globin minimal promoter coupled to several fluorophores, CreERT2 and Gal4. To extend the color spectrum for transgenic applications, we established middle entry vectors encoding the bright, blue-fluorescent protein mCerulean and mApple as an alternative red fluorophore. We present a series of p2A peptide-based 3' vectors with different fluorophores and subcellular localizations to co-label cells expressing proteins of interest. Finally, we established Tol2 destination vectors carrying the zebrafish exorh promoter driving different fluorophores as a pineal gland-specific transgenesis marker that is active before hatching and through adulthood. exorh-based reporters and transgenesis markers also drive specific pineal gland expression in the eye-less cavefish (Astyanax). Together, our vectors provide versatile reagents for transgenesis applications in zebrafish, cavefish and other models.
Collapse
Affiliation(s)
- Cassie L. Kemmler
- University of Colorado, School of Medicine, Anschutz Medical Campus, Department of Pediatrics, Section of Developmental Biology, 12801 E 17th Avenue, Aurora, CO 80045, USA
| | - Hannah R. Moran
- University of Colorado, School of Medicine, Anschutz Medical Campus, Department of Pediatrics, Section of Developmental Biology, 12801 E 17th Avenue, Aurora, CO 80045, USA
| | - Brooke F. Murray
- Department of Human Genetics, University of Utah, Salt Lake City, UT 84112, USA
| | - Aaron Scoresby
- Department of Human Genetics, University of Utah, Salt Lake City, UT 84112, USA
| | - John R. Klem
- Department of Biochemistry and Molecular Genetics, University of Louisville School of Medicine, Louisville, KY 40202, USA
| | - Rachel L. Eckert
- Department of Biochemistry and Molecular Genetics, University of Louisville School of Medicine, Louisville, KY 40202, USA
| | - Elizabeth Lepovsky
- Department of Biochemistry and Molecular Genetics, University of Louisville School of Medicine, Louisville, KY 40202, USA
| | - Sylvain Bertho
- Stowers Institute for Medical Research, Kansas City, MO 64110, USA
| | - Susan Nieuwenhuize
- University of Colorado, School of Medicine, Anschutz Medical Campus, Department of Pediatrics, Section of Developmental Biology, 12801 E 17th Avenue, Aurora, CO 80045, USA
- Department of Molecular Life Sciences, University of Zurich, 8057 Zürich, Switzerland
| | - Sibylle Burger
- Department of Molecular Life Sciences, University of Zurich, 8057 Zürich, Switzerland
| | - Gianluca D'Agati
- Department of Molecular Life Sciences, University of Zurich, 8057 Zürich, Switzerland
| | - Charles Betz
- Growth & Development, Biozentrum, Spitalstrasse 41, University of Basel, 4056 Basel, Switzerland
| | - Ann-Christin Puller
- Department of Molecular Life Sciences, University of Zurich, 8057 Zürich, Switzerland
| | - Anastasia Felker
- Department of Molecular Life Sciences, University of Zurich, 8057 Zürich, Switzerland
| | - Karolina Ditrychova
- Department of Molecular Life Sciences, University of Zurich, 8057 Zürich, Switzerland
| | - Seraina Bötschi
- Department of Molecular Life Sciences, University of Zurich, 8057 Zürich, Switzerland
| | - Markus Affolter
- Growth & Development, Biozentrum, Spitalstrasse 41, University of Basel, 4056 Basel, Switzerland
| | - Nicolas Rohner
- Stowers Institute for Medical Research, Kansas City, MO 64110, USA
| | - C. Ben Lovely
- Department of Biochemistry and Molecular Genetics, University of Louisville School of Medicine, Louisville, KY 40202, USA
| | - Kristen M. Kwan
- Department of Human Genetics, University of Utah, Salt Lake City, UT 84112, USA
| | - Alexa Burger
- University of Colorado, School of Medicine, Anschutz Medical Campus, Department of Pediatrics, Section of Developmental Biology, 12801 E 17th Avenue, Aurora, CO 80045, USA
| | - Christian Mosimann
- University of Colorado, School of Medicine, Anschutz Medical Campus, Department of Pediatrics, Section of Developmental Biology, 12801 E 17th Avenue, Aurora, CO 80045, USA
| |
Collapse
|
17
|
Coomer C, Naumova D, Talay M, Zolyomi B, Snell N, Sorkac A, Chanchu JM, Cheng J, Roman I, Li J, Robson D, Barnea G, Halpern ME. Transsynaptic labeling and transcriptional control of zebrafish neural circuits. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.03.535421. [PMID: 37066422 PMCID: PMC10103993 DOI: 10.1101/2023.04.03.535421] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/18/2023]
Abstract
Deciphering the connectome, the ensemble of synaptic connections that underlie brain function is a central goal of neuroscience research. The trans-Tango genetic approach, initially developed for anterograde transsynaptic tracing in Drosophila, can be used to map connections between presynaptic and postsynaptic partners and to drive gene expression in target neurons. Here, we describe the successful adaptation of trans-Tango to visualize neural connections in a living vertebrate nervous system, that of the zebrafish. Connections were validated between synaptic partners in the larval retina and brain. Results were corroborated by functional experiments in which optogenetic activation of retinal ganglion cells elicited responses in neurons of the optic tectum, as measured by trans-Tango-dependent expression of a genetically encoded calcium indicator. Transsynaptic signaling through trans-Tango reveals predicted as well as previously undescribed synaptic connections, providing a valuable in vivo tool to monitor and interrogate neural circuits over time.
Collapse
|
18
|
Baraban M, Gordillo Pi C, Bonnet I, Gilles JF, Lejeune C, Cabrera M, Tep F, Breau MA. Actomyosin contractility in olfactory placode neurons opens the skin epithelium to form the zebrafish nostril. Dev Cell 2023; 58:361-375.e5. [PMID: 36841243 PMCID: PMC10023511 DOI: 10.1016/j.devcel.2023.02.001] [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: 07/15/2022] [Revised: 12/07/2022] [Accepted: 02/02/2023] [Indexed: 02/27/2023]
Abstract
Despite their barrier function, epithelia can locally lose their integrity to create physiological openings during morphogenesis. The mechanisms driving the formation of these epithelial breaks are only starting to be investigated. Here, we study the formation of the zebrafish nostril (the olfactory orifice), which opens in the skin epithelium to expose the olfactory neurons to external odorant cues. Combining live imaging, drug treatments, laser ablation, and tissue-specific functional perturbations, we characterize a mechanical interplay between olfactory placode neurons and the skin, which plays a crucial role in the formation of the orifice: the neurons pull on the overlying skin cells in an actomyosin-dependent manner which, in combination with a local reorganization of the skin epithelium, triggers the opening of the orifice. This work identifies an original mechanism to break an epithelial sheet, in which an adjacent group of cells mechanically assists the epithelium to induce its local rupture.
Collapse
Affiliation(s)
- Marion Baraban
- Sorbonne Université, Centre National de la Recherche Scientifique (CNRS), Institut de Biologie Paris-Seine (IBPS), Developmental Biology Laboratory, 75005 Paris, France; Laboratoire Jean Perrin, 75005 Paris, France.
| | - Clara Gordillo Pi
- Sorbonne Université, Centre National de la Recherche Scientifique (CNRS), Institut de Biologie Paris-Seine (IBPS), Developmental Biology Laboratory, 75005 Paris, France
| | - Isabelle Bonnet
- Institut Curie, Université PSL, Sorbonne Université, CNRS UMR168, Laboratoire Physico Chimie Curie, 75005 Paris, France
| | | | - Camille Lejeune
- Sorbonne Université, Centre National de la Recherche Scientifique (CNRS), Institut de Biologie Paris-Seine (IBPS), Developmental Biology Laboratory, 75005 Paris, France
| | - Mélody Cabrera
- Sorbonne Université, Centre National de la Recherche Scientifique (CNRS), Institut de Biologie Paris-Seine (IBPS), Developmental Biology Laboratory, 75005 Paris, France
| | - Florian Tep
- Sorbonne Université, Centre National de la Recherche Scientifique (CNRS), Institut de Biologie Paris-Seine (IBPS), Developmental Biology Laboratory, 75005 Paris, France
| | - Marie Anne Breau
- Sorbonne Université, Centre National de la Recherche Scientifique (CNRS), Institut de Biologie Paris-Seine (IBPS), Developmental Biology Laboratory, 75005 Paris, France; Laboratoire Jean Perrin, 75005 Paris, France; Institut National de la Santé et de la Recherche Médicale (INSERM), Paris, France.
| |
Collapse
|
19
|
Burgess HA, Burton EA. A Critical Review of Zebrafish Neurological Disease Models-1. The Premise: Neuroanatomical, Cellular and Genetic Homology and Experimental Tractability. OXFORD OPEN NEUROSCIENCE 2023; 2:kvac018. [PMID: 37649777 PMCID: PMC10464506 DOI: 10.1093/oons/kvac018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/02/2022] [Revised: 11/13/2022] [Indexed: 09/01/2023]
Abstract
The last decade has seen a dramatic rise in the number of genes linked to neurological disorders, necessitating new models to explore underlying mechanisms and to test potential therapies. Over a similar period, many laboratories adopted zebrafish as a tractable model for studying brain development, defining neural circuits and performing chemical screens. Here we discuss strengths and limitations of using the zebrafish system to model neurological disorders. The underlying premise for many disease models is the high degree of homology between human and zebrafish genes, coupled with the conserved vertebrate Bauplan and repertoire of neurochemical signaling molecules. Yet, we caution that important evolutionary divergences often limit the extent to which human symptoms can be modeled meaningfully in zebrafish. We outline advances in genetic technologies that allow human mutations to be reproduced faithfully in zebrafish. Together with methods that visualize the development and function of neuronal pathways at the single cell level, there is now an unprecedented opportunity to understand how disease-associated genetic changes disrupt neural circuits, a level of analysis that is ideally suited to uncovering pathogenic changes in human brain disorders.
Collapse
Affiliation(s)
- Harold A Burgess
- Division of Developmental Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD, 20892, USA
| | - Edward A Burton
- Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh, Pittsburgh, PA, 15260, USA
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA,15260, USA
- Geriatric Research, Education, and Clinical Center, Pittsburgh VA Healthcare System, Pittsburgh, PA, 15240, USA
| |
Collapse
|
20
|
Begeman IJ, Emery B, Kurth A, Kang J. Regeneration and developmental enhancers are differentially compatible with minimal promoters. Dev Biol 2022; 492:47-58. [PMID: 36167150 PMCID: PMC10211259 DOI: 10.1016/j.ydbio.2022.09.007] [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: 07/11/2022] [Revised: 09/16/2022] [Accepted: 09/19/2022] [Indexed: 12/01/2022]
Abstract
Enhancers and promoters are cis-regulatory elements that control gene expression. Enhancers are activated in a cell type-, tissue-, and condition-specific manner to stimulate promoter function and transcription. Zebrafish have emerged as a powerful animal model for examining the activities of enhancers derived from various species through transgenic enhancer assays, in which an enhancer is coupled with a minimal promoter. However, the efficiency of minimal promoters and their compatibility with multiple developmental and regeneration enhancers have not been systematically tested in zebrafish. Thus, we assessed the efficiency of six minimal promoters and comprehensively interrogated the compatibility of the promoters with developmental and regeneration enhancers. We found that the fos minimal promoter and Drosophila synthetic core promoter (DSCP) yielded high rates of leaky expression that may complicate the interpretation of enhancer assays. Notably, the adenovirus E1b promoter, the zebrafish lepb 0.8-kb (P0.8) and lepb 2-kb (P2) promoters, and a new zebrafish synthetic promoter (ZSP) that combines elements of the E1b and P0.8 promoters drove little or no ectopic expression, making them suitable for transgenic assays. We also found significant differences in compatibility among specific combinations of promoters and enhancers, indicating the importance of promoters as key regulatory elements determining the specificity of gene expression. Our study provides guidelines for transgenic enhancer assays in zebrafish to aid in the discovery of functional enhancers regulating development and regeneration.
Collapse
Affiliation(s)
- Ian J Begeman
- Department of Cell and Regenerative Biology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, 53705, USA
| | - Benjamin Emery
- Department of Cell and Regenerative Biology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, 53705, USA
| | - Andrew Kurth
- Department of Cell and Regenerative Biology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, 53705, USA
| | - Junsu Kang
- Department of Cell and Regenerative Biology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, 53705, USA; UW Carbone Cancer Center, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, 53705, USA.
| |
Collapse
|
21
|
Hossainian D, Shao E, Jiao B, Ilin VA, Parris RS, Zhou Y, Bai Q, Burton EA. Quantification of functional recovery in a larval zebrafish model of spinal cord injury. J Neurosci Res 2022; 100:2044-2054. [PMID: 35986577 PMCID: PMC10695274 DOI: 10.1002/jnr.25118] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 07/19/2022] [Accepted: 08/01/2022] [Indexed: 11/12/2023]
Abstract
Human spinal cord injury (SCI) is characterized by permanent loss of damaged axons, resulting in chronic disability. In contrast, zebrafish can regenerate axonal projections following central nervous system injury and re-establish synaptic contacts with distant targets; elucidation of the underlying molecular events is an important goal with translational potential for improving outcomes in SCI patients. We generated transgenic zebrafish with GFP-labeled axons and transected their spinal cords at 10 days post-fertilization. Intravital confocal microscopy revealed robust axonal regeneration following the procedure, with abundant axons bridging the transection site by 48 h post-injury. In order to analyze neurological function in this model, we developed and validated new open-source software to measure zebrafish lateral trunk curvature during propulsive and turning movements at high temporal resolution. Immediately following spinal cord transection, axial movements were dramatically decreased caudal to the lesion site, but preserved rostral to the injury, suggesting the induction of motor paralysis below the transection level. Over the subsequent 96 h, the magnitude of movements caudal to the lesion recovered to baseline, but the rate of change of truncal curvature did not fully recover, suggesting incomplete restoration of caudal strength over this time course. Quantification of both morphological and functional recovery following SCI will be important for the analysis of axonal regeneration and downstream events necessary for restoration of motor function. An extensive array of genetic and pharmacological interventions can be deployed in the larval zebrafish model to investigate the underlying molecular mechanisms.
Collapse
Affiliation(s)
- Darius Hossainian
- Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA
- Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA
| | - Enhua Shao
- Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA
- Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA
- Tsinghua University Medical School, Beijing, China
| | - Binxuan Jiao
- Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA
- Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA
- Tsinghua University Medical School, Beijing, China
| | - Vladimir A. Ilin
- Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA
- Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA
| | - Ritika S. Parris
- Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA
- Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA
| | - Yangzhong Zhou
- Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA
- Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA
- Tsinghua University Medical School, Beijing, China
| | - Qing Bai
- Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA
- Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA
| | - Edward A. Burton
- Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA
- Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA
- Geriatric Research, Education and Clinical Center, Pittsburgh VA Healthcare System, Pittsburgh, PA, 15213, USA
| |
Collapse
|
22
|
Kumari A, Zeng XA, Rahaman A, Farooq MA, Huang Y, Alee M, Yao R, Ali M, Khalifa I, Badr O. Phenotype-based drug screening: An in vivo strategy to classify and identify the chemical compounds modulating zebrafish M-cell regeneration. Front Mol Biosci 2022; 9:984461. [PMID: 36353729 PMCID: PMC9637979 DOI: 10.3389/fmolb.2022.984461] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Accepted: 09/20/2022] [Indexed: 02/03/2023] Open
Abstract
Several disease-modulatory FDA-approved drugs are being used in patients with neurodegenerative diseases. However, information on their toxicity-related profiles is very limited. Therefore, measurement of drug toxicity is essential to increase the knowledge of their side effects. This study aimed to identify compounds that can modulate M-cell regeneration by causing neuro-protection and -toxicity. Here, we developed a simple and efficient in vivo assay using Tg (hsp: Gal4FF62A; UAS: nfsB-mCherry) transgenic zebrafish larvae. Interestingly, via the phenotype-based drug screening approach, we rapidly investigated 1,260 compounds from the United States drug collection and validated these in large numbers, including 14 compounds, that were obstructing this regeneration process. Next, 4 FDA-approved drugs out of 14 compounds were selected as the lead hits for in silico analysis to clarify their binding patterns with PTEN and SOCS3 signaling due to their significant potential in the inhibition of axon regeneration. Molecular docking studies indicated good binding affinity of all 4 drugs with the respective signaling molecules. This may point to PTEN and SOCS3 as the signaling molecules responsible for reducing axon regeneration. Moreover, the acute effect of compounds in reducing M-cell regeneration delineated their toxic effect. In conclusion, our in vivo along with in silico screening strategy will promote the rapid translation of new therapeutics to improve knowledge of the toxicity profile of approved/non-approved drugs efficiently.
Collapse
Affiliation(s)
- Ankita Kumari
- School of Food Science and Engineering, South China University of Technology, Guangzhou, China,Guangdong Key Laboratory of Food Intelligent Manufacturing, Foshan University, Foshan, Guangdong, China,Overseas Expertise Introduction Centre for Discipline Innovation of Food Nutrition and Human Health (111 Centre), Guangzhou, China
| | - Xin-An Zeng
- School of Food Science and Engineering, South China University of Technology, Guangzhou, China,Guangdong Key Laboratory of Food Intelligent Manufacturing, Foshan University, Foshan, Guangdong, China,Overseas Expertise Introduction Centre for Discipline Innovation of Food Nutrition and Human Health (111 Centre), Guangzhou, China,*Correspondence: Xin-An Zeng, ; Abdul Rahaman, ; Ibrahim Khalifa,
| | - Abdul Rahaman
- School of Food Science and Engineering, South China University of Technology, Guangzhou, China,Guangdong Key Laboratory of Food Intelligent Manufacturing, Foshan University, Foshan, Guangdong, China,Overseas Expertise Introduction Centre for Discipline Innovation of Food Nutrition and Human Health (111 Centre), Guangzhou, China,*Correspondence: Xin-An Zeng, ; Abdul Rahaman, ; Ibrahim Khalifa,
| | - Muhammad Adil Farooq
- Department of Food Science and Technology, Khwaja Fareed University of Engineering and Information Technology, Rahimyar Khan, Punjab, Pakistan
| | - Yanyan Huang
- Guangdong Key Laboratory of Food Intelligent Manufacturing, Foshan University, Foshan, Guangdong, China
| | - Mahafooj Alee
- School of Food Science and Engineering, South China University of Technology, Guangzhou, China
| | - Runyu Yao
- School of Food Science and Engineering, South China University of Technology, Guangzhou, China,Overseas Expertise Introduction Centre for Discipline Innovation of Food Nutrition and Human Health (111 Centre), Guangzhou, China
| | - Murtaza Ali
- School of Food Science and Engineering, South China University of Technology, Guangzhou, China,Guangdong Key Laboratory of Food Intelligent Manufacturing, Foshan University, Foshan, Guangdong, China,Overseas Expertise Introduction Centre for Discipline Innovation of Food Nutrition and Human Health (111 Centre), Guangzhou, China
| | - Ibrahim Khalifa
- Department of Food Science and Technology, Khwaja Fareed University of Engineering and Information Technology, Rahimyar Khan, Punjab, Pakistan,Food Technology Department, Faculty of Agriculture, Benha University, Qalyubia, Egypt,*Correspondence: Xin-An Zeng, ; Abdul Rahaman, ; Ibrahim Khalifa,
| | - Omnia Badr
- Department of Food Science and Technology, Khwaja Fareed University of Engineering and Information Technology, Rahimyar Khan, Punjab, Pakistan,Department of Genetics and Genetic Engineering, Faculty of Agriculture, Benha University, Qalyubia, Egypt
| |
Collapse
|
23
|
Flex E, Albadri S, Radio FC, Cecchetti S, Lauri A, Priolo M, Kissopoulos M, Carpentieri G, Fasano G, Venditti M, Magliocca V, Bellacchio E, Welch CL, Colombo PC, Kochav SM, Chang R, Barrick R, Trivisano M, Micalizzi A, Borghi R, Messina E, Mancini C, Pizzi S, De Santis F, Rosello M, Specchio N, Compagnucci C, McWalter K, Chung WK, Del Bene F, Tartaglia M. Dominantly acting KIF5B variants with pleiotropic cellular consequences cause variable clinical phenotypes. Hum Mol Genet 2022; 32:473-488. [PMID: 36018820 PMCID: PMC9851748 DOI: 10.1093/hmg/ddac213] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Revised: 08/05/2022] [Accepted: 08/23/2022] [Indexed: 01/25/2023] Open
Abstract
Kinesins are motor proteins involved in microtubule (MT)-mediated intracellular transport. They contribute to key cellular processes, including intracellular trafficking, organelle dynamics and cell division. Pathogenic variants in kinesin-encoding genes underlie several human diseases characterized by an extremely variable clinical phenotype, ranging from isolated neurodevelopmental/neurodegenerative disorders to syndromic phenotypes belonging to a family of conditions collectively termed as 'ciliopathies.' Among kinesins, kinesin-1 is the most abundant MT motor for transport of cargoes towards the plus end of MTs. Three kinesin-1 heavy chain isoforms exist in mammals. Different from KIF5A and KIF5C, which are specifically expressed in neurons and established to cause neurological diseases when mutated, KIF5B is an ubiquitous protein. Three de novo missense KIF5B variants were recently described in four subjects with a syndromic skeletal disorder characterized by kyphomelic dysplasia, hypotonia and DD/ID. Here, we report three dominantly acting KIF5B variants (p.Asn255del, p.Leu498Pro and p.Leu537Pro) resulting in a clinically wide phenotypic spectrum, ranging from dilated cardiomyopathy with adult-onset ophthalmoplegia and progressive skeletal myopathy to a neurodevelopmental condition characterized by severe hypotonia with or without seizures. In vitro and in vivo analyses provide evidence that the identified disease-associated KIF5B variants disrupt lysosomal, autophagosome and mitochondrial organization, and impact cilium biogenesis. All variants, and one of the previously reported missense changes, were shown to affect multiple developmental processes in zebrafish. These findings document pleiotropic consequences of aberrant KIF5B function on development and cell homeostasis, and expand the phenotypic spectrum resulting from altered kinesin-mediated processes.
Collapse
Affiliation(s)
- Elisabetta Flex
- To whom correspondence should be addressed at: Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy. Tel: +39 06 4990 2866; ; Marco Tartaglia, Genetics and Rare Disease Research Division, Ospedale Pediatrico Bambino Gesù, IRCCS, Viale di San Paolo 15, 00146 Rome, Italy. Tel: +39 06 6859 3742;
| | | | - Francesca Clementina Radio
- Genetics and Rare Diseases Research Division, Ospedale Pediatrico Bambino Gesù, IRCCS, 00146 Rome, Italy
| | - Serena Cecchetti
- Core Facilities, Istituto Superiore di Sanità, 00161 Rome, Italy
| | - Antonella Lauri
- Genetics and Rare Diseases Research Division, Ospedale Pediatrico Bambino Gesù, IRCCS, 00146 Rome, Italy
| | - Manuela Priolo
- UOSD Genetica Medica, Grande Ospedale Metropolitano "Bianchi Melacrino Morelli", 89124 Reggio Calabria, Italy
| | - Marta Kissopoulos
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy
| | - Giovanna Carpentieri
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy,Genetics and Rare Diseases Research Division, Ospedale Pediatrico Bambino Gesù, IRCCS, 00146 Rome, Italy
| | - Giulia Fasano
- Genetics and Rare Diseases Research Division, Ospedale Pediatrico Bambino Gesù, IRCCS, 00146 Rome, Italy
| | - Martina Venditti
- Genetics and Rare Diseases Research Division, Ospedale Pediatrico Bambino Gesù, IRCCS, 00146 Rome, Italy
| | - Valentina Magliocca
- Genetics and Rare Diseases Research Division, Ospedale Pediatrico Bambino Gesù, IRCCS, 00146 Rome, Italy
| | - Emanuele Bellacchio
- Genetics and Rare Diseases Research Division, Ospedale Pediatrico Bambino Gesù, IRCCS, 00146 Rome, Italy
| | - Carrie L Welch
- Department of Pediatrics, Columbia University Irving Medical Center, NY, New York 10032, USA
| | - Paolo C Colombo
- Department of Medicine, Columbia University Irving Medical Center, NY, New York 10032, USA
| | - Stephanie M Kochav
- Department of Medicine, Columbia University Irving Medical Center, NY, New York 10032, USA
| | - Richard Chang
- Division of Metabolic Disorders, Children's Hospital of Orange County (CHOC), CA, Orange 92868, USA
| | - Rebekah Barrick
- Division of Metabolic Disorders, Children's Hospital of Orange County (CHOC), CA, Orange 92868, USA
| | - Marina Trivisano
- Department of Neuroscience, Ospedale Pediatrico Bambino Gesù, IRCCS, 00146 Rome, Italy
| | - Alessia Micalizzi
- Translational Cytogenomics Research Unit, Bambino Gesù Children's Hospital, IRCCS, 00146 Rome, Italy
| | - Rossella Borghi
- Genetics and Rare Diseases Research Division, Ospedale Pediatrico Bambino Gesù, IRCCS, 00146 Rome, Italy
| | - Elena Messina
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy,Genetics and Rare Diseases Research Division, Ospedale Pediatrico Bambino Gesù, IRCCS, 00146 Rome, Italy
| | - Cecilia Mancini
- Genetics and Rare Diseases Research Division, Ospedale Pediatrico Bambino Gesù, IRCCS, 00146 Rome, Italy
| | - Simone Pizzi
- Genetics and Rare Diseases Research Division, Ospedale Pediatrico Bambino Gesù, IRCCS, 00146 Rome, Italy
| | - Flavia De Santis
- Institut Curie, PSL Research University, INSERM U934, CNRS UMR3215 Paris, France
| | - Marion Rosello
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, 17 Rue Moreau, F-75012 Paris, France
| | - Nicola Specchio
- Department of Neuroscience, Ospedale Pediatrico Bambino Gesù, IRCCS, 00146 Rome, Italy
| | - Claudia Compagnucci
- Genetics and Rare Diseases Research Division, Ospedale Pediatrico Bambino Gesù, IRCCS, 00146 Rome, Italy
| | | | - Wendy K Chung
- Department of Pediatrics, Columbia University Irving Medical Center, NY, New York 10032, USA,Department of Medicine, Columbia University Irving Medical Center, NY, New York 10032, USA
| | | | - Marco Tartaglia
- To whom correspondence should be addressed at: Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy. Tel: +39 06 4990 2866; ; Marco Tartaglia, Genetics and Rare Disease Research Division, Ospedale Pediatrico Bambino Gesù, IRCCS, Viale di San Paolo 15, 00146 Rome, Italy. Tel: +39 06 6859 3742;
| |
Collapse
|
24
|
Caviglia S, Unterweger IA, Gasiūnaitė A, Vanoosthuyse AE, Cutrale F, Trinh LA, Fraser SE, Neuhauss SCF, Ober EA. FRaeppli: a multispectral imaging toolbox for cell tracing and dense tissue analysis in zebrafish. Development 2022; 149:276363. [DOI: 10.1242/dev.199615] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Accepted: 07/20/2022] [Indexed: 12/15/2022]
Abstract
ABSTRACT
Visualizing cell shapes and interactions of differentiating cells is instrumental for understanding organ development and repair. Across species, strategies for stochastic multicolour labelling have greatly facilitated in vivo cell tracking and mapping neuronal connectivity. Yet integrating multi-fluorophore information into the context of developing zebrafish tissues is challenging given their cytoplasmic localization and spectral incompatibility with common fluorescent markers. Inspired by Drosophila Raeppli, we developed FRaeppli (Fish-Raeppli) by expressing bright membrane- or nuclear-targeted fluorescent proteins for efficient cell shape analysis and tracking. High spatiotemporal activation flexibility is provided by the Gal4/UAS system together with Cre/lox and/or PhiC31 integrase. The distinct spectra of the FRaeppli fluorescent proteins allow simultaneous imaging with GFP and infrared subcellular reporters or tissue landmarks. We demonstrate the suitability of FRaeppli for live imaging of complex internal organs, such as the liver, and have tailored hyperspectral protocols for time-efficient acquisition. Combining FRaeppli with polarity markers revealed previously unknown canalicular topologies between differentiating hepatocytes, reminiscent of the mammalian liver, suggesting common developmental mechanisms. The multispectral FRaeppli toolbox thus enables the comprehensive analysis of intricate cellular morphologies, topologies and lineages at single-cell resolution in zebrafish.
Collapse
Affiliation(s)
- Sara Caviglia
- University of Copenhagen, NNF Center for Stem Cell Biology (DanStem) 1 , Blegdamsvej 3B, 2200 Copenhagen N , Denmark
- University of Zurich 2 , Department of Molecular and Life Sciences, Winterthurerstrasse 190, 8057 Zürich , Switzerland
| | - Iris A. Unterweger
- University of Copenhagen, NNF Center for Stem Cell Biology (DanStem) 1 , Blegdamsvej 3B, 2200 Copenhagen N , Denmark
| | - Akvilė Gasiūnaitė
- University of Copenhagen, NNF Center for Stem Cell Biology (DanStem) 1 , Blegdamsvej 3B, 2200 Copenhagen N , Denmark
| | - Alexandre E. Vanoosthuyse
- University of Copenhagen, NNF Center for Stem Cell Biology (DanStem) 1 , Blegdamsvej 3B, 2200 Copenhagen N , Denmark
| | - Francesco Cutrale
- Translational Imaging Center, University of Southern California 3 , 1002 West Childs Way, Los Angeles, CA 90089 , USA
- Biomedical Engineering, University of Southern California 4 , 1002 West Childs Way, Los Angeles, CA 90089 , USA
| | - Le A. Trinh
- Translational Imaging Center, University of Southern California 3 , 1002 West Childs Way, Los Angeles, CA 90089 , USA
- University of Southern California 5 Molecular and Computational Biology , , 1002 West Childs Way, Los Angeles, CA 90089 , USA
| | - Scott E. Fraser
- Translational Imaging Center, University of Southern California 3 , 1002 West Childs Way, Los Angeles, CA 90089 , USA
- Biomedical Engineering, University of Southern California 4 , 1002 West Childs Way, Los Angeles, CA 90089 , USA
- University of Southern California 5 Molecular and Computational Biology , , 1002 West Childs Way, Los Angeles, CA 90089 , USA
| | - Stephan C. F. Neuhauss
- University of Zurich 2 , Department of Molecular and Life Sciences, Winterthurerstrasse 190, 8057 Zürich , Switzerland
| | - Elke A. Ober
- University of Copenhagen, NNF Center for Stem Cell Biology (DanStem) 1 , Blegdamsvej 3B, 2200 Copenhagen N , Denmark
| |
Collapse
|
25
|
Dendrite regeneration in the vertebrate spinal cord. Dev Biol 2022; 488:114-119. [PMID: 35644253 PMCID: PMC10046145 DOI: 10.1016/j.ydbio.2022.05.014] [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: 04/12/2022] [Revised: 05/18/2022] [Accepted: 05/22/2022] [Indexed: 11/20/2022]
Abstract
Axon regeneration in response to injury has been documented in many animals over several hundred years. In contrast, how neurons respond to dendrite injury has been examined only in the last decade. So far, dendrite regeneration after injury has been documented in invertebrate model systems, but has not been assayed in a vertebrate. In this study, we use zebrafish motor neurons to track neurons after dendrite injury. We address two major gaps in our knowledge of dendrite regeneration: 1) whether post-synaptic dendrites can regenerate and 2) whether vertebrate dendrites can regenerate. We find that motor neurons survive laser microsurgery to remove one or all dendrites. Outgrowth of new dendrites typically initiated one to three days after injury, and a new, stable dendrite arbor was in place by five days after injury. We conclude that zebrafish motor neurons have the capacity to regenerate a new dendrite arbor.
Collapse
|
26
|
Liu F, Kambakam S, Almeida MP, Ming Z, Welker JM, Wierson WA, Schultz-Rogers LE, Ekker SC, Clark KJ, Essner JJ, McGrail M. Cre/ lox regulated conditional rescue and inactivation with zebrafish UFlip alleles generated by CRISPR-Cas9 targeted integration. eLife 2022; 11:71478. [PMID: 35713402 PMCID: PMC9270027 DOI: 10.7554/elife.71478] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2021] [Accepted: 06/16/2022] [Indexed: 11/13/2022] Open
Abstract
The ability to regulate gene activity spatially and temporally is essential to investigate cell type-specific gene function during development and in postembryonic processes and disease models. The Cre/lox system has been widely used for performing cell and tissue-specific conditional analysis of gene function in zebrafish. However, simple and efficient methods for isolation of stable, Cre/lox regulated zebrafish alleles are lacking. Here we applied our GeneWeld CRISPR-Cas9 targeted integration strategy to generate floxed alleles that provide robust conditional inactivation and rescue. A universal targeting vector, UFlip, with sites for cloning short homology arms flanking a floxed 2A-mRFP gene trap, was integrated into an intron in rbbp4 and rb1. rbbp4off and rb1off integration alleles resulted in strong mRFP expression, >99% reduction of endogenous gene expression, and recapitulated known indel loss of function phenotypes. Introduction of Cre led to stable inversion of the floxed cassette, loss of mRFP expression, and phenotypic rescue. rbbp4on and rb1on integration alleles did not cause phenotypes in combination with a loss of function mutation. Addition of Cre led to conditional inactivation by stable inversion of the cassette, gene trapping and mRFP expression, and the expected mutant phenotype. Neural progenitor Cre drivers were used for conditional inactivation and phenotypic rescue to showcase how this approach can be used in specific cell populations. Together these results validate a simplified approach for efficient isolation of Cre/lox responsive conditional alleles in zebrafish. Our strategy provides a new toolkit for generating genetic mosaics and represents a significant advance in zebrafish genetics.
Collapse
Affiliation(s)
- Fang Liu
- Department of Genetics, Development and Cell Biology, Iowa State University, Ames, United States
| | - Sekhar Kambakam
- Department of Genetics, Development and Cell Biology, Iowa State University, Ames, United States
| | - Maira P Almeida
- Department of Genetics, Development and Cell Biology, Iowa State University, Ames, United States
| | - Zhitao Ming
- Department of Genetics, Development and Cell Biology, Iowa State University, Ames, United States
| | - Jordan M Welker
- Department of Genetics, Development and Cell Biology, Iowa State University, Ames, United States
| | - Wesley A Wierson
- Department of Genetics, Development and Cell Biology, Iowa State University, Ames, United States
| | - Laura E Schultz-Rogers
- Department of Genetics, Development and Cell Biology, Iowa State University, Ames, United States
| | - Stephen C Ekker
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, United States
| | - Karl J Clark
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, United States
| | - Jeffrey J Essner
- Department of Genetics, Development and Cell Biology, Iowa State University, Ames, United States
| | - Maura McGrail
- Department of Genetics, Development and Cell Biology, Iowa State University, Ames, United States
| |
Collapse
|
27
|
Hevia CF, Engel-Pizcueta C, Udina F, Pujades C. The neurogenic fate of the hindbrain boundaries relies on Notch3-dependent asymmetric cell divisions. Cell Rep 2022; 39:110915. [PMID: 35675784 DOI: 10.1016/j.celrep.2022.110915] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 01/16/2022] [Accepted: 05/11/2022] [Indexed: 11/19/2022] Open
Abstract
Elucidating the cellular and molecular mechanisms that regulate the balance between progenitor cell proliferation and neuronal differentiation in the construction of the embryonic brain demands the combination of cell lineage and functional approaches. Here, we generate the comprehensive lineage of hindbrain boundary cells by using a CRISPR-based knockin zebrafish transgenic line that specifically labels the boundaries. We unveil that boundary cells asynchronously engage in neurogenesis undergoing a functional transition from neuroepithelial progenitors to radial glia cells, coinciding with the onset of Notch3 signaling that triggers their asymmetrical cell division. Upon notch3 loss of function, boundary cells lose radial glia properties and symmetrically divide undergoing neuronal differentiation. Finally, we show that the fate of boundary cells is to become neurons, the subtype of which relies on their axial position, suggesting that boundary cells contribute to refine the number and proportion of the distinct neuronal populations.
Collapse
Affiliation(s)
| | | | - Frederic Udina
- Department of Economics and Business, Universitat Pompeu Fabra, 08002 Barcelona, Spain; Data Science Center, Barcelona School of Economics, 08002 Barcelona, Spain
| | - Cristina Pujades
- Department of Medicine and Life Sciences, 08003 Barcelona, Spain.
| |
Collapse
|
28
|
Mazzolini J, Le Clerc S, Morisse G, Coulonges C, Zagury J, Sieger D. Wasl is crucial to maintain microglial core activities during glioblastoma initiation stages. Glia 2022; 70:1027-1051. [PMID: 35194846 PMCID: PMC9306864 DOI: 10.1002/glia.24154] [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: 09/01/2021] [Revised: 01/19/2022] [Accepted: 01/21/2022] [Indexed: 11/28/2022]
Abstract
Microglia actively promotes the growth of high-grade gliomas. Within the glioma microenvironment an amoeboid microglial morphology has been observed, however the underlying causes and the related impact on microglia functions and their tumor promoting activities is unclear. Using the advantages of the larval zebrafish model, we identified the underlying mechanism and show that microglial morphology and functions are already impaired during glioma initiation stages. The presence of pre-neoplastic HRasV12 expressing cells induces an amoeboid morphology of microglia, increases microglial numbers and decreases their motility and phagocytic activity. RNA sequencing analysis revealed lower expression levels of the actin nucleation promoting factor wasla in microglia. Importantly, a microglia specific rescue of wasla expression restores microglial morphology and functions. This results in increased phagocytosis of pre-neoplastic cells and slows down tumor progression. In conclusion, we identified a mechanism that de-activates core microglial functions within the emerging glioma microenvironment. Restoration of this mechanism might provide a way to impair glioma growth.
Collapse
Affiliation(s)
- Julie Mazzolini
- Centre for Discovery Brain SciencesUniversity of EdinburghEdinburghUK
| | - Sigrid Le Clerc
- Laboratoire GBCM, EA7528, Conservatoire National des Arts et MétiersHESAM UniversitéParisFrance
| | - Gregoire Morisse
- Centre for Discovery Brain SciencesUniversity of EdinburghEdinburghUK
| | - Cédric Coulonges
- Laboratoire GBCM, EA7528, Conservatoire National des Arts et MétiersHESAM UniversitéParisFrance
| | - Jean‐François Zagury
- Laboratoire GBCM, EA7528, Conservatoire National des Arts et MétiersHESAM UniversitéParisFrance
| | - Dirk Sieger
- Centre for Discovery Brain SciencesUniversity of EdinburghEdinburghUK
| |
Collapse
|
29
|
Lichtenegger A, Mukherjee P, Zhu L, Morishita R, Tomita K, Oida D, Leskovar K, Abd El-Sadek I, Makita S, Kirchberger S, Distel M, Baumann B, Yasuno Y. Non-destructive characterization of adult zebrafish models using Jones matrix optical coherence tomography. BIOMEDICAL OPTICS EXPRESS 2022; 13:2202-2223. [PMID: 35519284 PMCID: PMC9045912 DOI: 10.1364/boe.455876] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 03/03/2022] [Accepted: 03/03/2022] [Indexed: 06/14/2023]
Abstract
The zebrafish is a valuable vertebrate animal model in pre-clinical cancer research. A Jones matrix optical coherence tomography (JM-OCT) prototype operating at 1310 nm and an intensity-based spectral-domain OCT setup at 840 nm were utilized to investigate adult wildtype and a tumor-developing zebrafish model. Various anatomical features were characterized based on their inherent scattering and polarization signature. A motorized translation stage in combination with the JM-OCT prototype enabled large field-of-view imaging to investigate adult zebrafish in a non-destructive way. The diseased animals exhibited tumor-related abnormalities in the brain and near the eye region. The scatter intensity, the attenuation coefficients and local polarization parameters such as the birefringence and the degree of polarization uniformity were analyzed to quantify differences in tumor versus control regions. The proof-of-concept study in a limited number of animals revealed a significant decrease in birefringence in tumors found in the brain and near the eye compared to control regions. The presented work showed the potential of OCT and JM-OCT as non-destructive, high-resolution, and real-time imaging modalities for pre-clinical research based on zebrafish.
Collapse
Affiliation(s)
- Antonia Lichtenegger
- Center for Medical Physics and Biomedical Engineering, Medical University of Vienna, Austria
- Computational Optics Group, Institute of Applied Physics, University of Tsukuba, Japan
| | - Pradipta Mukherjee
- Computational Optics Group, Institute of Applied Physics, University of Tsukuba, Japan
| | - Lida Zhu
- Computational Optics Group, Institute of Applied Physics, University of Tsukuba, Japan
| | - Rion Morishita
- Computational Optics Group, Institute of Applied Physics, University of Tsukuba, Japan
| | - Kiriko Tomita
- Computational Optics Group, Institute of Applied Physics, University of Tsukuba, Japan
| | - Daisuke Oida
- Computational Optics Group, Institute of Applied Physics, University of Tsukuba, Japan
| | - Konrad Leskovar
- Center for Medical Physics and Biomedical Engineering, Medical University of Vienna, Austria
| | - Ibrahim Abd El-Sadek
- Computational Optics Group, Institute of Applied Physics, University of Tsukuba, Japan
- Department of Physics, Faculty of Science, Damietta University, Egypt
| | - Shuichi Makita
- Computational Optics Group, Institute of Applied Physics, University of Tsukuba, Japan
| | | | - Martin Distel
- St. Anna Children’s Cancer Research Institute (CCRI), Austria
| | - Bernhard Baumann
- Center for Medical Physics and Biomedical Engineering, Medical University of Vienna, Austria
| | - Yoshiaki Yasuno
- Computational Optics Group, Institute of Applied Physics, University of Tsukuba, Japan
| |
Collapse
|
30
|
Wada Y, Tsukatani H, Kuroda C, Miyazaki Y, Otoshi M, Kobayashi I. Jagged 2b induces intercellular signaling within somites to establish hematopoietic stem cell fate in zebrafish. Development 2022; 149:274970. [DOI: 10.1242/dev.200339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Accepted: 03/17/2022] [Indexed: 11/20/2022]
Abstract
ABSTRACT
During development, the somites play a key role in the specification of hematopoietic stem cells (HSCs). In zebrafish, the somitic Notch ligands Delta-c (Dlc) and Dld, both of which are regulated by Wnt16, directly instruct HSC fate in a shared vascular precursor. However, it remains unclear how this signaling cascade is spatially and temporally regulated within somites. Here, we show in zebrafish that an additional somitic Notch ligand, Jagged 2b (Jag2b), induces intercellular signaling to drive wnt16 expression. Jag2b activated Notch signaling in segmented somites at the early stage of somitogenesis. Loss of jag2b led to a reduction in the expression of wnt16 in the somites and an HSC marker, runx1, in the dorsal aorta, whereas overexpression of jag2b increased both. However, Notch-activated cells were adjacent to, but did not overlap with, wnt16-expressing cells within the somites, suggesting that an additional signaling molecule mediates this intercellular signal transduction. We uncover that Jag2b-driven Notch signaling induces efna1b expression, which regulates wnt16 expression in neighboring somitic cells. Collectively, we provide evidence for previously unidentified spatiotemporal regulatory mechanisms of HSC specification by somites.
Collapse
Affiliation(s)
- Yukino Wada
- Division of Life Sciences, Graduate School of Natural Science and Technology, Kanazawa University, Ishikawa 920-1192, Japan
| | - Hikaru Tsukatani
- Faculty of Natural System, Institute of Science and Engineering, Kanazawa University, Ishikawa 920-1192, Japan
| | - Chihiro Kuroda
- Faculty of Natural System, Institute of Science and Engineering, Kanazawa University, Ishikawa 920-1192, Japan
| | - Yurika Miyazaki
- Faculty of Natural System, Institute of Science and Engineering, Kanazawa University, Ishikawa 920-1192, Japan
| | - Miku Otoshi
- Faculty of Biological Science and Technology, Institute of Science and Engineering, Kanazawa University, Ishikawa 920-1192, Japan
| | - Isao Kobayashi
- Faculty of Biological Science and Technology, Institute of Science and Engineering, Kanazawa University, Ishikawa 920-1192, Japan
| |
Collapse
|
31
|
Gao L, Meiring JCM, Varady A, Ruider IE, Heise C, Wranik M, Velasco CD, Taylor JA, Terni B, Weinert T, Standfuss J, Cabernard CC, Llobet A, Steinmetz MO, Bausch AR, Distel M, Thorn-Seshold J, Akhmanova A, Thorn-Seshold O. In Vivo Photocontrol of Microtubule Dynamics and Integrity, Migration and Mitosis, by the Potent GFP-Imaging-Compatible Photoswitchable Reagents SBTubA4P and SBTub2M. J Am Chem Soc 2022; 144:5614-5628. [PMID: 35290733 PMCID: PMC8972266 DOI: 10.1021/jacs.2c01020] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Photoswitchable reagents are powerful tools for high-precision studies in cell biology. When these reagents are globally administered yet locally photoactivated in two-dimensional (2D) cell cultures, they can exert micron- and millisecond-scale biological control. This gives them great potential for use in biologically more relevant three-dimensional (3D) models and in vivo, particularly for studying systems with inherent spatiotemporal complexity, such as the cytoskeleton. However, due to a combination of photoswitch isomerization under typical imaging conditions, metabolic liabilities, and insufficient water solubility at effective concentrations, the in vivo potential of photoswitchable reagents addressing cytosolic protein targets remains largely unrealized. Here, we optimized the potency and solubility of metabolically stable, druglike colchicinoid microtubule inhibitors based on the styrylbenzothiazole (SBT) scaffold that are nonresponsive to typical fluorescent protein imaging wavelengths and so enable multichannel imaging studies. We applied these reagents both to 3D organoids and tissue explants and to classic model organisms (zebrafish, clawed frog) in one- and two-protein imaging experiments, in which spatiotemporally localized illuminations allowed them to photocontrol microtubule dynamics, network architecture, and microtubule-dependent processes in vivo with cellular precision and second-level resolution. These nanomolar, in vivo capable photoswitchable reagents should open up new dimensions for high-precision cytoskeleton research in cargo transport, cell motility, cell division, and development. More broadly, their design can also inspire similarly capable optical reagents for a range of cytosolic protein targets, thus bringing in vivo photopharmacology one step closer to general realization.
Collapse
Affiliation(s)
- Li Gao
- Department of Pharmacy, Ludwig-Maximilians University of Munich, Munich 81377, Germany
| | - Joyce C M Meiring
- Cell Biology, Neurobiology and Biophysics, Department of Biology, Faculty of Science, Utrecht University, Utrecht CH 3584, Netherlands
| | - Adam Varady
- St. Anna Children's Cancer Research Institute (CCRI), Vienna 1090, Austria
| | - Iris E Ruider
- Physics Department and Center for Protein Assemblies CPA, Technical University of Munich, Garching 85747, Germany
| | - Constanze Heise
- Department of Pharmacy, Ludwig-Maximilians University of Munich, Munich 81377, Germany
| | - Maximilian Wranik
- Laboratory of Biomolecular Research, Division of Biology and Chemistry, Paul Scherrer Institut, Villigen 5232, Switzerland
| | - Cecilia D Velasco
- Laboratory of Neurobiology, Department of Pathology and Experimental Therapy, Institute of Neurosciences, University of Barcelona, L'Hospitalet de Llobregat, Barcelona 08907, Spain
- Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat, Barcelona 08907, Spain
| | - Jennifer A Taylor
- Department of Biology, University of Washington, Seattle, Washington 98195, United States
| | - Beatrice Terni
- Laboratory of Neurobiology, Department of Pathology and Experimental Therapy, Institute of Neurosciences, University of Barcelona, L'Hospitalet de Llobregat, Barcelona 08907, Spain
- Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat, Barcelona 08907, Spain
| | - Tobias Weinert
- Laboratory of Biomolecular Research, Division of Biology and Chemistry, Paul Scherrer Institut, Villigen 5232, Switzerland
| | - Jörg Standfuss
- Laboratory of Biomolecular Research, Division of Biology and Chemistry, Paul Scherrer Institut, Villigen 5232, Switzerland
| | - Clemens C Cabernard
- Department of Biology, University of Washington, Seattle, Washington 98195, United States
| | - Artur Llobet
- Laboratory of Neurobiology, Department of Pathology and Experimental Therapy, Institute of Neurosciences, University of Barcelona, L'Hospitalet de Llobregat, Barcelona 08907, Spain
- Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat, Barcelona 08907, Spain
| | - Michel O Steinmetz
- Laboratory of Biomolecular Research, Division of Biology and Chemistry, Paul Scherrer Institut, Villigen 5232, Switzerland
- Biozentrum, University of Basel, Basel 4056, Switzerland
| | - Andreas R Bausch
- Physics Department and Center for Protein Assemblies CPA, Technical University of Munich, Garching 85747, Germany
| | - Martin Distel
- St. Anna Children's Cancer Research Institute (CCRI), Vienna 1090, Austria
- Zebrafish Platform Austria for Preclinical Drug Screening (ZANDR), Vienna 1090, Austria
| | - Julia Thorn-Seshold
- Department of Pharmacy, Ludwig-Maximilians University of Munich, Munich 81377, Germany
| | - Anna Akhmanova
- Cell Biology, Neurobiology and Biophysics, Department of Biology, Faculty of Science, Utrecht University, Utrecht CH 3584, Netherlands
| | - Oliver Thorn-Seshold
- Department of Pharmacy, Ludwig-Maximilians University of Munich, Munich 81377, Germany
| |
Collapse
|
32
|
Sharrock AV, Mulligan TS, Hall KR, Williams EM, White DT, Zhang L, Emmerich K, Matthews F, Nimmagadda S, Washington S, Le KD, Meir-Levi D, Cox OL, Saxena MT, Calof AL, Lopez-Burks ME, Lander AD, Ding D, Ji H, Ackerley DF, Mumm JS. NTR 2.0: a rationally engineered prodrug-converting enzyme with substantially enhanced efficacy for targeted cell ablation. Nat Methods 2022; 19:205-215. [PMID: 35132245 PMCID: PMC8851868 DOI: 10.1038/s41592-021-01364-4] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Accepted: 11/29/2021] [Indexed: 11/12/2022]
Abstract
Transgenic expression of bacterial nitroreductase (NTR) enzymes sensitizes eukaryotic cells to prodrugs such as metronidazole (MTZ), enabling selective cell-ablation paradigms that have expanded studies of cell function and regeneration in vertebrates. However, first-generation NTRs required confoundingly toxic prodrug treatments to achieve effective cell ablation, and some cell types have proven resistant. Here we used rational engineering and cross-species screening to develop an NTR variant, NTR 2.0, which exhibits ~100-fold improvement in MTZ-mediated cell-specific ablation efficacy, eliminating the need for near-toxic prodrug treatment regimens. NTR 2.0 therefore enables sustained cell-loss paradigms and ablation of previously resistant cell types. These properties permit enhanced interrogations of cell function, extended challenges to the regenerative capacities of discrete stem cell niches, and novel modeling of chronic degenerative diseases. Accordingly, we have created a series of bipartite transgenic reporter/effector resources to facilitate dissemination of NTR 2.0 to the research community.
Collapse
Affiliation(s)
- Abigail V Sharrock
- School of Biological Sciences, Victoria University of Wellington, Wellington, New Zealand
| | - Timothy S Mulligan
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University, Baltimore, MD, USA
| | - Kelsi R Hall
- School of Biological Sciences, Victoria University of Wellington, Wellington, New Zealand
| | - Elsie M Williams
- School of Biological Sciences, Victoria University of Wellington, Wellington, New Zealand
| | - David T White
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University, Baltimore, MD, USA
| | - Liyun Zhang
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University, Baltimore, MD, USA
| | - Kevin Emmerich
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University, Baltimore, MD, USA
| | - Frazer Matthews
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University, Baltimore, MD, USA
| | - Saumya Nimmagadda
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University, Baltimore, MD, USA
| | - Selena Washington
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University, Baltimore, MD, USA
| | - Katherine D Le
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University, Baltimore, MD, USA
| | - Danielle Meir-Levi
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University, Baltimore, MD, USA
| | - Olivia L Cox
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University, Baltimore, MD, USA
| | - Meera T Saxena
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University, Baltimore, MD, USA
- Luminomics, Baltimore, MD, USA
| | - Anne L Calof
- Department of Anatomy and Neurobiology, University of California, Irvine, CA, USA
- Department of Developmental and Cell Biology, University of California, Irvine, CA, USA
- Center for Complex Biological Systems, University of California, Irvine, CA, USA
| | - Martha E Lopez-Burks
- Department of Developmental and Cell Biology, University of California, Irvine, CA, USA
- Center for Complex Biological Systems, University of California, Irvine, CA, USA
| | - Arthur D Lander
- Department of Developmental and Cell Biology, University of California, Irvine, CA, USA
- Center for Complex Biological Systems, University of California, Irvine, CA, USA
| | - Ding Ding
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Hongkai Ji
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - David F Ackerley
- School of Biological Sciences, Victoria University of Wellington, Wellington, New Zealand.
- Centre for Biodiscovery and Maurice Wilkins Centre for Molecular Biodiscovery, Victoria University of Wellington, Wellington, New Zealand.
| | - Jeff S Mumm
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University, Baltimore, MD, USA.
- The Center for Nanomedicine, Wilmer Eye Institute, Johns Hopkins University, Baltimore, MD, USA.
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University, Baltimore, MD, USA.
- Department of Genetic Medicine, McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University, Baltimore, MD, USA.
| |
Collapse
|
33
|
Ákos Z, Dunipace L, Stathopoulos A. NaNuTrap: a technique for in vivo cell nucleus labelling using nanobodies. Development 2021; 148:dev199822. [PMID: 34328170 PMCID: PMC10656463 DOI: 10.1242/dev.199822] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Accepted: 07/12/2021] [Indexed: 11/20/2022]
Abstract
In vivo cell labelling is challenging in fast developmental processes because many cell types differentiate more quickly than the maturation time of fluorescent proteins, making visualization of these tissues impossible with standard techniques. Here, we present a nanobody-based method, Nanobody Nuclear Trap (NaNuTrap), which works with the existing Gal4/UAS system in Drosophila and allows for early in vivo cell nuclei labelling independently of the maturation time of the fluorescent protein. This restores the utility of fluorescent proteins that have longer maturation times, such as those used in two-photon imaging, for live imaging of fast or very early developmental processes. We also present a more general application of this system, whereby NaNuTrap can convert cytoplasmic GFP expressed in any existing transgenic fly line into a nuclear label. This nuclear re-localization of the fluorescent signal can improve the utility of the GFP label, e.g. in cell counting, as well as resulting in a general increase in intensity of the live fluorescent signal. We demonstrate these capabilities of NaNuTrap by effectively tracking subsets of cells during the fast movements associated with gastrulation.
Collapse
Affiliation(s)
- Zsuzsa Ákos
- California Institute of Technology, 1200 East California Blvd, Pasadena, CA 91125, USA
| | - Leslie Dunipace
- California Institute of Technology, 1200 East California Blvd, Pasadena, CA 91125, USA
| | - Angelike Stathopoulos
- California Institute of Technology, 1200 East California Blvd, Pasadena, CA 91125, USA
| |
Collapse
|
34
|
Choe CP, Choi SY, Kee Y, Kim MJ, Kim SH, Lee Y, Park HC, Ro H. Transgenic fluorescent zebrafish lines that have revolutionized biomedical research. Lab Anim Res 2021; 37:26. [PMID: 34496973 PMCID: PMC8424172 DOI: 10.1186/s42826-021-00103-2] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Accepted: 08/26/2021] [Indexed: 12/22/2022] Open
Abstract
Since its debut in the biomedical research fields in 1981, zebrafish have been used as a vertebrate model organism in more than 40,000 biomedical research studies. Especially useful are zebrafish lines expressing fluorescent proteins in a molecule, intracellular organelle, cell or tissue specific manner because they allow the visualization and tracking of molecules, intracellular organelles, cells or tissues of interest in real time and in vivo. In this review, we summarize representative transgenic fluorescent zebrafish lines that have revolutionized biomedical research on signal transduction, the craniofacial skeletal system, the hematopoietic system, the nervous system, the urogenital system, the digestive system and intracellular organelles.
Collapse
Affiliation(s)
- Chong Pyo Choe
- Division of Life Science, Gyeongsang National University, Jinju, 52828, Republic of Korea.,Division of Applied Life Science, Plant Molecular Biology and Biotechnology Research Center, Gyeongsang National University, Jinju, 52828, Republic of Korea
| | - Seok-Yong Choi
- Department of Biomedical Sciences, Chonnam National University Medical School, Hwasun, 58128, Republic of Korea
| | - Yun Kee
- Division of Biomedical Convergence, College of Biomedical Science, Kangwon National University, Chuncheon, 24341, Republic of Korea.
| | - Min Jung Kim
- Department of Biological Sciences, Sookmyung Women's University, Seoul, 04310, Republic of Korea
| | - Seok-Hyung Kim
- Department of Marine Life Sciences and Fish Vaccine Research Center, Jeju National University, Jeju, 63243, Republic of Korea
| | - Yoonsung Lee
- Center for Genomic Integrity, Institute for Basic Science (IBS), Ulsan, 44919, Republic of Korea
| | - Hae-Chul Park
- Department of Biomedical Sciences, College of Medicine, Korea University, Ansan, 15355, Republic of Korea
| | - Hyunju Ro
- Department of Biological Sciences, College of Bioscience and Biotechnology, Chungnam National University, Daejeon, 34134, Republic of Korea
| |
Collapse
|
35
|
Andreana M, Sturtzel C, Spielvogel CP, Papp L, Leitgeb R, Drexler W, Distel M, Unterhuber A. Toward Quantitative in vivo Label-Free Tracking of Lipid Distribution in a Zebrafish Cancer Model. Front Cell Dev Biol 2021; 9:675636. [PMID: 34277618 PMCID: PMC8280786 DOI: 10.3389/fcell.2021.675636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Accepted: 05/04/2021] [Indexed: 11/26/2022] Open
Abstract
Cancer cells often adapt their lipid metabolism to accommodate the increased fatty acid demand for membrane biogenesis and energy production. Upregulation of fatty acid uptake from the environment of cancer cells has also been reported as an alternative mechanism. To investigate the role of lipids in tumor onset and progression and to identify potential diagnostic biomarkers, lipids are ideally imaged directly within the intact tumor tissue in a label-free way. In this study, we investigated lipid accumulation and distribution in living zebrafish larvae developing a tumor by means of coherent anti-Stokes Raman scattering microscopy. Quantitative textural features based on radiomics revealed higher lipid accumulation in oncogene-expressing larvae compared to healthy ones. This high lipid accumulation could reflect an altered lipid metabolism in the hyperproliferating oncogene-expressing cells.
Collapse
Affiliation(s)
- Marco Andreana
- Center for Medical Physics and Biomedical Engineering, Medical University of Vienna, Vienna, Austria
| | - Caterina Sturtzel
- Innovative Cancer Models, St. Anna Children's Cancer Research Institute, Vienna, Austria.,Zebrafish Platform Austria for Preclinical Drug Screening (ZANDR), Vienna, Austria
| | - Clemens P Spielvogel
- Division of Nuclear Medicine, Department of Medical Imaging and Image-Guided Therapy, Medical University of Vienna, Vienna, Austria.,Christian Doppler Laboratory for Applied Metabolomics, Medical University of Vienna, Vienna, Austria
| | - Laszlo Papp
- Center for Medical Physics and Biomedical Engineering, Medical University of Vienna, Vienna, Austria
| | - Rainer Leitgeb
- Center for Medical Physics and Biomedical Engineering, Medical University of Vienna, Vienna, Austria.,Christian Doppler Laboratory OPTRAMED, Medical University of Vienna, Vienna, Austria
| | - Wolfgang Drexler
- Center for Medical Physics and Biomedical Engineering, Medical University of Vienna, Vienna, Austria
| | - Martin Distel
- Innovative Cancer Models, St. Anna Children's Cancer Research Institute, Vienna, Austria.,Zebrafish Platform Austria for Preclinical Drug Screening (ZANDR), Vienna, Austria
| | - Angelika Unterhuber
- Center for Medical Physics and Biomedical Engineering, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
36
|
Manuel R, Iglesias Gonzalez AB, Habicher J, Koning HK, Boije H. Characterization of Individual Projections Reveal That Neuromasts of the Zebrafish Lateral Line are Innervated by Multiple Inhibitory Efferent Cells. Front Neuroanat 2021; 15:666109. [PMID: 34234651 PMCID: PMC8255702 DOI: 10.3389/fnana.2021.666109] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Accepted: 04/19/2021] [Indexed: 11/13/2022] Open
Abstract
The zebrafish lateral line is a sensory system used to detect changes in water flow. It is comprized of clusters of superficial hair cells called neuromasts. Modulation occurs via excitatory and inhibitory efferent neurons located in the brain. Using mosaic transgenic labeling we provide an anatomical overview of the lateral line projections made by individual inhibitory efferent neurons in 5-day old zebrafish larvae. For each hemisphere we estimate there to be six inhibitory efferent neurons located in two different nuclei. Three distinct cell types were classified based on their projections; to the anterior lateral line around the head, to the posterior lateral line along the body, or to both. Our analyses corroborate previous studies employing back-fills, but our transgenic labeling allowed a more thorough characterization of their morphology. We found that individual inhibitory efferent cells connect to multiple neuromasts and that a single neuromast is connected by multiple inhibitory efferent cells. The efferent axons project to the sensory ganglia and follow the sensory axon tract along the lateral line. Time-lapse imaging revealed that inhibitory efferent axons do not migrate with the primordium as the primary sensory afferent does, but follow with an 8–14 h lag. These data bring new insights into the formation of a sensory circuit and support the hypothesis that different classes of inhibitory efferent cells have different functions. Our findings provide a foundation for future studies focussed toward unraveling how and when sensory perception is modulated by different efferent cells.
Collapse
Affiliation(s)
- Remy Manuel
- Department Neuroscience, Uppsala University, Uppsala, Sweden
| | | | - Judith Habicher
- Department Neuroscience, Uppsala University, Uppsala, Sweden
| | | | - Henrik Boije
- Department Neuroscience, Uppsala University, Uppsala, Sweden
| |
Collapse
|
37
|
Zebrafish Neural Crest: Lessons and Tools to Study In Vivo Cell Migration. Methods Mol Biol 2021. [PMID: 32939715 DOI: 10.1007/978-1-0716-0779-4_9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register]
Abstract
The study of cell migration has been greatly enhanced by the development of new model systems and analysis protocols to study this process in vivo. Zebrafish embryos have been a principal protagonist because they are easily accessible, genetically tractable, and optically transparent. Neural crest cells, on the other hand, are the ideal system to study cell migration. These cells migrate extensively, using different modalities of movement and sharing many traits with metastatic cancer cells. In this chapter, we present new tools and protocols that allow the study of NC development and migration in vivo.
Collapse
|
38
|
Theisen U, Ernst AU, Heyne RLS, Ring TP, Thorn-Seshold O, Köster RW. Microtubules and motor proteins support zebrafish neuronal migration by directing cargo. J Cell Biol 2021; 219:151951. [PMID: 32668451 PMCID: PMC7659711 DOI: 10.1083/jcb.201908040] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Revised: 04/08/2020] [Accepted: 06/18/2020] [Indexed: 11/22/2022] Open
Abstract
Neuronal migration during development is necessary to form an ordered and functional brain. Postmitotic neurons require microtubules and dynein to move, but the mechanisms by which they contribute to migration are not fully characterized. Using tegmental hindbrain nuclei neurons in zebrafish embryos together with subcellular imaging, optogenetics, and photopharmacology, we show that, in vivo, the centrosome's position relative to the nucleus is not linked to greatest motility in this cell type. Nevertheless, microtubules, dynein, and kinesin-1 are essential for migration, and we find that interference with endosome formation or the Golgi apparatus impairs migration to a similar extent as disrupting microtubules. In addition, an imbalance in the traffic of the model cargo Cadherin-2 also reduces neuronal migration. These results lead us to propose that microtubules act as cargo carriers to control spatiotemporal protein distribution, which in turn controls motility. This adds crucial insights into the variety of ways that microtubules can support successful neuronal migration in vivo.
Collapse
Affiliation(s)
- Ulrike Theisen
- Technical University of Braunschweig, Zoological Institute, Cellular and Molecular Neurobiology, Braunschweig, Germany
| | - Alexander U Ernst
- Technical University of Braunschweig, Zoological Institute, Cellular and Molecular Neurobiology, Braunschweig, Germany.,University of Bern, Institute of Anatomy, Bern, Switzerland
| | - Ronja L S Heyne
- Technical University of Braunschweig, Zoological Institute, Cellular and Molecular Neurobiology, Braunschweig, Germany.,Danish Stem Cell Center, University of Copenhagen, Copenhagen, Denmark
| | - Tobias P Ring
- Technical University of Braunschweig, Institute for Acoustics, Braunschweig, Germany
| | - Oliver Thorn-Seshold
- Department of Pharmacy, Ludwig Maximilians University of Munich, Munich, Germany
| | - Reinhard W Köster
- Technical University of Braunschweig, Zoological Institute, Cellular and Molecular Neurobiology, Braunschweig, Germany
| |
Collapse
|
39
|
López-Cuevas P, Deane L, Yang Y, Hammond CL, Kague E. Transformed notochordal cells trigger chronic wounds destabilizing the vertebral column and bone homeostasis. Dis Model Mech 2021; 14:dmm.047001. [PMID: 33579726 PMCID: PMC7988777 DOI: 10.1242/dmm.047001] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Accepted: 02/01/2021] [Indexed: 12/14/2022] Open
Abstract
Notochordal cells play a pivotal role in vertebral column patterning, contributing to the formation of the inner architecture of intervertebral discs (IVDs). Their disappearance during development has been associated with reduced repair capacity and IVD degeneration. Notochord cells can give rise to chordomas, a highly invasive bone cancer associated with late diagnosis. Understanding the impact of neoplastic cells during development and on the surrounding vertebral column could open avenues for earlier intervention and therapeutics. We investigated the impact of transformed notochord cells in the zebrafish skeleton using a line expressing RAS in the notochord under the control of the kita promoter, with the advantage of adulthood endurance. Transformed cells caused damage in the notochord and destabilised the sheath layer, triggering a wound repair mechanism, with enrolment of sheath cells (col9a2+) and expression of wt1b, similar to induced notochord wounds. Moreover, increased recruitment of neutrophils and macrophages, displaying abnormal behaviour in proximity to the notochord sheath and transformed cells, supported parallels between chordomas, wound and inflammation. Cancerous notochordal cells interfere with differentiation of sheath cells to form chordacentra domains, leading to fusions and vertebral clefts during development. Adults displayed IVD irregularities reminiscent of degeneration, including reduced bone mineral density and increased osteoclast activity, along with disorganised osteoblasts and collagen, indicating impaired bone homeostasis. By depleting inflammatory cells, we abrogated chordoma development and rescued the skeletal features of the vertebral column. Therefore, we showed that transformed notochord cells alter the skeleton during life, causing a wound-like phenotype and activating chronic wound response, suggesting parallels between chordoma, wound, IVD degeneration and inflammation, highlighting inflammation as a promising target for future therapeutics. This article has an associated First Person interview with the first author of the paper. Summary: Analyses using a zebrafish line expressing RAS in the notochord, under the control of the kita promoter, revealed that transformed notochord cells alter the skeleton during life, causing a wound-like phenotype and activating chronic wound response.
Collapse
Affiliation(s)
- Paco López-Cuevas
- The School of Biochemistry, Biomedical Sciences, University of Bristol, BS8 1TD, UK
| | - Luke Deane
- The School of Biochemistry, Biomedical Sciences, University of Bristol, BS8 1TD, UK
| | - Yushi Yang
- School of Physics, HH Wills Physics Laboratory, University of Bristol, BS8 1TL, UK.,Centre for Nanoscience and Quantum Information, University of Bristol, Bristol, BS8 1FD, UK.,Bristol Centre for Functional Nanomaterials, University of Bristol, Bristol, BS8 1TL, UK
| | - Chrissy L Hammond
- The School of Physiology, Pharmacology and Neuroscience, Biomedical Sciences, University of Bristol, BS8 1TD, UK
| | - Erika Kague
- The School of Physiology, Pharmacology and Neuroscience, Biomedical Sciences, University of Bristol, BS8 1TD, UK
| |
Collapse
|
40
|
Mao A, Zhang M, Li L, Liu J, Ning G, Cao Y, Wang Q. Pharyngeal pouches provide a niche microenvironment for arch artery progenitor specification. Development 2021; 148:dev.192658. [PMID: 33334861 PMCID: PMC7847271 DOI: 10.1242/dev.192658] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Accepted: 12/10/2020] [Indexed: 11/20/2022]
Abstract
The paired pharyngeal arch arteries (PAAs) are transient blood vessels connecting the heart with the dorsal aorta during embryogenesis. Although PAA malformations often occur along with pharyngeal pouch defects, the functional interaction between these adjacent tissues remains largely unclear. Here, we report that pharyngeal pouches are essential for PAA progenitor specification in zebrafish embryos. We reveal that the segmentation of pharyngeal pouches coincides spatiotemporally with the emergence of PAA progenitor clusters. These pouches physically associate with pharyngeal mesoderm in discrete regions and provide a niche microenvironment for PAA progenitor commitment by expressing BMP proteins. Specifically, pouch-derived BMP2a and BMP5 are the primary niche cues responsible for activating the BMP/Smad pathway in pharyngeal mesoderm, thereby promoting progenitor specification. In addition, BMP2a and BMP5 play an inductive function in the expression of the cloche gene npas4l in PAA progenitors. cloche mutants exhibit a striking failure to specify PAA progenitors and display ectopic expression of head muscle markers in the pharyngeal mesoderm. Therefore, our results support a crucial role for pharyngeal pouches in establishing a progenitor niche for PAA morphogenesis via BMP2a/5 expression.
Collapse
Affiliation(s)
- Aihua Mao
- State Key Laboratory of Membrane Biology, Institute of Zoology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing 100101, China
| | - Mingming Zhang
- State Key Laboratory of Membrane Biology, Institute of Zoology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing 100101, China
| | - Linwei Li
- State Key Laboratory of Membrane Biology, Institute of Zoology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing 100101, China
| | - Jie Liu
- State Key Laboratory of Membrane Biology, Institute of Zoology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing 100101, China
| | - Guozhu Ning
- State Key Laboratory of Membrane Biology, Institute of Zoology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing 100101, China
| | - Yu Cao
- State Key Laboratory of Membrane Biology, Institute of Zoology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing 100101, China
| | - Qiang Wang
- State Key Laboratory of Membrane Biology, Institute of Zoology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing 100101, China .,Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China
| |
Collapse
|
41
|
Noel NCL, MacDonald IM, Allison WT. Zebrafish Models of Photoreceptor Dysfunction and Degeneration. Biomolecules 2021; 11:78. [PMID: 33435268 PMCID: PMC7828047 DOI: 10.3390/biom11010078] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2020] [Revised: 01/02/2021] [Accepted: 01/04/2021] [Indexed: 12/15/2022] Open
Abstract
Zebrafish are an instrumental system for the generation of photoreceptor degeneration models, which can be utilized to determine underlying causes of photoreceptor dysfunction and death, and for the analysis of potential therapeutic compounds, as well as the characterization of regenerative responses. We review the wealth of information from existing zebrafish models of photoreceptor disease, specifically as they relate to currently accepted taxonomic classes of human rod and cone disease. We also highlight that rich, detailed information can be derived from studying photoreceptor development, structure, and function, including behavioural assessments and in vivo imaging of zebrafish. Zebrafish models are available for a diversity of photoreceptor diseases, including cone dystrophies, which are challenging to recapitulate in nocturnal mammalian systems. Newly discovered models of photoreceptor disease and drusenoid deposit formation may not only provide important insights into pathogenesis of disease, but also potential therapeutic approaches. Zebrafish have already shown their use in providing pre-clinical data prior to testing genetic therapies in clinical trials, such as antisense oligonucleotide therapy for Usher syndrome.
Collapse
Affiliation(s)
- Nicole C. L. Noel
- Department of Medical Genetics, University of Alberta, Edmonton, AB T6G 2H7, Canada; (I.M.M.); (W.T.A.)
| | - Ian M. MacDonald
- Department of Medical Genetics, University of Alberta, Edmonton, AB T6G 2H7, Canada; (I.M.M.); (W.T.A.)
- Department of Ophthalmology and Visual Sciences, University of Alberta, Edmonton, AB T6G 2R7, Canada
| | - W. Ted Allison
- Department of Medical Genetics, University of Alberta, Edmonton, AB T6G 2H7, Canada; (I.M.M.); (W.T.A.)
- Department of Biological Sciences, University of Alberta, Edmonton, AB T6G 2E9, Canada
- Centre for Prions and Protein Folding Diseases, University of Alberta, Edmonton, AB T6G 2M8, Canada
| |
Collapse
|
42
|
Studying the Tumor Microenvironment in Zebrafish. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1329:69-92. [PMID: 34664234 DOI: 10.1007/978-3-030-73119-9_4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The tumor microenvironment significantly contributes to tumor initiation, progression, neo-angiogenesis, and metastasis, and a better understanding of the role of the different cellular players would facilitate the development of novel therapeutic strategies for cancer treatment. Towards this goal, intravital imaging is a powerful method to unravel interaction partners of tumor cells. Among vertebrate model organisms, zebrafish is uniquely suited for in vivo imaging studies. In recent years zebrafish has also become a valuable model in cancer research. In this chapter, we will summarize, how zebrafish has been used to characterize cells of the tumor microenvironment. We will cover both genetically engineered cancer models and xenograft models in zebrafish. The majority of work has been done on the role of innate immune cells and their role during tumor initiation and metastasis, but we will also cover studies focusing on adipocytes, fibroblasts, and endothelial cells. Taken together, we will highlight the versatile use of the zebrafish model for in vivo tumor microenvironment studies.
Collapse
|
43
|
Symonds AC, Buckley CE, Williams CA, Clarke JDW. Coordinated assembly and release of adhesions builds apical junctional belts during de novo polarisation of an epithelial tube. Development 2020; 147:dev191494. [PMID: 33361092 PMCID: PMC7774892 DOI: 10.1242/dev.191494] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Accepted: 10/28/2020] [Indexed: 01/05/2023]
Abstract
Using the zebrafish neural tube as a model, we uncover the in vivo mechanisms allowing the generation of two opposing apical epithelial surfaces within the centre of an initially unpolarised, solid organ. We show that Mpp5a and Rab11a play a dual role in coordinating the generation of ipsilateral junctional belts whilst simultaneously releasing contralateral adhesions across the centre of the tissue. We show that Mpp5a- and Rab11a-mediated resolution of cell-cell adhesions are both necessary for midline lumen opening and contribute to later maintenance of epithelial organisation. We propose that these roles for both Mpp5a and Rab11a operate through the transmembrane protein Crumbs. In light of a recent conflicting publication, we also clarify that the junction-remodelling role of Mpp5a is not specific to dividing cells.
Collapse
Affiliation(s)
- Andrew C Symonds
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King's College London, Guy's Campus, London SE1 1UL, UK
| | - Clare E Buckley
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King's College London, Guy's Campus, London SE1 1UL, UK
- Department of Physiology, Development and Neuroscience, University of Cambridge, Downing Street, Cambridge CB2 3BY, UK
| | - Charlotte A Williams
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King's College London, Guy's Campus, London SE1 1UL, UK
| | - Jonathan D W Clarke
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King's College London, Guy's Campus, London SE1 1UL, UK
| |
Collapse
|
44
|
Rascher M, Wittstein K, Winter B, Rupcic Z, Wolf-Asseburg A, Stadler M, Köster RW. Erinacine C Activates Transcription from a Consensus ETS DNA Binding Site in Astrocytic Cells in Addition to NGF Induction. Biomolecules 2020; 10:E1440. [PMID: 33066380 PMCID: PMC7602259 DOI: 10.3390/biom10101440] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Revised: 10/08/2020] [Accepted: 10/13/2020] [Indexed: 11/16/2022] Open
Abstract
Medicinal mushrooms of the genus Hericium are known to produce secondary metabolites with homeostatic properties for the central nervous system. We and others have recently demonstrated that among these metabolites cyathane diterpenoids and in particular erinacine C possess potent neurotrophin inducing properties in astrocytic cells. Yet, the signaling events downstream of erinacine C induced neurotrophin acitivity in neural-like adrenal phaeochromocytoma cells (PC12) cells have remained elusive. Similar, signaling events activated by erinacine C in astrocytic cells are unknown. Using a combination of genetic and pharmacological inhibitors we show that erinacine C induced neurotrophic activity mediates PC12 cell differentiation via the TrkA receptor and likely its associated PLCγ-, PI3K-, and MAPK/ERK pathways. Furthermore, a small library of transcriptional activation reporters revealed that erinacine C induces transcriptional activation mediated by DNA consensus binding sites of selected conserved transcription factor families. Among these, transcription is activated from an ETS consensus in a concentration dependent manner. Interestingly, induced ETS-consensus transcription occurs in parallel and independent of neurotrophin induction. This finding helps to explain the many pleiotropic functions of cyathane diterpenoids. Moreover, our studies provide genetic access to cyathane diterpenoid functions in astrocytic cells and help to mechanistically understand the action of cyathanes in glial cells.
Collapse
Affiliation(s)
- Monique Rascher
- Division of Cellular and Molecular Biology, Zoological Institute, Technische Universität Braunschweig, Spielmannstraße 7, 38106 Braunschweig, Germany; (M.R.); (B.W.); (A.W.-A.)
- Department Microbial Drugs, Helmholtz Centre for Infection Research GmbH and Institute of Microbiology, Technische Universität Braunschweig, Inhoffenstraße 7, 38124 Braunschweig, Germany; (K.W.); (Z.R.)
- German Centre for Infection Research (DZIF), Technische Universität Braunschweig, Partner Site Hannover-Braunschweig, 38124 Braunschweig, Germany
| | - Kathrin Wittstein
- Department Microbial Drugs, Helmholtz Centre for Infection Research GmbH and Institute of Microbiology, Technische Universität Braunschweig, Inhoffenstraße 7, 38124 Braunschweig, Germany; (K.W.); (Z.R.)
- German Centre for Infection Research (DZIF), Technische Universität Braunschweig, Partner Site Hannover-Braunschweig, 38124 Braunschweig, Germany
| | - Barbara Winter
- Division of Cellular and Molecular Biology, Zoological Institute, Technische Universität Braunschweig, Spielmannstraße 7, 38106 Braunschweig, Germany; (M.R.); (B.W.); (A.W.-A.)
| | - Zeljka Rupcic
- Department Microbial Drugs, Helmholtz Centre for Infection Research GmbH and Institute of Microbiology, Technische Universität Braunschweig, Inhoffenstraße 7, 38124 Braunschweig, Germany; (K.W.); (Z.R.)
- German Centre for Infection Research (DZIF), Technische Universität Braunschweig, Partner Site Hannover-Braunschweig, 38124 Braunschweig, Germany
| | - Alexandra Wolf-Asseburg
- Division of Cellular and Molecular Biology, Zoological Institute, Technische Universität Braunschweig, Spielmannstraße 7, 38106 Braunschweig, Germany; (M.R.); (B.W.); (A.W.-A.)
| | - Marc Stadler
- Department Microbial Drugs, Helmholtz Centre for Infection Research GmbH and Institute of Microbiology, Technische Universität Braunschweig, Inhoffenstraße 7, 38124 Braunschweig, Germany; (K.W.); (Z.R.)
- German Centre for Infection Research (DZIF), Technische Universität Braunschweig, Partner Site Hannover-Braunschweig, 38124 Braunschweig, Germany
- Institute of Microbiology, Technische Universität Braunschweig, Spielmannstraße 7, 38106 Braunschweig, Germany
| | - Reinhard W. Köster
- Division of Cellular and Molecular Biology, Zoological Institute, Technische Universität Braunschweig, Spielmannstraße 7, 38106 Braunschweig, Germany; (M.R.); (B.W.); (A.W.-A.)
| |
Collapse
|
45
|
Burgess J, Burrows JT, Sadhak R, Chiang S, Weiss A, D'Amata C, Molinaro AM, Zhu S, Long M, Hu C, Krause HM, Pearson BJ. An optimized QF-binary expression system for use in zebrafish. Dev Biol 2020; 465:144-156. [PMID: 32697972 DOI: 10.1016/j.ydbio.2020.07.007] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Revised: 07/05/2020] [Accepted: 07/09/2020] [Indexed: 11/15/2022]
Abstract
The zebrafish model organism has been of exceptional utility for the study of vertebrate development and disease through the application of tissue-specific labelling and overexpression of genes carrying patient-derived mutations. However, there remains a need for a binary expression system that is both non-toxic and not silenced over animal generations by DNA methylation. The Q binary expression system derived from the fungus Neurospora crassa is ideal, because the consensus binding site for the QF transcription factor lacks CpG dinucleotides, precluding silencing by CpG-meditated methylation. To optimize this system for zebrafish, we systematically tested several variants of the QF transcription factor: QF full length; QF2, which lacks the middle domain; QF2w, which is an attenuated version of QF2; and chimeric QFGal4. We found that full length QF and QF2 were strongly toxic to zebrafish embryos, QF2w was mildly toxic, and QFGal4 was well tolerated, when injected as RNA or expressed ubiquitously from stable transgenes. In addition, QFGal4 robustly activated a Tg(QUAS:GFPNLS) reporter transgene. To increase the utility of this system, we also modified the QF effector sequence termed QUAS, which consists of five copies of the QF binding site. Specifically, we decreased both the CpG dinucleotide content, as well as the repetitiveness of QUAS, to reduce the risk of transgene silencing via CpG methylation. Moreover, these modifications to QUAS removed leaky QF-independent neural expression that we detected in the original QUAS sequence. To demonstrate the utility of our QF optimizations, we show how the Q-system can be used for lineage tracing using a Cre-dependent Tg(ubi:QFGal4-switch) transgene. We also demonstrate that QFGal4 can be used in transient injections to tag and label endogenous genes by knocking in QFGal4 into sox2 and ubiquitin C genes.
Collapse
Affiliation(s)
- Jason Burgess
- The Hospital for Sick Children, Program in Developmental and Stem Cell Biology, Toronto, ON, M5G 0A4, Canada
| | - Jeffrey T Burrows
- The Hospital for Sick Children, Program in Developmental and Stem Cell Biology, Toronto, ON, M5G 0A4, Canada
| | - Roshan Sadhak
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Sharon Chiang
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Alex Weiss
- The Hospital for Sick Children, Program in Developmental and Stem Cell Biology, Toronto, ON, M5G 0A4, Canada
| | - Cassandra D'Amata
- The Hospital for Sick Children, Program in Developmental and Stem Cell Biology, Toronto, ON, M5G 0A4, Canada
| | - Alyssa M Molinaro
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Shujun Zhu
- The Hospital for Sick Children, Program in Developmental and Stem Cell Biology, Toronto, ON, M5G 0A4, Canada
| | - Michael Long
- The Donnelly Centre for Cellular and Biomolecular Research (CCBR), University of Toronto, Toronto, ON, M5S 3E1, Canada
| | - Chun Hu
- The Donnelly Centre for Cellular and Biomolecular Research (CCBR), University of Toronto, Toronto, ON, M5S 3E1, Canada
| | - Henry M Krause
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada; The Donnelly Centre for Cellular and Biomolecular Research (CCBR), University of Toronto, Toronto, ON, M5S 3E1, Canada
| | - Bret J Pearson
- The Hospital for Sick Children, Program in Developmental and Stem Cell Biology, Toronto, ON, M5G 0A4, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada; Ontario Institute for Cancer Research, Toronto, ON, Canada.
| |
Collapse
|
46
|
Collins RT, Coxam B, Fechner I, Unterweger I, Szymborska A, Meier K, Gerhardt H. Intron with transgenic marker (InTraM) facilitates high-throughput screening of endogenous gene reporter lines. Genesis 2020; 58:e23391. [PMID: 32783355 DOI: 10.1002/dvg.23391] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 07/15/2020] [Accepted: 07/16/2020] [Indexed: 11/07/2022]
Abstract
The generation and maintenance of genome edited zebrafish lines is typically labor intensive due to the lack of an easy visual read-out for the modification. To facilitate this process, we have developed a novel method that relies on the inclusion of an artificial intron with a transgenic marker (InTraM) within the knock-in sequence of interest, which upon splicing produces a transcript with a precise and seamless modification. We have demonstrated this technology by replacing the stop codon of the zebrafish fli1a gene with a transcriptional activator KALTA4, using an InTraM that enables red fluorescent protein expression in the heart.
Collapse
Affiliation(s)
- Russell T Collins
- Integrative Vascular Biology Lab, Max-Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Baptiste Coxam
- Integrative Vascular Biology Lab, Max-Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany
| | - Ines Fechner
- Integrative Vascular Biology Lab, Max-Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany
| | - Iris Unterweger
- Integrative Vascular Biology Lab, Max-Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- Novo Nordisk Foundation Center for Stem Cell Biology, Copenhagen, Denmark
| | - Anna Szymborska
- Integrative Vascular Biology Lab, Max-Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany
| | - Katja Meier
- Integrative Vascular Biology Lab, Max-Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany
| | - Holger Gerhardt
- Integrative Vascular Biology Lab, Max-Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany
- Berlin Institute of Health (BIH), Berlin, Germany
| |
Collapse
|
47
|
Hartmann J, Wong M, Gallo E, Gilmour D. An image-based data-driven analysis of cellular architecture in a developing tissue. eLife 2020; 9:e55913. [PMID: 32501214 PMCID: PMC7274788 DOI: 10.7554/elife.55913] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Accepted: 05/24/2020] [Indexed: 12/22/2022] Open
Abstract
Quantitative microscopy is becoming increasingly crucial in efforts to disentangle the complexity of organogenesis, yet adoption of the potent new toolbox provided by modern data science has been slow, primarily because it is often not directly applicable to developmental imaging data. We tackle this issue with a newly developed algorithm that uses point cloud-based morphometry to unpack the rich information encoded in 3D image data into a straightforward numerical representation. This enabled us to employ data science tools, including machine learning, to analyze and integrate cell morphology, intracellular organization, gene expression and annotated contextual knowledge. We apply these techniques to construct and explore a quantitative atlas of cellular architecture for the zebrafish posterior lateral line primordium, an experimentally tractable model of complex self-organized organogenesis. In doing so, we are able to retrieve both previously established and novel biologically relevant patterns, demonstrating the potential of our data-driven approach.
Collapse
Affiliation(s)
- Jonas Hartmann
- Cell Biology and Biophysics Unit, European Molecular Biology Laboratory (EMBL)HeidelbergGermany
| | - Mie Wong
- Institute of Molecular Life Sciences, University of Zurich (UZH)ZurichSwitzerland
| | - Elisa Gallo
- Cell Biology and Biophysics Unit, European Molecular Biology Laboratory (EMBL)HeidelbergGermany
- Institute of Molecular Life Sciences, University of Zurich (UZH)ZurichSwitzerland
- Collaboration for joint PhD degree between EMBL and Heidelberg University, Faculty of BiosciencesHeidelbergGermany
| | - Darren Gilmour
- Institute of Molecular Life Sciences, University of Zurich (UZH)ZurichSwitzerland
| |
Collapse
|
48
|
Varady A, Distel M. Non-neuromodulatory Optogenetic Tools in Zebrafish. Front Cell Dev Biol 2020; 8:418. [PMID: 32582702 PMCID: PMC7283495 DOI: 10.3389/fcell.2020.00418] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Accepted: 05/05/2020] [Indexed: 01/18/2023] Open
Abstract
The zebrafish (Danio rerio) is a popular vertebrate model organism to investigate molecular mechanisms driving development and disease. Due to its transparency at embryonic and larval stages, investigations in the living organism are possible with subcellular resolution using intravital microscopy. The beneficial optical characteristics of zebrafish not only allow for passive observation, but also active manipulation of proteins and cells by light using optogenetic tools. Initially, photosensitive ion channels have been applied for neurobiological studies in zebrafish to dissect complex behaviors on a cellular level. More recently, exciting non-neural optogenetic tools have been established to control gene expression or protein localization and activity, allowing for unprecedented non-invasive and precise manipulation of various aspects of cellular physiology. Zebrafish will likely be a vertebrate model organism at the forefront of in vivo application of non-neural optogenetic tools and pioneering work has already been performed. In this review, we provide an overview of non-neuromodulatory optogenetic tools successfully applied in zebrafish to control gene expression, protein localization, cell signaling, migration and cell ablation.
Collapse
Affiliation(s)
- Adam Varady
- St. Anna Children's Cancer Research Institute, Innovative Cancer Models, Vienna, Austria
| | - Martin Distel
- St. Anna Children's Cancer Research Institute, Innovative Cancer Models, Vienna, Austria
| |
Collapse
|
49
|
Kuil LE, Oosterhof N, Ferrero G, Mikulášová T, Hason M, Dekker J, Rovira M, van der Linde HC, van Strien PMH, de Pater E, Schaaf G, Bindels EMJ, Wittamer V, van Ham TJ. Zebrafish macrophage developmental arrest underlies depletion of microglia and reveals Csf1r-independent metaphocytes. eLife 2020; 9:e53403. [PMID: 32367800 PMCID: PMC7237208 DOI: 10.7554/elife.53403] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Accepted: 04/24/2020] [Indexed: 12/18/2022] Open
Abstract
Macrophages derive from multiple sources of hematopoietic progenitors. Most macrophages require colony-stimulating factor 1 receptor (CSF1R), but some macrophages persist in the absence of CSF1R. Here, we analyzed mpeg1:GFP-expressing macrophages in csf1r-deficient zebrafish and report that embryonic macrophages emerge followed by their developmental arrest. In larvae, mpeg1+ cell numbers then increased showing two distinct types in the skin: branched, putative Langerhans cells, and amoeboid cells. In contrast, although numbers also increased in csf1r-mutants, exclusively amoeboid mpeg1+ cells were present, which we showed by genetic lineage tracing to have a non-hematopoietic origin. They expressed macrophage-associated genes, but also showed decreased phagocytic gene expression and increased epithelial-associated gene expression, characteristic of metaphocytes, recently discovered ectoderm-derived cells. We further demonstrated that juvenile csf1r-deficient zebrafish exhibit systemic macrophage depletion. Thus, csf1r deficiency disrupts embryonic to adult macrophage development. Zebrafish deficient for csf1r are viable and permit analyzing the consequences of macrophage loss throughout life.
Collapse
Affiliation(s)
- Laura E Kuil
- Department of Clinical Genetics, Erasmus University Medical Center RotterdamRotterdamNetherlands
| | - Nynke Oosterhof
- Department of Clinical Genetics, Erasmus University Medical Center RotterdamRotterdamNetherlands
| | - Giuliano Ferrero
- Institut de Recherche Interdisciplinaire en Biologie Humaine et Moléculaire (IRIBHM), Université Libre de Bruxelles (ULB)BrusselsBelgium
| | - Tereza Mikulášová
- Laboratory of Cell Differentiation, Institute of Molecular Genetics of the Czech Academy of SciencesPragueCzech Republic
| | - Martina Hason
- Laboratory of Cell Differentiation, Institute of Molecular Genetics of the Czech Academy of SciencesPragueCzech Republic
| | - Jordy Dekker
- Department of Clinical Genetics, Erasmus University Medical Center RotterdamRotterdamNetherlands
| | - Mireia Rovira
- Institut de Recherche Interdisciplinaire en Biologie Humaine et Moléculaire (IRIBHM), Université Libre de Bruxelles (ULB)BrusselsBelgium
| | - Herma C van der Linde
- Department of Clinical Genetics, Erasmus University Medical Center RotterdamRotterdamNetherlands
| | | | - Emma de Pater
- Department of Hematology, Erasmus University Medical CenterRotterdamNetherlands
| | - Gerben Schaaf
- Department of Clinical Genetics, Erasmus University Medical Center RotterdamRotterdamNetherlands
| | - Erik MJ Bindels
- Department of Hematology, Erasmus University Medical CenterRotterdamNetherlands
| | - Valerie Wittamer
- Institut de Recherche Interdisciplinaire en Biologie Humaine et Moléculaire (IRIBHM), Université Libre de Bruxelles (ULB)BrusselsBelgium
- WELBIO, ULBBrusselsBelgium
| | - Tjakko J van Ham
- Department of Clinical Genetics, Erasmus University Medical Center RotterdamRotterdamNetherlands
| |
Collapse
|
50
|
Shrestha R, Lieberth J, Tillman S, Natalizio J, Bloomekatz J. Using Zebrafish to Analyze the Genetic and Environmental Etiologies of Congenital Heart Defects. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1236:189-223. [PMID: 32304074 DOI: 10.1007/978-981-15-2389-2_8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Congenital heart defects (CHDs) are among the most common human birth defects. However, the etiology of a large proportion of CHDs remains undefined. Studies identifying the molecular and cellular mechanisms that underlie cardiac development have been critical to elucidating the origin of CHDs. Building upon this knowledge to understand the pathogenesis of CHDs requires examining how genetic or environmental stress changes normal cardiac development. Due to strong molecular conservation to humans and unique technical advantages, studies using zebrafish have elucidated both fundamental principles of cardiac development and have been used to create cardiac disease models. In this chapter we examine the unique toolset available to zebrafish researchers and how those tools are used to interrogate the genetic and environmental contributions to CHDs.
Collapse
Affiliation(s)
- Rabina Shrestha
- Department of Biology, University of Mississippi, Oxford, MS, USA
| | - Jaret Lieberth
- Department of Biology, University of Mississippi, Oxford, MS, USA
| | - Savanna Tillman
- Department of Biology, University of Mississippi, Oxford, MS, USA
| | - Joseph Natalizio
- Department of Biology, University of Mississippi, Oxford, MS, USA
| | | |
Collapse
|