1
|
Lush ME, Tsai YY, Chen S, Münch D, Peloggia J, Sandler JE, Piotrowski T. Stem and progenitor cell proliferation are independently regulated by cell type-specific cyclinD genes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.21.619490. [PMID: 39484411 PMCID: PMC11526906 DOI: 10.1101/2024.10.21.619490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
Regeneration and homeostatic turnover of solid tissues depend on the proliferation of symmetrically dividing adult stem cells, which either remain stem cells or differentiate based on their niche position. Here we demonstrate that in zebrafish lateral line sensory organs, stem and progenitor cell proliferation are independently regulated by two cyclinD genes. Loss of ccnd2a impairs stem cell proliferation during development, while loss of ccndx disrupts hair cell progenitor proliferation but allows normal differentiation. Notably, ccnd2a can functionally replace ccndx , indicating that the respective effects of these Cyclins on proliferation are due to cell type-specific expression. However, even though hair cell progenitors differentiate normally in ccndx mutants, they are mispolarized due to hes2 and Emx2 downregulation. Thus, regulated proliferation ensures that equal numbers of hair cells are polarized in opposite directions. Our study reveals cell type-specific roles for cyclinD genes in regulating the different populations of symmetrically dividing cells governing organ development and regeneration, with implications for regenerative medicine and disease.
Collapse
|
2
|
Brooks PM, Lewis P, Million-Perez S, Yandulskaya AS, Khalil M, Janes M, Porco J, Walker E, Meyers JR. Pharmacological reprogramming of zebrafish lateral line supporting cells to a migratory progenitor state. Dev Biol 2024; 512:70-88. [PMID: 38729405 DOI: 10.1016/j.ydbio.2024.05.003] [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: 10/27/2023] [Revised: 04/17/2024] [Accepted: 05/07/2024] [Indexed: 05/12/2024]
Abstract
In the zebrafish lateral line, non-sensory supporting cells readily re-enter the cell cycle to generate new hair cells and supporting cells during homeostatic maintenance and following damage to hair cells. This contrasts with supporting cells from mammalian vestibular and auditory sensory epithelia which rarely re-enter the cell cycle, and hence loss of hair cells results in permanent sensory deficit. Lateral line supporting cells are derived from multipotent progenitor cells that migrate down the trunk midline as a primordium and are deposited to differentiate into a neuromast. We have found that we can revert zebrafish support cells back to a migratory progenitor state by pharmacologically altering the signaling environment to mimic that of the migratory primordium, with active Wnt signaling and repressed FGF signaling. The reverted supporting cells migrate anteriorly and posteriorly along the horizontal myoseptum and will re-epithelialize to form an increased number of neuromasts along the midline when the pharmacological agents are removed. These data demonstrate that supporting cells can be readily reprogrammed to a migratory multipotent progenitor state that can form new sensory neuromasts, which has important implications for our understanding of how the lateral line system matures and expands in fish and also suggest avenues for returning mammalian supporting cells back to a proliferative state.
Collapse
Affiliation(s)
- Paige M Brooks
- Dept. of Biology and Program in Neuroscience, Colgate University, 13 Oak Drive, Hamilton, NY, 13346, USA
| | - Parker Lewis
- Dept. of Biology and Program in Neuroscience, Colgate University, 13 Oak Drive, Hamilton, NY, 13346, USA
| | - Sara Million-Perez
- Dept. of Biology and Program in Neuroscience, Colgate University, 13 Oak Drive, Hamilton, NY, 13346, USA
| | - Anastasia S Yandulskaya
- Dept. of Biology and Program in Neuroscience, Colgate University, 13 Oak Drive, Hamilton, NY, 13346, USA
| | - Mahmoud Khalil
- Dept. of Biology and Program in Neuroscience, Colgate University, 13 Oak Drive, Hamilton, NY, 13346, USA
| | - Meredith Janes
- Dept. of Biology and Program in Neuroscience, Colgate University, 13 Oak Drive, Hamilton, NY, 13346, USA
| | - Joseph Porco
- Dept. of Biology and Program in Neuroscience, Colgate University, 13 Oak Drive, Hamilton, NY, 13346, USA
| | - Eleanor Walker
- Dept. of Biology and Program in Neuroscience, Colgate University, 13 Oak Drive, Hamilton, NY, 13346, USA
| | - Jason R Meyers
- Dept. of Biology and Program in Neuroscience, Colgate University, 13 Oak Drive, Hamilton, NY, 13346, USA.
| |
Collapse
|
3
|
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
|
4
|
Aman AJ, Parichy DM. Anatomy, development and regeneration of zebrafish elasmoid scales. Dev Biol 2024; 510:1-7. [PMID: 38458375 PMCID: PMC11015963 DOI: 10.1016/j.ydbio.2024.03.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2023] [Revised: 02/22/2024] [Accepted: 03/05/2024] [Indexed: 03/10/2024]
Abstract
Vertebrate skin appendages - particularly avian feathers and mammalian hairs, glands and teeth - are perennially useful systems for investigating fundamental mechanisms of development. The most common type of skin appendage in teleost fishes is the elasmoid scale, yet this structure has received much less attention than the skin appendages of tetrapods. Elasmoid scales are thin, overlapping plates of partially mineralized extracellular matrices, deposited in the skin in a hexagonal pattern by a specialized population of dermal cells in cooperation with the overlying epidermis. Recent years have seen rapid progress in our understanding of elasmoid scale development and regeneration, driven by the deployment of developmental genetics, live imaging and transcriptomics in larval and adult zebrafish. These findings are reviewed together with histological and ultrastructural approaches to understanding scale development and regeneration.
Collapse
Affiliation(s)
- Andrew J Aman
- Department of Biology, University of Virginia, Charlottesville, VA, 22903, USA.
| | - David M Parichy
- Department of Biology, University of Virginia, Charlottesville, VA, 22903, USA; Department of Cell Biology, University of Virginia, Charlottesville, VA, 22903, USA.
| |
Collapse
|
5
|
Han M, Perkins MH, Novaes LS, Xu T, Chang H. Advances in transposable elements: from mechanisms to applications in mammalian genomics. Front Genet 2023; 14:1290146. [PMID: 38098473 PMCID: PMC10719622 DOI: 10.3389/fgene.2023.1290146] [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: 09/07/2023] [Accepted: 11/13/2023] [Indexed: 12/17/2023] Open
Abstract
It has been 70 years since Barbara McClintock discovered transposable elements (TE), and the mechanistic studies and functional applications of transposable elements have been at the forefront of life science research. As an essential part of the genome, TEs have been discovered in most species of prokaryotes and eukaryotes, and the relative proportion of the total genetic sequence they comprise gradually increases with the expansion of the genome. In humans, TEs account for about 40% of the genome and are deeply involved in gene regulation, chromosome structure maintenance, inflammatory response, and the etiology of genetic and non-genetic diseases. In-depth functional studies of TEs in mammalian cells and the human body have led to a greater understanding of these fundamental biological processes. At the same time, as a potent mutagen and efficient genome editing tool, TEs have been transformed into biological tools critical for developing new techniques. By controlling the random insertion of TEs into the genome to change the phenotype in cells and model organisms, critical proteins of many diseases have been systematically identified. Exploiting the TE's highly efficient in vitro insertion activity has driven the development of cutting-edge sequencing technologies. Recently, a new technology combining CRISPR with TEs was reported, which provides a novel targeted insertion system to both academia and industry. We suggest that interrogating biological processes that generally depend on the actions of TEs with TEs-derived genetic tools is a very efficient strategy. For example, excessive activation of TEs is an essential factor in the occurrence of cancer in humans. As potent mutagens, TEs have also been used to unravel the key regulatory elements and mechanisms of carcinogenesis. Through this review, we aim to effectively combine the traditional views of TEs with recent research progress, systematically link the mechanistic discoveries of TEs with the technological developments of TE-based tools, and provide a comprehensive approach and understanding for researchers in different fields.
Collapse
Affiliation(s)
- Mei Han
- Guangzhou National Laboratory, Guangzhou, China
| | - Matthew H. Perkins
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Leonardo Santana Novaes
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Tao Xu
- Guangzhou National Laboratory, Guangzhou, China
| | - Hao Chang
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| |
Collapse
|
6
|
Zhao L, Fong SH, Yang Q, Jiang YJ, Korzh V, Liou YC. The prolyl isomerase Pin1 stabilizes NeuroD during differentiation of mechanoreceptors. Front Cell Dev Biol 2023; 11:1225128. [PMID: 37791075 PMCID: PMC10543749 DOI: 10.3389/fcell.2023.1225128] [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: 05/18/2023] [Accepted: 08/11/2023] [Indexed: 10/05/2023] Open
Abstract
The peptidyl prolyl cis-trans isomerase Pin1 plays vital roles in diverse cellular processes and pathological conditions. NeuroD is a differentiation and survival factor for a subset of neurons and pancreatic endocrine cells. Although multiple phosphorylation events are known to be crucial for NeuroD function, their mechanisms remain elusive. In this study, we demonstrate that zebrafish embryos deficient in Pin1 displayed phenotypes resembling those associated with NeuroD depletion, characterized by defects in formation of mechanosensory hair cells. Furthermore, zebrafish Pin1 interacts with NeuroD in a phosphorylation-dependent manner. In Pin1-deficient cell lines, NeuroD is rapidly degraded. However, the protein stability of NeuroD is restored upon overexpression of Pin1. These findings suggest that Pin1 functionally regulates NeuroD protein levels by post-phosphorylation cis-trans isomerization during neuronal specification.
Collapse
Affiliation(s)
- Liqun Zhao
- Department of Biological Sciences, Faculty of Science, National University of Singapore, Singapore, Singapore
| | - Steven H. Fong
- Department of Biological Sciences, Faculty of Science, National University of Singapore, Singapore, Singapore
- Genes and Development Division, Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A-STAR), Singapore, Singapore
| | - Qiaoyun Yang
- Department of Biological Sciences, Faculty of Science, National University of Singapore, Singapore, Singapore
| | - Yun-Jin Jiang
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, Zhunan, Taiwan
| | - Vladimir Korzh
- Department of Biological Sciences, Faculty of Science, National University of Singapore, Singapore, Singapore
- Genes and Development Division, Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A-STAR), Singapore, Singapore
| | - Yih-Cherng Liou
- Department of Biological Sciences, Faculty of Science, National University of Singapore, Singapore, Singapore
| |
Collapse
|
7
|
Aman AJ, Saunders LM, Carr AA, Srivatasan S, Eberhard C, Carrington B, Watkins-Chow D, Pavan WJ, Trapnell C, Parichy DM. Transcriptomic profiling of tissue environments critical for post-embryonic patterning and morphogenesis of zebrafish skin. eLife 2023; 12:RP86670. [PMID: 37695017 PMCID: PMC10495112 DOI: 10.7554/elife.86670] [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: 09/12/2023] Open
Abstract
Pigment patterns and skin appendages are prominent features of vertebrate skin. In zebrafish, regularly patterned pigment stripes and an array of calcified scales form simultaneously in the skin during post-embryonic development. Understanding the mechanisms that regulate stripe patterning and scale morphogenesis may lead to the discovery of fundamental mechanisms that govern the development of animal form. To learn about cell types and signaling interactions that govern skin patterning and morphogenesis, we generated and analyzed single-cell transcriptomes of skin from wild-type fish as well as fish having genetic or transgenically induced defects in squamation or pigmentation. These data reveal a previously undescribed population of epidermal cells that express transcripts encoding enamel matrix proteins, suggest hormonal control of epithelial-mesenchymal signaling, clarify the signaling network that governs scale papillae development, and identify a critical role for the hypodermis in supporting pigment cell development. Additionally, these comprehensive single-cell transcriptomic data representing skin phenotypes of biomedical relevance should provide a useful resource for accelerating the discovery of mechanisms that govern skin development and homeostasis.
Collapse
Affiliation(s)
- Andrew J Aman
- Department of Biology, University of VirginiaCharlottesvilleUnited States
| | - Lauren M Saunders
- Department of Genome Sciences, University of WashingtonSeattleUnited States
| | - August A Carr
- Department of Biology, University of VirginiaCharlottesvilleUnited States
| | - Sanjay Srivatasan
- Department of Genome Sciences, University of WashingtonSeattleUnited States
| | - Colten Eberhard
- National Human Genome Research Institute, National Institutes of HealthBethesdaUnited States
| | - Blake Carrington
- National Human Genome Research Institute, National Institutes of HealthBethesdaUnited States
| | - Dawn Watkins-Chow
- National Human Genome Research Institute, National Institutes of HealthBethesdaUnited States
| | - William J Pavan
- National Human Genome Research Institute, National Institutes of HealthBethesdaUnited States
| | - Cole Trapnell
- Department of Genome Sciences, University of WashingtonSeattleUnited States
| | - David M Parichy
- Department of Biology, University of VirginiaCharlottesvilleUnited States
- Department of Cell Biology, University of VirginiaCharlottesvilleUnited States
| |
Collapse
|
8
|
Baldera D, Baxendale S, van Hateren NJ, Marzo M, Glendenning E, Geng F, Yokoya K, Knight RD, Whitfield TT. Enhancer trap lines with GFP driven by smad6b and frizzled1 regulatory sequences for the study of epithelial morphogenesis in the developing zebrafish inner ear. J Anat 2023; 243:78-89. [PMID: 36748120 PMCID: PMC10273346 DOI: 10.1111/joa.13845] [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: 11/30/2022] [Revised: 01/23/2023] [Accepted: 01/23/2023] [Indexed: 02/08/2023] Open
Abstract
Live imaging in the zebrafish embryo using tissue-specific expression of fluorescent proteins can yield important insights into the mechanisms that drive sensory organ morphogenesis and cell differentiation. Morphogenesis of the semicircular canal ducts of the vertebrate inner ear requires a complex rearrangement of epithelial cells, including outgrowth, adhesion, fusion and perforation of epithelial projections to generate pillars of tissue that form the hubs of each canal. We report the insertion sites and expression patterns of two enhancer trap lines in the developing zebrafish embryo, each of which highlight different aspects of epithelial cell morphogenesis in the inner ear. A membrane-linked EGFP driven by smad6b regulatory sequences is expressed throughout the otic epithelium, most strongly on the lateral side of the ear and in the sensory cristae. A second enhancer trap line, with cytoplasmic EGFP driven by frizzled1 (fzd1) regulatory sequences, specifically marks cells of the ventral projection and pillar in the developing ear, and marginal cells in the sensory cristae, together with variable expression in the retina and epiphysis, and neurons elsewhere in the developing central nervous system. We have used a combination of methods to identify the insertion sites of these two transgenes, which were generated through random insertion, and show that Targeted Locus Amplification is a rapid and reliable method for the identification of insertion sites of randomly inserted transgenes.
Collapse
Affiliation(s)
- Davide Baldera
- School of BiosciencesUniversity of SheffieldSheffieldUK
- Present address:
CeSASt, University of CagliariCagliariItaly
| | | | | | - Mar Marzo
- School of BiosciencesUniversity of SheffieldSheffieldUK
| | | | - Fan‐Suo Geng
- Brain and Mind Research Institute, University of SydneySydneyNew South WalesAustralia
- Present address:
Data Science Institute, The University of Technology SydneySydneyAustralia
| | - Kazutomo Yokoya
- Centre for Craniofacial and Regenerative Biology, King's College London, Guy's HospitalLondonUK
| | - Robert D. Knight
- Centre for Craniofacial and Regenerative Biology, King's College London, Guy's HospitalLondonUK
| | | |
Collapse
|
9
|
Murayama E, Vivier C, Schmidt A, Herbomel P. Alcam-a and Pdgfr-α are essential for the development of sclerotome-derived stromal cells that support hematopoiesis. Nat Commun 2023; 14:1171. [PMID: 36859431 PMCID: PMC9977867 DOI: 10.1038/s41467-023-36612-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Accepted: 02/09/2023] [Indexed: 03/03/2023] Open
Abstract
Mesenchymal stromal cells are essential components of hematopoietic stem and progenitor cell (HSPC) niches, regulating HSPC proliferation and fates. Their developmental origins are largely unknown. In zebrafish, we previously found that the stromal cells of the caudal hematopoietic tissue (CHT), a niche functionally homologous to the mammalian fetal liver, arise from the ventral part of caudal somites. We have now found that this ventral domain is the sclerotome, and that two markers of mammalian mesenchymal stem/stromal cells, Alcam and Pdgfr-α, are distinctively expressed there and instrumental for the emergence and migration of stromal cell progenitors, which in turn conditions the proper assembly of the vascular component of the CHT niche. Furthermore, we find that trunk somites are similarly dependent on Alcam and Pdgfr-α to produce mesenchymal cells that foster HSPC emergence from the aorta. Thus the sclerotome contributes essential stromal cells for each of the key steps of developmental hematopoiesis.
Collapse
Affiliation(s)
- Emi Murayama
- Institut Pasteur, Department of Developmental & Stem Cell Biology, Paris, 75015, France. .,INSERM, Paris, 75013, France. .,CNRS, UMR3738, Paris, 75015, France.
| | - Catherine Vivier
- Institut Pasteur, Department of Developmental & Stem Cell Biology, Paris, 75015, France.,CNRS, UMR3738, Paris, 75015, France
| | - Anne Schmidt
- Institut Pasteur, Department of Developmental & Stem Cell Biology, Paris, 75015, France.,CNRS, UMR3738, Paris, 75015, France
| | - Philippe Herbomel
- Institut Pasteur, Department of Developmental & Stem Cell Biology, Paris, 75015, France.,CNRS, UMR3738, Paris, 75015, France
| |
Collapse
|
10
|
Wu M, Xu J, Zhang Y, Wen Z. Learning from Zebrafish Hematopoiesis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1442:137-157. [PMID: 38228963 DOI: 10.1007/978-981-99-7471-9_9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/18/2024]
Abstract
Hematopoiesis is a complex process that tightly regulates the generation, proliferation, differentiation, and maintenance of hematopoietic cells. Disruptions in hematopoiesis can lead to various diseases affecting both hematopoietic and non-hematopoietic systems, such as leukemia, anemia, thrombocytopenia, rheumatoid arthritis, and chronic granuloma. The zebrafish serves as a powerful vertebrate model for studying hematopoiesis, offering valuable insights into both hematopoietic regulation and hematopoietic diseases. In this chapter, we present a comprehensive overview of zebrafish hematopoiesis, highlighting its distinctive characteristics in hematopoietic processes. We discuss the ontogeny and modulation of both primitive and definitive hematopoiesis, as well as the microenvironment that supports hematopoietic stem/progenitor cells. Additionally, we explore the utility of zebrafish as a disease model and its potential in drug discovery, which not only advances our understanding of the regulatory mechanisms underlying hematopoiesis but also facilitates the exploration of novel therapeutic strategies for hematopoietic diseases.
Collapse
Affiliation(s)
- Mei Wu
- Affiliated Hospital of Guangdong Medical University and Key Laboratory of Zebrafish Model for Development and Disease of Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Jin Xu
- South China University of Technology, School of Medicine, Guangzhou, Guangdong, China.
| | - Yiyue Zhang
- South China University of Technology, School of Medicine, Guangzhou, Guangdong, China.
| | - Zilong Wen
- Southern University of Science and Technology, School of Life Sciences, Shenzhen, Guangdong, China.
| |
Collapse
|
11
|
Lalonde RL, Kemmler CL, Riemslagh FW, Aman AJ, Kresoja-Rakic J, Moran HR, Nieuwenhuize S, Parichy DM, Burger A, Mosimann C. Heterogeneity and genomic loci of ubiquitous transgenic Cre reporter lines in zebrafish. Dev Dyn 2022; 251:1754-1773. [PMID: 35582941 PMCID: PMC10069295 DOI: 10.1002/dvdy.499] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Revised: 05/10/2022] [Accepted: 05/11/2022] [Indexed: 11/09/2022] Open
Abstract
BACKGROUND The most-common strategy for zebrafish Cre/lox-mediated lineage labeling experiments combines ubiquitously expressed, lox-based Switch reporter transgenes with tissue-specific Cre or 4-OH-Tamoxifen-inducible CreERT2 driver lines. Although numerous Cre driver lines have been produced, only a few broadly expressed Switch reporters exist in zebrafish and their generation by random transgene integration has been challenging due to position-effect sensitivity of the lox-flanked recombination cassettes. Here, we compare commonly used Switch reporter lines for their recombination efficiency and reporter expression pattern during zebrafish development. RESULTS Using different experimental setups, we show that ubi:Switch and hsp70l:Switch outperform current generations of the two additional Switch reporters actb2:BFP-DsRed and actb2:Stop-DsRed. Our comparisons also document preferential Cre-dependent recombination of ubi:Switch and hsp70l:Switch in distinct zebrafish tissues at early developmental stages. To investigate what genomic features may influence Cre accessibility and lox recombination efficiency in highly functional Switch lines, we mapped these transgenes and charted chromatin dynamics at their integration sites. CONCLUSIONS Our data documents the heterogeneity among lox-based Switch transgenes toward informing suitable transgene selection for lineage labeling experiments. Our work further proposes that ubi:Switch and hsp70l:Switch define genomic integration sites suitable for universal transgene or switch reporter knock-in in zebrafish.
Collapse
Affiliation(s)
- Robert L Lalonde
- Department of Pediatrics, Section of Developmental Biology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, Colorado, USA
| | - Cassie L Kemmler
- Department of Pediatrics, Section of Developmental Biology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, Colorado, USA
| | - Fréderike W Riemslagh
- Department of Pediatrics, Section of Developmental Biology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, Colorado, USA
| | - Andrew J Aman
- Department of Biology, University of Virginia, Charlottesville, Virginia, USA.,Department of Biology and Department of Cell Biology, University of Virginia, Charlottesville, Virginia, USA
| | - Jelena Kresoja-Rakic
- Department of Pediatrics, Section of Developmental Biology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, Colorado, USA
| | - Hannah R Moran
- Department of Pediatrics, Section of Developmental Biology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, Colorado, USA
| | - Susan Nieuwenhuize
- Department of Pediatrics, Section of Developmental Biology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, Colorado, USA
| | - David M Parichy
- Department of Biology, University of Virginia, Charlottesville, Virginia, USA.,Department of Biology and Department of Cell Biology, University of Virginia, Charlottesville, Virginia, USA
| | - Alexa Burger
- Department of Pediatrics, Section of Developmental Biology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, Colorado, USA
| | - Christian Mosimann
- Department of Pediatrics, Section of Developmental Biology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, Colorado, USA
| |
Collapse
|
12
|
The Annotation of Zebrafish Enhancer Trap Lines Generated with PB Transposon. Curr Issues Mol Biol 2022; 44:2614-2621. [PMID: 35735619 PMCID: PMC9221761 DOI: 10.3390/cimb44060178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 05/30/2022] [Accepted: 05/31/2022] [Indexed: 11/16/2022] Open
Abstract
An enhancer trap (ET) mediated by a transposon is an effective method for functional gene research. Here, an ET system based on a PB transposon that carries a mini Krt4 promoter (the keratin4 minimal promoter from zebrafish) and the green fluorescent protein gene (GFP) has been used to produce zebrafish ET lines. One enhancer trap line with eye-specific expression GFP named EYE was used to identify the trapped enhancers and genes. Firstly, GFP showed a temporal and spatial expression pattern with whole-embryo expression at 6, 12, and 24 hpf stages and eye-specific expression from 2 to 7 dpf. Then, the genome insertion sites were detected by splinkerette PCR (spPCR). The Krt4-GFP was inserted into the fourth intron of the gene itgav (integrin, alpha V) in chromosome 9 of the zebrafish genome, with the GFP direction the same as that of the itgav gene. By the alignment of homologous gene sequences in different species, three predicted endogenous enhancers were obtained. The trapped endogenous gene itgav, whose overexpression is related to hepatocellular carcinoma, showed a similar expression pattern as GFP detected by in situ hybridization, which suggested that GFP and itgav were possibly regulated by the same enhancers. In short, the zebrafish enhancer trap lines generated by the PB transposon-mediated enhancer trap technology in this study were valuable resources as visual markers to study the regulators and genes. This work provides an efficient method to identify and isolate tissue-specific enhancer sequences.
Collapse
|
13
|
Korzh VP, Gasanov EV. Genetics of Atavism. Russ J Dev Biol 2022. [DOI: 10.1134/s1062360422030043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Abstract
Atavisms have attracted people’s attention for a long time. First, atavisms excited their imagination and created fertile ground for myths and superstitions. With the development of science, atavisms became the subject of investigation, which soon provided evidence to support evolutionary theory. However, at the molecular level, the formation of atavisms remained insufficiently understood. Recent progress in comparative genomics and molecular developmental biology has helped in understanding the processes underlying the formation of one of the human atavisms: the vestigial tail.
Collapse
|
14
|
Single-cell transcriptome analysis reveals three sequential phases of gene expression during zebrafish sensory hair cell regeneration. Dev Cell 2022; 57:799-819.e6. [PMID: 35316618 PMCID: PMC9188816 DOI: 10.1016/j.devcel.2022.03.001] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 11/19/2021] [Accepted: 02/28/2022] [Indexed: 12/20/2022]
Abstract
Loss of sensory hair cells (HCs) in the mammalian inner ear leads to permanent hearing and vestibular defects, whereas loss of HCs in zebrafish results in their regeneration. We used single-cell RNA sequencing (scRNA-seq) to characterize the transcriptional dynamics of HC regeneration in zebrafish at unprecedented spatiotemporal resolution. We uncovered three sequentially activated modules: first, an injury/inflammatory response and downregulation of progenitor cell maintenance genes within minutes after HC loss; second, the transient activation of regeneration-specific genes; and third, a robust re-activation of developmental gene programs, including HC specification, cell-cycle activation, ribosome biogenesis, and a metabolic switch to oxidative phosphorylation. The results are relevant not only for our understanding of HC regeneration and how we might be able to trigger it in mammals but also for regenerative processes in general. The data are searchable and publicly accessible via a web-based interface.
Collapse
|
15
|
Corbacho J, Sanabria-Reinoso E, Buono L, Fernández-Miñan A, Martínez-Morales JR. Trap-TRAP, a Versatile Tool for Tissue-Specific Translatomics in Zebrafish. Front Cell Dev Biol 2022; 9:817191. [PMID: 35174174 PMCID: PMC8841413 DOI: 10.3389/fcell.2021.817191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Accepted: 12/24/2021] [Indexed: 11/13/2022] Open
Abstract
Developmental and physiological processes depend on the transcriptional and translational activity of heterogeneous cell populations. A main challenge in gene expression studies is dealing with this intrinsic complexity while keeping sequencing efficiency. Translating ribosome affinity purification (TRAP) methods have allowed cell-specific recovery of polyribosome-associated RNAs by genetic tagging of ribosomes in selected cell populations. Here we combined the TRAP approach with adapted enhancer trap methods (trap-TRAP) to systematically generate zebrafish transgenic lines suitable for tissue-specific translatome interrogation. Through the random integration of a GFP-tagged version of the large subunit ribosomal protein L10a (EGFP-Rpl10a), we have generated stable lines driving expression in a variety of tissues, including the retina, skeletal muscle, lateral line primordia, rhombomeres, or jaws. To increase the range of applications, a UAS:TRAP transgenic line compatible with available Gal4 lines was also generated and tested. The resulting collection of lines and applications constitutes a resource for the zebrafish community in developmental genetics, organ physiology and disease modelling.
Collapse
|
16
|
Luo Z, Guo S, Ho NY, Takamiya M, Strähle U, Yang L. Methylmercury-induced hair cell loss requires hydrogen peroxide production and leukocytes in zebrafish embryos. Toxicol Lett 2021; 356:151-160. [PMID: 34954246 DOI: 10.1016/j.toxlet.2021.12.012] [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: 08/24/2021] [Revised: 12/07/2021] [Accepted: 12/17/2021] [Indexed: 10/19/2022]
Abstract
Hearing impairment and deafness is frequently observed as one of the neurological signs in patients with Minamata disease caused by methylmercury (MeHg) poisoning. Loss of hair cells in humans and animals is a consequence of MeHg poisoning. However, it is still not clear how MeHg causes hearing deficits. We employed the hair cells of the lateral line system of zebrafish embryos as a model to explore this question. We exposed transgenic zebrafish embryos to MeHg (30-360 μg/L) at the different stages, and scored the numbers of hair cells. We find that MeHg-induced reduction of hair cells is in a concentration dependent manner. By employing antisense morpholino against to pu.1, we confirm that loss of hair cells involves the action of leukocytes. Moreover, hair cell loss is attenuated by co-treating MeHg-exposed embryos with pharmacological inhibitors of NADPH oxidases named diphenyleneiodonium (DPI) and VAS2870. In situ gene expression analysis showed that genes encoding the SQSTM1-Keap1-Nrf2 systems involved in combating oxidative stress and immune responses are highly expressed in the lateral line organs of embryos exposed to MeHg. This suggests that induction of hydrogen peroxide (H2O2) is the primary effect of MeHg on the hair cells. Genes induced by MeHg are also involved in regeneration of the hair cells. These features are likely related to the capacity of the zebrafish to regenerate the lost hair cells.
Collapse
Affiliation(s)
- Zidie Luo
- State Key Laboratory of Environmental Criteria and Risk Assessment, Chinese Research Academy of Environmental Sciences, 100012, Beijing, China
| | - Shaojuan Guo
- State Key Laboratory of Environmental Criteria and Risk Assessment, Chinese Research Academy of Environmental Sciences, 100012, Beijing, China
| | - Nga Yu Ho
- Institute of Biological and Chemical Systems-Biological Information Processing, Karlsruhe Institute of Technology (KIT), Karlsruhe, Germany
| | - Masanari Takamiya
- Institute of Biological and Chemical Systems-Biological Information Processing, Karlsruhe Institute of Technology (KIT), Karlsruhe, Germany
| | - Uwe Strähle
- Institute of Biological and Chemical Systems-Biological Information Processing, Karlsruhe Institute of Technology (KIT), Karlsruhe, Germany
| | - Lixin Yang
- State Key Laboratory of Environmental Criteria and Risk Assessment, Chinese Research Academy of Environmental Sciences, 100012, Beijing, China.
| |
Collapse
|
17
|
Minhas R, Loeffler-Wirth H, Siddiqui YH, Obrębski T, Vashisht S, Abu Nahia K, Paterek A, Brzozowska A, Bugajski L, Piwocka K, Korzh V, Binder H, Winata CL. Transcriptome profile of the sinoatrial ring reveals conserved and novel genetic programs of the zebrafish pacemaker. BMC Genomics 2021; 22:715. [PMID: 34600492 PMCID: PMC8487553 DOI: 10.1186/s12864-021-08016-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Accepted: 09/16/2021] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Sinoatrial Node (SAN) is part of the cardiac conduction system, which controls the rhythmic contraction of the vertebrate heart. The SAN consists of a specialized pacemaker cell population that has the potential to generate electrical impulses. Although the SAN pacemaker has been extensively studied in mammalian and teleost models, including the zebrafish, their molecular nature remains inadequately comprehended. RESULTS To characterize the molecular profile of the zebrafish sinoatrial ring (SAR) and elucidate the mechanism of pacemaker function, we utilized the transgenic line sqet33mi59BEt to isolate cells of the SAR of developing zebrafish embryos and profiled their transcriptome. Our analyses identified novel candidate genes and well-known conserved signaling pathways involved in pacemaker development. We show that, compared to the rest of the heart, the zebrafish SAR overexpresses several mammalian SAN pacemaker signature genes, which include hcn4 as well as those encoding calcium- and potassium-gated channels. Moreover, genes encoding components of the BMP and Wnt signaling pathways, as well as members of the Tbx family, which have previously been implicated in pacemaker development, were also overexpressed in the SAR. Among SAR-overexpressed genes, 24 had human homologues implicated in 104 different ClinVar phenotype entries related to various forms of congenital heart diseases, which suggest the relevance of our transcriptomics resource to studying human heart conditions. Finally, functional analyses of three SAR-overexpressed genes, pard6a, prom2, and atp1a1a.2, uncovered their novel role in heart development and physiology. CONCLUSION Our results established conserved aspects between zebrafish and mammalian pacemaker function and revealed novel factors implicated in maintaining cardiac rhythm. The transcriptome data generated in this study represents a unique and valuable resource for the study of pacemaker function and associated heart diseases.
Collapse
Affiliation(s)
- Rashid Minhas
- International Institute of Molecular and Cell Biology in Warsaw, Warsaw, Poland
- Randall Centre of Cell and Molecular Biophysics, Faculty of Life Sciences and Medicine, King's College London, London, UK
| | - Henry Loeffler-Wirth
- Interdisciplinary Centre for Bioinformatics, University of Leipzig, Leipzig, Germany
| | - Yusra H Siddiqui
- International Institute of Molecular and Cell Biology in Warsaw, Warsaw, Poland
- School of Human Sciences, College of Science and Engineering, University of Derby, Derby, UK
| | - Tomasz Obrębski
- International Institute of Molecular and Cell Biology in Warsaw, Warsaw, Poland
| | - Shikha Vashisht
- International Institute of Molecular and Cell Biology in Warsaw, Warsaw, Poland
| | - Karim Abu Nahia
- International Institute of Molecular and Cell Biology in Warsaw, Warsaw, Poland
| | - Alexandra Paterek
- International Institute of Molecular and Cell Biology in Warsaw, Warsaw, Poland
| | - Angelika Brzozowska
- International Institute of Molecular and Cell Biology in Warsaw, Warsaw, Poland
| | - Lukasz Bugajski
- Nencki Institute of Experimental Biology, Laboratory of Cytometry, Warsaw, Poland
| | - Katarzyna Piwocka
- Nencki Institute of Experimental Biology, Laboratory of Cytometry, Warsaw, Poland
| | - Vladimir Korzh
- International Institute of Molecular and Cell Biology in Warsaw, Warsaw, Poland
| | - Hans Binder
- Interdisciplinary Centre for Bioinformatics, University of Leipzig, Leipzig, Germany
| | - Cecilia Lanny Winata
- International Institute of Molecular and Cell Biology in Warsaw, Warsaw, Poland.
| |
Collapse
|
18
|
Abu Nahia K, Migdał M, Quinn TA, Poon KL, Łapiński M, Sulej A, Liu J, Mondal SS, Pawlak M, Bugajski Ł, Piwocka K, Brand T, Kohl P, Korzh V, Winata C. Genomic and physiological analyses of the zebrafish atrioventricular canal reveal molecular building blocks of the secondary pacemaker region. Cell Mol Life Sci 2021; 78:6669-6687. [PMID: 34557935 PMCID: PMC8558220 DOI: 10.1007/s00018-021-03939-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 08/06/2021] [Accepted: 09/10/2021] [Indexed: 01/06/2023]
Abstract
The atrioventricular canal (AVC) is the site where key structures responsible for functional division between heart regions are established, most importantly, the atrioventricular (AV) conduction system and cardiac valves. To elucidate the mechanism underlying AVC development and function, we utilized transgenic zebrafish line sqet31Et expressing EGFP in the AVC to isolate this cell population and profile its transcriptome at 48 and 72 hpf. The zebrafish AVC transcriptome exhibits hallmarks of mammalian AV node, including the expression of genes implicated in its development and those encoding connexins forming low conductance gap junctions. Transcriptome analysis uncovered protein-coding and noncoding transcripts enriched in AVC, which have not been previously associated with this structure, as well as dynamic expression of epithelial-to-mesenchymal transition markers and components of TGF-β, Notch, and Wnt signaling pathways likely reflecting ongoing AVC and valve development. Using transgenic line Tg(myl7:mermaid) encoding voltage-sensitive fluorescent protein, we show that abolishing the pacemaker-containing sinoatrial ring (SAR) through Isl1 loss of function resulted in spontaneous activation in the AVC region, suggesting that it possesses inherent automaticity although insufficient to replace the SAR. The SAR and AVC transcriptomes express partially overlapping species of ion channels and gap junction proteins, reflecting their distinct roles. Besides identifying conserved aspects between zebrafish and mammalian conduction systems, our results established molecular hallmarks of the developing AVC which underlies its role in structural and electrophysiological separation between heart chambers. This data constitutes a valuable resource for studying AVC development and function, and identification of novel candidate genes implicated in these processes.
Collapse
Affiliation(s)
- Karim Abu Nahia
- International Institute of Molecular and Cell Biology in Warsaw, Warsaw, Poland
| | - Maciej Migdał
- International Institute of Molecular and Cell Biology in Warsaw, Warsaw, Poland
| | - T Alexander Quinn
- Department of Physiology and Biophysics, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Kar-Lai Poon
- Institute of Molecular and Cell Biology, 61 Biopolis Dr, Singapore , Singapore.,Developmental Dynamics, National Heart and Lung Institute, Imperial College London, London, UK
| | - Maciej Łapiński
- International Institute of Molecular and Cell Biology in Warsaw, Warsaw, Poland
| | - Agata Sulej
- International Institute of Molecular and Cell Biology in Warsaw, Warsaw, Poland
| | - Jiandong Liu
- McAllister Heart Institute, University of North Carolina, Chapel Hill, USA
| | - Shamba S Mondal
- International Institute of Molecular and Cell Biology in Warsaw, Warsaw, Poland
| | - Michał Pawlak
- International Institute of Molecular and Cell Biology in Warsaw, Warsaw, Poland
| | | | | | - Thomas Brand
- Developmental Dynamics, National Heart and Lung Institute, Imperial College London, London, UK
| | - Peter Kohl
- Institute for Experimental Cardiovascular Medicine, University Heart Centre, Faculty of Medicine, and Faculty of Engineering, University of Freiburg, Freiburg im Breisgau, Germany
| | - Vladimir Korzh
- International Institute of Molecular and Cell Biology in Warsaw, Warsaw, Poland.
| | - Cecilia Winata
- International Institute of Molecular and Cell Biology in Warsaw, Warsaw, Poland.
| |
Collapse
|
19
|
Vandestadt C, Vanwalleghem GC, Khabooshan MA, Douek AM, Castillo HA, Li M, Schulze K, Don E, Stamatis SA, Ratnadiwakara M, Änkö ML, Scott EK, Kaslin J. RNA-induced inflammation and migration of precursor neurons initiates neuronal circuit regeneration in zebrafish. Dev Cell 2021; 56:2364-2380.e8. [PMID: 34428400 DOI: 10.1016/j.devcel.2021.07.021] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Revised: 06/18/2021] [Accepted: 07/27/2021] [Indexed: 11/29/2022]
Abstract
Tissue regeneration and functional restoration after injury are considered as stem- and progenitor-cell-driven processes. In the central nervous system, stem cell-driven repair is slow and problematic because function needs to be restored rapidly for vital tasks. In highly regenerative vertebrates, such as zebrafish, functional recovery is rapid, suggesting a capability for fast cell production and functional integration. Surprisingly, we found that migration of dormant "precursor neurons" to the injury site pioneers functional circuit regeneration after spinal cord injury and controls the subsequent stem-cell-driven repair response. Thus, the precursor neurons make do before the stem cells make new. Furthermore, RNA released from the dying or damaged cells at the site of injury acts as a signal to attract precursor neurons for repair. Taken together, our data demonstrate an unanticipated role of neuronal migration and RNA as drivers of neural repair.
Collapse
Affiliation(s)
- Celia Vandestadt
- Australian Regenerative Medicine Institute, Monash University, Clayton VIC, 3800, Australia
| | - Gilles C Vanwalleghem
- The Queensland Brain Institute, the University of Queensland, St. Lucia, QLD, Australia
| | - Mitra Amiri Khabooshan
- Australian Regenerative Medicine Institute, Monash University, Clayton VIC, 3800, Australia
| | - Alon M Douek
- Australian Regenerative Medicine Institute, Monash University, Clayton VIC, 3800, Australia
| | - Hozana Andrade Castillo
- Australian Regenerative Medicine Institute, Monash University, Clayton VIC, 3800, Australia; Brazilian Biosciences National Laboratory, Brazilian Centre for Research in Energy and Materials, Campinas CEP 13083-100, Brazil
| | - Mei Li
- Australian Regenerative Medicine Institute, Monash University, Clayton VIC, 3800, Australia
| | - Keith Schulze
- Monash Micro Imaging, Monash University, Monash University, Clayton, VIC 3800, Australia
| | - Emily Don
- Centre for Motor Neuron Disease Research, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Macquarie University, Macquarie Park, NSW 2109, Australia
| | | | - Madara Ratnadiwakara
- Centre for Reproductive Health and Center for Cancer Research, Hudson Institute of Medical Research, Clayton, VIC 3168, Australia
| | - Minna-Liisa Änkö
- Centre for Reproductive Health and Center for Cancer Research, Hudson Institute of Medical Research, Clayton, VIC 3168, Australia; Department of Molecular and Translational Sciences, Monash University, Clayton, VIC 3800, Australia
| | - Ethan K Scott
- The Queensland Brain Institute, the University of Queensland, St. Lucia, QLD, Australia
| | - Jan Kaslin
- Australian Regenerative Medicine Institute, Monash University, Clayton VIC, 3800, Australia.
| |
Collapse
|
20
|
Wilson MH, Ekker SC, Farber SA. Imaging cytoplasmic lipid droplets in vivo with fluorescent perilipin 2 and perilipin 3 knock-in zebrafish. eLife 2021; 10:e66393. [PMID: 34387191 PMCID: PMC8460263 DOI: 10.7554/elife.66393] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Accepted: 08/10/2021] [Indexed: 12/26/2022] Open
Abstract
Cytoplasmic lipid droplets are highly dynamic storage organelles that are critical for cellular lipid homeostasis. While the molecular details of lipid droplet dynamics are a very active area of investigation, this work has been primarily performed in cultured cells. Taking advantage of the powerful transgenic and in vivo imaging opportunities available in zebrafish, we built a suite of tools to study lipid droplets in real time from the subcellular to the whole organism level. Fluorescently tagging the lipid droplet-associated proteins, perilipin 2 and perilipin 3, in the endogenous loci permits visualization of lipid droplets in the intestine, liver, and adipose tissue. Using these tools, we found that perilipin 3 is rapidly loaded on intestinal lipid droplets following a high-fat meal and later replaced by perilipin 2. These powerful new tools will facilitate studies on the role of lipid droplets in different tissues, under different genetic and physiological manipulations, and in a variety of human disease models.
Collapse
Affiliation(s)
- Meredith H Wilson
- Carnegie Institution for Science Department of EmbryologyBaltimoreUnited States
| | - Stephen C Ekker
- Department of Biochemistry and Molecular Biology, Mayo ClinicRochesterUnited States
| | - Steven A Farber
- Carnegie Institution for Science Department of EmbryologyBaltimoreUnited States
- Johns Hopkins University Department of BiologyBaltimoreUnited States
| |
Collapse
|
21
|
Aman AJ, Kim M, Saunders LM, Parichy DM. Thyroid hormone regulates abrupt skin morphogenesis during zebrafish postembryonic development. Dev Biol 2021; 477:205-218. [PMID: 34089732 PMCID: PMC10069294 DOI: 10.1016/j.ydbio.2021.05.019] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 05/28/2021] [Accepted: 05/29/2021] [Indexed: 10/21/2022]
Abstract
Thyroid hormone is a key regulator of post-embryonic vertebrate development. Skin is a biomedically important thyroid hormone target organ, but the cellular and molecular mechanisms underlying skin pathologies associated with thyroid dysfunction remain obscure. The transparent skin of zebrafish is an accessible model system for studying vertebrate skin development. During post-embryonic development of the zebrafish, scales emerge in the skin from a hexagonally patterned array of dermal papillae, like other vertebrate skin appendages such as feathers and hair follicles. We show here that thyroid hormone regulates the rate of post-embryonic dermal development through interaction with nuclear hormone receptors. This couples skin development with body growth to generate a well ordered array of correctly proportioned scales. This work extends our knowledge of thyroid hormone actions on skin by providing in-vivo evidence that thyroid hormone regulates multiple aspects of dermal development.
Collapse
Affiliation(s)
- Andrew J Aman
- Department of Biology, University of Virginia, Charlottesville, VA, 22903, USA
| | - Margaret Kim
- Department of Biology, University of Virginia, Charlottesville, VA, 22903, USA
| | - Lauren M Saunders
- Department of Genome Sciences, University of Washington, Seattle, WA, 98195, USA
| | - David M Parichy
- Department of Biology, University of Virginia, Charlottesville, VA, 22903, USA; Department of Cell Biology, University of Virginia, Charlottesville, VA, 22903, USA.
| |
Collapse
|
22
|
Sandoval-Villegas N, Nurieva W, Amberger M, Ivics Z. Contemporary Transposon Tools: A Review and Guide through Mechanisms and Applications of Sleeping Beauty, piggyBac and Tol2 for Genome Engineering. Int J Mol Sci 2021; 22:ijms22105084. [PMID: 34064900 PMCID: PMC8151067 DOI: 10.3390/ijms22105084] [Citation(s) in RCA: 55] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 04/30/2021] [Accepted: 05/05/2021] [Indexed: 01/19/2023] Open
Abstract
Transposons are mobile genetic elements evolved to execute highly efficient integration of their genes into the genomes of their host cells. These natural DNA transfer vehicles have been harnessed as experimental tools for stably introducing a wide variety of foreign DNA sequences, including selectable marker genes, reporters, shRNA expression cassettes, mutagenic gene trap cassettes, and therapeutic gene constructs into the genomes of target cells in a regulated and highly efficient manner. Given that transposon components are typically supplied as naked nucleic acids (DNA and RNA) or recombinant protein, their use is simple, safe, and economically competitive. Thus, transposons enable several avenues for genome manipulations in vertebrates, including transgenesis for the generation of transgenic cells in tissue culture comprising the generation of pluripotent stem cells, the production of germline-transgenic animals for basic and applied research, forward genetic screens for functional gene annotation in model species and therapy of genetic disorders in humans. This review describes the molecular mechanisms involved in transposition reactions of the three most widely used transposon systems currently available (Sleeping Beauty, piggyBac, and Tol2), and discusses the various parameters and considerations pertinent to their experimental use, highlighting the state-of-the-art in transposon technology in diverse genetic applications.
Collapse
Affiliation(s)
| | | | | | - Zoltán Ivics
- Correspondence: ; Tel.: +49-6103-77-6000; Fax: +49-6103-77-1280
| |
Collapse
|
23
|
Peloggia J, Münch D, Meneses-Giles P, Romero-Carvajal A, Lush ME, Lawson ND, McClain M, Pan YA, Piotrowski T. Adaptive cell invasion maintains lateral line organ homeostasis in response to environmental changes. Dev Cell 2021; 56:1296-1312.e7. [PMID: 33878346 DOI: 10.1016/j.devcel.2021.03.027] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Revised: 02/21/2021] [Accepted: 03/24/2021] [Indexed: 10/21/2022]
Abstract
Mammalian inner ear and fish lateral line sensory hair cells (HCs) detect fluid motion to transduce environmental signals. Actively maintained ionic homeostasis of the mammalian inner ear endolymph is essential for HC function. In contrast, fish lateral line HCs are exposed to the fluctuating ionic composition of the aqueous environment. Using lineage labeling, in vivo time-lapse imaging and scRNA-seq, we discovered highly motile skin-derived cells that invade mature mechanosensory organs of the zebrafish lateral line and differentiate into Neuromast-associated (Nm) ionocytes. This invasion is adaptive as it is triggered by environmental fluctuations. Our discovery of Nm ionocytes challenges the notion of an entirely placodally derived lateral line and identifies Nm ionocytes as likely regulators of HC function possibly by modulating the ionic microenvironment. Nm ionocytes provide an experimentally accessible in vivo system to study cell invasion and migration, as well as the physiological adaptation of vertebrate organs to changing environmental conditions.
Collapse
Affiliation(s)
- Julia Peloggia
- Stowers Institute for Medical Research, Kansas City, MO 64110, USA
| | - Daniela Münch
- Stowers Institute for Medical Research, Kansas City, MO 64110, USA
| | | | - Andrés Romero-Carvajal
- Stowers Institute for Medical Research, Kansas City, MO 64110, USA; Pontificia Universidad Católica del Ecuador, Escuela de Ciencias Biológicas, Quito, Ecuador
| | - Mark E Lush
- Stowers Institute for Medical Research, Kansas City, MO 64110, USA
| | - Nathan D Lawson
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester 01605, USA
| | - Melainia McClain
- Stowers Institute for Medical Research, Kansas City, MO 64110, USA
| | - Y Albert Pan
- Center for Neurobiology Research, Fralin Biomedical Research Institute at Virginia Tech Carilion, Virginia Tech, Roanoke, VA 24016, USA; Department of Biomedical Sciences and Pathobiology, Virginia-Maryland Regional College of Veterinary Medicine, Virginia Tech, Blacksburg, VA 24060, USA; Department of Psychiatry and Behavioral Medicine, Virginia Tech Carilion School of Medicine, Roanoke, VA 24016, USA
| | | |
Collapse
|
24
|
Parisi C, Vashisht S, Winata CL. Fish-Ing for Enhancers in the Heart. Int J Mol Sci 2021; 22:3914. [PMID: 33920121 PMCID: PMC8069060 DOI: 10.3390/ijms22083914] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 04/07/2021] [Accepted: 04/08/2021] [Indexed: 12/19/2022] Open
Abstract
Precise control of gene expression is crucial to ensure proper development and biological functioning of an organism. Enhancers are non-coding DNA elements which play an essential role in regulating gene expression. They contain specific sequence motifs serving as binding sites for transcription factors which interact with the basal transcription machinery at their target genes. Heart development is regulated by intricate gene regulatory network ensuring precise spatiotemporal gene expression program. Mutations affecting enhancers have been shown to result in devastating forms of congenital heart defect. Therefore, identifying enhancers implicated in heart biology and understanding their mechanism is key to improve diagnosis and therapeutic options. Despite their crucial role, enhancers are poorly studied, mainly due to a lack of reliable way to identify them and determine their function. Nevertheless, recent technological advances have allowed rapid progress in enhancer discovery. Model organisms such as the zebrafish have contributed significant insights into the genetics of heart development through enabling functional analyses of genes and their regulatory elements in vivo. Here, we summarize the current state of knowledge on heart enhancers gained through studies in model organisms, discuss various approaches to discover and study their function, and finally suggest methods that could further advance research in this field.
Collapse
Affiliation(s)
- Costantino Parisi
- International Institute of Molecular and Cell Biology in Warsaw, 02-109 Warsaw, Poland; (C.P.); (S.V.)
| | - Shikha Vashisht
- International Institute of Molecular and Cell Biology in Warsaw, 02-109 Warsaw, Poland; (C.P.); (S.V.)
| | - Cecilia Lanny Winata
- International Institute of Molecular and Cell Biology in Warsaw, 02-109 Warsaw, Poland; (C.P.); (S.V.)
- Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
| |
Collapse
|
25
|
Yang S, Emelyanov A, You MS, Sin M, Korzh V. Camel regulates development of the brain ventricular system. Cell Tissue Res 2021; 383:835-852. [PMID: 32902807 PMCID: PMC7904751 DOI: 10.1007/s00441-020-03270-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Accepted: 07/29/2020] [Indexed: 10/25/2022]
Abstract
Development of the brain ventricular system of vertebrates and the molecular mechanisms involved are not fully understood. The developmental genes expressed in the elements of the brain ventricular system such as the ependyma and circumventricular organs act as molecular determinants of cell adhesion critical for the formation of brain ventricular system. They control brain development and function, including the flow of cerebrospinal fluid. Here, we describe the novel distantly related member of the zebrafish L1-CAM family of genes-camel. Whereas its maternal transcripts distributed uniformly, the zygotic transcripts demonstrate clearly defined expression patterns, in particular in the axial structures: floor plate, hypochord, and roof plate. camel expresses in several other cell lineages with access to the brain ventricular system, including the midbrain roof plate, subcommissural organ, organum vasculosum lamina terminalis, median eminence, paraventricular organ, flexural organ, and inter-rhombomeric boundaries. This expression pattern suggests a role of Camel in neural development. Several isoforms of Camel generated by differential splicing of exons encoding the sixth fibronectin type III domain enhance cell adhesion differentially. The antisense oligomer morpholino-mediated loss-of-function of Camel affects cell adhesion and causes hydrocephalus and scoliosis manifested via the tail curled down phenotype. The subcommissural organ's derivative-the Reissner fiber-participates in the flow of cerebrospinal fluid. The Reissner fiber fails to form upon morpholino-mediated Camel loss-of-function. The Camel mRNA-mediated gain-of-function causes the Reissner fiber misdirection. This study revealed a link between Chl1a/Camel and Reissner fiber formation, and this supports the idea that CHL1 is one of the scoliosis factors.
Collapse
Affiliation(s)
- Shulan Yang
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore, Singapore
- Translational Medicine Centre, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Alexander Emelyanov
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore, Singapore
- Institute for Research on Cancer and Aging, Nice, France
| | - May-Su You
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore, Singapore
- National Health Research Institutes, Zhunan, Taiwan
| | - Melvin Sin
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore, Singapore
| | - Vladimir Korzh
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore, Singapore.
- International Institute of Molecular and Cell Biology, Warsaw, Poland.
| |
Collapse
|
26
|
Is zebrafish heart regeneration "complete"? Lineage-restricted cardiomyocytes proliferate to pre-injury numbers but some fail to differentiate in fibrotic hearts. Dev Biol 2020; 471:106-118. [PMID: 33309949 DOI: 10.1016/j.ydbio.2020.12.004] [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: 08/26/2020] [Revised: 11/13/2020] [Accepted: 12/03/2020] [Indexed: 12/28/2022]
Abstract
Adult zebrafish are frequently described to be able to "completely" regenerate the heart. Yet, the extent to which cardiomyocytes lost to injury are replaced is unknown, since existing evidence for cardiomyocyte proliferation is indirect or non-quantitative. We established stereological methods to quantify the number of cardiomyocytes at several time-points post cryoinjury. Intriguingly, after cryoinjuries that killed about 1/3 of the ventricular cardiomyocytes, pre-injury cardiomyocyte numbers were restored already within 30 days. Yet, many hearts retained small residual scars, and a subset of cardiomyocytes bordering these fibrotic areas remained smaller, lacked differentiated sarcomeric structures, and displayed defective calcium signaling. Thus, a subset of regenerated cardiomyocytes failed to fully mature. While lineage-tracing experiments have shown that regenerating cardiomyocytes are derived from differentiated cardiomyocytes, technical limitations have previously made it impossible to test whether cardiomyocyte trans-differentiation contributes to regeneration of non-myocyte cell lineages. Using Cre responder lines that are expressed in all major cell types of the heart, we found no evidence for cardiomyocyte transdifferentiation into endothelial, epicardial, fibroblast or immune cell lineages. Overall, our results imply a refined answer to the question whether zebrafish can completely regenerate the heart: in response to cryoinjury, preinjury cardiomyocyte numbers are indeed completely regenerated by proliferation of lineage-restricted cardiomyocytes, while restoration of cardiomyocyte differentiation and function, as well as resorption of scar tissue, is less robustly achieved.
Collapse
|
27
|
Tobias IC, Abatti LE, Moorthy SD, Mullany S, Taylor T, Khader N, Filice MA, Mitchell JA. Transcriptional enhancers: from prediction to functional assessment on a genome-wide scale. Genome 2020; 64:426-448. [PMID: 32961076 DOI: 10.1139/gen-2020-0104] [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] [Indexed: 12/20/2022]
Abstract
Enhancers are cis-regulatory sequences located distally to target genes. These sequences consolidate developmental and environmental cues to coordinate gene expression in a tissue-specific manner. Enhancer function and tissue specificity depend on the expressed set of transcription factors, which recognize binding sites and recruit cofactors that regulate local chromatin organization and gene transcription. Unlike other genomic elements, enhancers are challenging to identify because they function independently of orientation, are often distant from their promoters, have poorly defined boundaries, and display no reading frame. In addition, there are no defined genetic or epigenetic features that are unambiguously associated with enhancer activity. Over recent years there have been developments in both empirical assays and computational methods for enhancer prediction. We review genome-wide tools, CRISPR advancements, and high-throughput screening approaches that have improved our ability to both observe and manipulate enhancers in vitro at the level of primary genetic sequences, chromatin states, and spatial interactions. We also highlight contemporary animal models and their importance to enhancer validation. Together, these experimental systems and techniques complement one another and broaden our understanding of enhancer function in development, evolution, and disease.
Collapse
Affiliation(s)
- Ian C Tobias
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON, M5S 3G5, Canada.,Department of Cell and Systems Biology, University of Toronto, Toronto, ON, M5S 3G5, Canada
| | - Luis E Abatti
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON, M5S 3G5, Canada.,Department of Cell and Systems Biology, University of Toronto, Toronto, ON, M5S 3G5, Canada
| | - Sakthi D Moorthy
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON, M5S 3G5, Canada.,Department of Cell and Systems Biology, University of Toronto, Toronto, ON, M5S 3G5, Canada
| | - Shanelle Mullany
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON, M5S 3G5, Canada.,Department of Cell and Systems Biology, University of Toronto, Toronto, ON, M5S 3G5, Canada
| | - Tiegh Taylor
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON, M5S 3G5, Canada.,Department of Cell and Systems Biology, University of Toronto, Toronto, ON, M5S 3G5, Canada
| | - Nawrah Khader
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON, M5S 3G5, Canada.,Department of Cell and Systems Biology, University of Toronto, Toronto, ON, M5S 3G5, Canada
| | - Mario A Filice
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON, M5S 3G5, Canada.,Department of Cell and Systems Biology, University of Toronto, Toronto, ON, M5S 3G5, Canada
| | - Jennifer A Mitchell
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON, M5S 3G5, Canada.,Department of Cell and Systems Biology, University of Toronto, Toronto, ON, M5S 3G5, Canada
| |
Collapse
|
28
|
Ichino N, Serres MR, Urban RM, Urban MD, Treichel AJ, Schaefbauer KJ, Greif LE, Varshney GK, Skuster KJ, McNulty MS, Daby CL, Wang Y, Liao HK, El-Rass S, Ding Y, Liu W, Anderson JL, Wishman MD, Sabharwal A, Schimmenti LA, Sivasubbu S, Balciunas D, Hammerschmidt M, Farber SA, Wen XY, Xu X, McGrail M, Essner JJ, Burgess SM, Clark KJ, Ekker SC. Building the vertebrate codex using the gene breaking protein trap library. eLife 2020; 9:54572. [PMID: 32779569 PMCID: PMC7486118 DOI: 10.7554/elife.54572] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Accepted: 08/07/2020] [Indexed: 12/14/2022] Open
Abstract
One key bottleneck in understanding the human genome is the relative under-characterization of 90% of protein coding regions. We report a collection of 1200 transgenic zebrafish strains made with the gene-break transposon (GBT) protein trap to simultaneously report and reversibly knockdown the tagged genes. Protein trap-associated mRFP expression shows previously undocumented expression of 35% and 90% of cloned genes at 2 and 4 days post-fertilization, respectively. Further, investigated alleles regularly show 99% gene-specific mRNA knockdown. Homozygous GBT animals in ryr1b, fras1, tnnt2a, edar and hmcn1 phenocopied established mutants. 204 cloned lines trapped diverse proteins, including 64 orthologs of human disease-associated genes with 40 as potential new disease models. Severely reduced skeletal muscle Ca2+ transients in GBT ryr1b homozygous animals validated the ability to explore molecular mechanisms of genetic diseases. This GBT system facilitates novel functional genome annotation towards understanding cellular and molecular underpinnings of vertebrate biology and human disease.
Collapse
Affiliation(s)
- Noriko Ichino
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, United States
| | - MaKayla R Serres
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, United States
| | - Rhianna M Urban
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, United States
| | - Mark D Urban
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, United States
| | - Anthony J Treichel
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, United States
| | - Kyle J Schaefbauer
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, United States
| | - Lauren E Greif
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, United States
| | - Gaurav K Varshney
- Translational and Functional Genomics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, United States.,Functional & Chemical Genomics Program, Oklahoma Medical Research Foundation, Oklahoma City, United States
| | - Kimberly J Skuster
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, United States
| | - Melissa S McNulty
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, United States
| | - Camden L Daby
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, United States
| | - Ying Wang
- Department of Genetics, Development and Cell Biology, Iowa State University, Ames, United States
| | - Hsin-Kai Liao
- Department of Genetics, Development and Cell Biology, Iowa State University, Ames, United States
| | - Suzan El-Rass
- Zebrafish Centre for Advanced Drug Discovery & Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute, St. Michael's Hospital, Unity Health Toronto & University of Toronto, Toronto, Canada
| | - Yonghe Ding
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, United States.,Department of Cardiovascular Medicine, Mayo Clinic, Rochester, United States
| | - Weibin Liu
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, United States.,Department of Cardiovascular Medicine, Mayo Clinic, Rochester, United States
| | - Jennifer L Anderson
- Department of Embryology, Carnegie Institution for Science, Baltimore, United States
| | - Mark D Wishman
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, United States
| | - Ankit Sabharwal
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, United States
| | - Lisa A Schimmenti
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, United States.,Department of Clinical Genomics, Mayo Clinic, Rochester, United States.,Department of Otorhinolaryngology, Mayo Clinic, Rochester, United States
| | - Sridhar Sivasubbu
- Genomics and Molecular Medicine Unit, CSIR-Institute of Genomics and Integrative Biology, Delhi, India
| | - Darius Balciunas
- Department of Biology, Temple University, Philadelphia, United States
| | - Matthias Hammerschmidt
- Institute of Zoology, Developmental Biology Unit, University of Cologne, Cologne, Germany
| | - Steven Arthur Farber
- Department of Embryology, Carnegie Institution for Science, Baltimore, United States
| | - Xiao-Yan Wen
- Zebrafish Centre for Advanced Drug Discovery & Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute, St. Michael's Hospital, Unity Health Toronto & University of Toronto, Toronto, Canada
| | - Xiaolei Xu
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, United States.,Department of Cardiovascular Medicine, Mayo Clinic, Rochester, United States
| | - Maura McGrail
- Department of Genetics, Development and Cell Biology, Iowa State University, Ames, United States
| | - Jeffrey J Essner
- Department of Genetics, Development and Cell Biology, Iowa State University, Ames, United States
| | - Shawn M Burgess
- Translational and Functional Genomics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, United States
| | - Karl J Clark
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, United States
| | - Stephen C Ekker
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, United States
| |
Collapse
|
29
|
Ye Z, Su Z, Xie S, Liu Y, Wang Y, Xu X, Zheng Y, Zhao M, Jiang L. Yap-lin28a axis targets let7-Wnt pathway to restore progenitors for initiating regeneration. eLife 2020; 9:55771. [PMID: 32352377 PMCID: PMC7250571 DOI: 10.7554/elife.55771] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Accepted: 04/29/2020] [Indexed: 12/12/2022] Open
Abstract
The sox2 expressing (sox2+) progenitors in adult mammalian inner ear lose the capacity to regenerate while progenitors in the zebrafish lateral line are able to proliferate and regenerate damaged HCs throughout lifetime. To mimic the HC damage in mammals, we have established a zebrafish severe injury model to eliminate both progenitors and HCs. The atoh1a expressing (atoh1a+) HC precursors were the main population that survived post severe injury, and gained sox2 expression to initiate progenitor regeneration. In response to severe injury, yap was activated to upregulate lin28a transcription. Severe-injury-induced progenitor regeneration was disabled in lin28a or yap mutants. In contrary, overexpression of lin28a initiated the recovery of sox2+ progenitors. Mechanistically, microRNA let7 acted downstream of lin28a to activate Wnt pathway for promoting regeneration. Our findings that lin28a is necessary and sufficient to regenerate the exhausted sox2+ progenitors shed light on restoration of progenitors to initiate HC regeneration in mammals.
Collapse
Affiliation(s)
- Zhian Ye
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Zhongwu Su
- Department of Otolaryngology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Siyu Xie
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China.,Key Laboratory of Stem Cells and Tissue Engineering, Zhongshan School of Medicine, Sun Yat-Sen University, Ministry of Education, Guangzhou, China
| | - Yuye Liu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Yongqiang Wang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Xi Xu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China.,Key Laboratory of Stem Cells and Tissue Engineering, Zhongshan School of Medicine, Sun Yat-Sen University, Ministry of Education, Guangzhou, China
| | - Yiqing Zheng
- Department of Otolaryngology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Meng Zhao
- Key Laboratory of Stem Cells and Tissue Engineering, Zhongshan School of Medicine, Sun Yat-Sen University, Ministry of Education, Guangzhou, China
| | - Linjia Jiang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| |
Collapse
|
30
|
Pei W, Xu L, Chen Z, Slevin CC, Pettie KP, Wincovitch S, Burgess SM. A subset of SMN complex members have a specific role in tissue regeneration via ERBB pathway-mediated proliferation. NPJ Regen Med 2020; 5:6. [PMID: 32218991 PMCID: PMC7096462 DOI: 10.1038/s41536-020-0089-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Accepted: 01/27/2020] [Indexed: 12/24/2022] Open
Abstract
Spinal muscular atrophy (SMA) is the most common genetic disease in children. SMA is generally caused by mutations in the gene SMN1. The survival of motor neurons (SMN) complex consists of SMN1, Gemins (2-8), and Strap/Unrip. We previously demonstrated smn1 and gemin5 inhibited tissue regeneration in zebrafish. Here we investigated each individual SMN complex member and identified gemin3 as another regeneration-essential gene. These three genes are likely pan-regenerative, since they affect the regeneration of hair cells, liver, and caudal fin. RNA-Seq analysis reveals that smn1, gemin3, and gemin5 are linked to a common set of genetic pathways, including the tp53 and ErbB pathways. Additional studies indicated all three genes facilitate regeneration by inhibiting the ErbB pathway, thereby allowing cell proliferation in the injured neuromasts. This study provides a new understanding of the SMN complex and a potential etiology for SMA and potentially other rare unidentified genetic diseases with similar symptoms.
Collapse
Affiliation(s)
- Wuhong Pei
- Translational and Functional Genomics Branch, National Human Genome Research Institute, Bethesda, MD 20892 USA
| | - Lisha Xu
- Translational and Functional Genomics Branch, National Human Genome Research Institute, Bethesda, MD 20892 USA
| | - Zelin Chen
- Translational and Functional Genomics Branch, National Human Genome Research Institute, Bethesda, MD 20892 USA
| | - Claire C. Slevin
- Translational and Functional Genomics Branch, National Human Genome Research Institute, Bethesda, MD 20892 USA
| | - Kade P. Pettie
- Translational and Functional Genomics Branch, National Human Genome Research Institute, Bethesda, MD 20892 USA
| | - Stephen Wincovitch
- Cytogenetics and Microscopy Core, National Human Genome Research Institute, Bethesda, MD 20892 USA
| | - Shawn M. Burgess
- Translational and Functional Genomics Branch, National Human Genome Research Institute, Bethesda, MD 20892 USA
| |
Collapse
|
31
|
Yao Q, Wang L, Mittal R, Yan D, Richmond MT, Denyer S, Requena T, Liu K, Varshney GK, Lu Z, Liu XZ. Transcriptomic Analyses of Inner Ear Sensory Epithelia in Zebrafish. Anat Rec (Hoboken) 2019; 303:527-543. [PMID: 31883312 DOI: 10.1002/ar.24331] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Revised: 08/01/2019] [Accepted: 11/18/2019] [Indexed: 12/25/2022]
Abstract
Analysis of gene expression has the potential to assist in the understanding of multiple cellular processes including proliferation, cell-fate specification, senesence, and activity in both healthy and disease states. Zebrafish model has been increasingly used to understand the process of hearing and the development of the vertebrate auditory system. Within the zebrafish inner ear, there are three otolith organs, each containing a sensory macula of hair cells. The saccular macula is primarily involved in hearing, the utricular macula is primarily involved in balance and the function of the lagenar macula is not completely understood. The goal of this study is to understand the transcriptional differences in the sensory macula associated with different otolith organs with the intention of understanding the genetic mechanisms responsible for the distinct role each organ plays in sensory perception. The sensory maculae of the saccule, utricle, and lagena were dissected out of adult Et(krt4:GFP)sqet4 zebrafish expressing green fluorescent protein in hair cells for transcriptional analysis. The total RNAs of the maculae were isolated and analyzed by RNA GeneChip microarray. Several of the differentially expressed genes are known to be involved in deafness, otolith development and balance. Gene expression among these otolith organs was very well conserved with less than 10% of genes showing differential expression. Data from this study will help to elucidate which genes are involved in hearing and balance. Furthermore, the findings of this study will assist in the development of the zebrafish model for human hearing and balance disorders. Anat Rec, 303:527-543, 2020. © 2019 American Association for Anatomy.
Collapse
Affiliation(s)
- Qi Yao
- Department of Otolaryngology, Miller School of Medicine, University of Miami, Miami, Florida.,Department of Biology, University of Miami, Miami, Florida
| | - Lingyu Wang
- Department of Biology, University of Miami, Miami, Florida
| | - Rahul Mittal
- Department of Otolaryngology, Miller School of Medicine, University of Miami, Miami, Florida
| | - Denise Yan
- Department of Otolaryngology, Miller School of Medicine, University of Miami, Miami, Florida
| | | | - Steven Denyer
- Department of Biology, University of Miami, Miami, Florida
| | - Teresa Requena
- Genes & Human Disease Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma
| | - Kaili Liu
- Genes & Human Disease Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma
| | - Gaurav K Varshney
- Genes & Human Disease Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma
| | - Zhongmin Lu
- Department of Biology, University of Miami, Miami, Florida
| | - Xue Zhong Liu
- Department of Otolaryngology, Miller School of Medicine, University of Miami, Miami, Florida.,Department of Otolaryngology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| |
Collapse
|
32
|
Rissone A, Jimenez E, Bishop K, Carrington B, Slevin C, Wincovitch SM, Sood R, Candotti F, Burgess SM. A model for reticular dysgenesis shows impaired sensory organ development and hair cell regeneration linked to cellular stress. Dis Model Mech 2019; 12:dmm040170. [PMID: 31727854 PMCID: PMC6955229 DOI: 10.1242/dmm.040170] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Accepted: 11/05/2019] [Indexed: 12/14/2022] Open
Abstract
Mutations in the gene AK2 are responsible for reticular dysgenesis (RD), a rare and severe form of primary immunodeficiency in children. RD patients have a severely shortened life expectancy and without treatment die, generally from sepsis soon after birth. The only available therapeutic option for RD is hematopoietic stem cell transplantation (HSCT). To gain insight into the pathophysiology of RD, we previously created zebrafish models for Ak2 deficiencies. One of the clinical features of RD is hearing loss, but its pathophysiology and causes have not been determined. In adult mammals, sensory hair cells of the inner ear do not regenerate; however, their regeneration has been observed in several non-mammalian vertebrates, including zebrafish. Therefore, we used our RD zebrafish models to determine whether Ak2 deficiency affects sensory organ development and/or hair cell regeneration. Our studies indicated that Ak2 is required for the correct development, survival and regeneration of sensory hair cells. Interestingly, Ak2 deficiency induces the expression of several oxidative stress markers and it triggers an increased level of cell death in the hair cells. Finally, we show that glutathione treatment can partially rescue hair cell development in the sensory organs in our RD models, pointing to the potential use of antioxidants as a therapeutic treatment supplementing HSCT to prevent or ameliorate sensorineural hearing deficits in RD patients.
Collapse
Affiliation(s)
- Alberto Rissone
- Translational and Functional Genomics Branch, National Human Genome Research Institute (NHGRI), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Erin Jimenez
- Translational and Functional Genomics Branch, National Human Genome Research Institute (NHGRI), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Kevin Bishop
- NHGRI Zebrafish Core, Translational and Functional Genomics Branch, NHGRI, NIH, Bethesda, MD, USA
| | - Blake Carrington
- NHGRI Zebrafish Core, Translational and Functional Genomics Branch, NHGRI, NIH, Bethesda, MD, USA
| | - Claire Slevin
- Translational and Functional Genomics Branch, National Human Genome Research Institute (NHGRI), National Institutes of Health (NIH), Bethesda, MD, USA
| | | | - Raman Sood
- Translational and Functional Genomics Branch, National Human Genome Research Institute (NHGRI), National Institutes of Health (NIH), Bethesda, MD, USA
- NHGRI Zebrafish Core, Translational and Functional Genomics Branch, NHGRI, NIH, Bethesda, MD, USA
| | - Fabio Candotti
- Division of Immunology and Allergy, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Shawn M Burgess
- Translational and Functional Genomics Branch, National Human Genome Research Institute (NHGRI), National Institutes of Health (NIH), Bethesda, MD, USA
| |
Collapse
|
33
|
Korzh V, Kondrychyn I. Origin and development of circumventricular organs in living vertebrate. Semin Cell Dev Biol 2019; 102:13-20. [PMID: 31706729 DOI: 10.1016/j.semcdb.2019.10.010] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Accepted: 10/17/2019] [Indexed: 01/22/2023]
Abstract
The circumventricular organs (CVOs) function by mediating chemical communication between blood and brain across the blood-brain barrier. Their origin and developmental mechanisms involved are not understood in enough detail due to a lack of molecular markers common for CVOs. These rather small and inconspicuous organs are found in close vicinity to the third and fourth brain ventricles suggestive of ancient evolutionary origin. Recently, an integrated approach based on analysis of CVOs development in the enhancer-trap transgenic zebrafish led to an idea that almost all of CVOs could be highlighted by GFP expression in this transgenic line. This in turn suggested that an enhancer along with a set of genes it regulates may illustrate the first common element of developmental regulation of CVOs. It seems to be related to a mechanism of suppression of the canonical Wnt/ β-catenin signaling that functions in development of fenestrated capillaries typical for CVOs. Based on that observation the common molecular elements of the putative developmental mechanism of CVOs will be discussed in this review.
Collapse
Affiliation(s)
- Vladimir Korzh
- International Institute of Molecular and Cell Biology in Warsaw, Poland.
| | | |
Collapse
|
34
|
PCP and Wnt pathway components act in parallel during zebrafish mechanosensory hair cell orientation. Nat Commun 2019; 10:3993. [PMID: 31488837 PMCID: PMC6728366 DOI: 10.1038/s41467-019-12005-y] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Accepted: 08/05/2019] [Indexed: 12/03/2022] Open
Abstract
Planar cell polarity (PCP) plays crucial roles in developmental processes such as gastrulation, neural tube closure and hearing. Wnt pathway mutants are often classified as PCP mutants due to similarities between their phenotypes. Here, we show that in the zebrafish lateral line, disruptions of the PCP and Wnt pathways have differential effects on hair cell orientations. While mutations in the PCP genes vangl2 and scrib cause random orientations of hair cells, mutations in wnt11f1, gpc4 and fzd7a/b induce hair cells to adopt a concentric pattern. This concentric pattern is not caused by defects in PCP but is due to misaligned support cells. The molecular basis of the support cell defect is unknown but we demonstrate that the PCP and Wnt pathways work in parallel to establish proper hair cell orientation. Consequently, hair cell orientation defects are not solely explained by defects in PCP signaling, and some hair cell phenotypes warrant re-evaluation. Planar cell polarity (PCP) regulates hair cell orientation in the zebrafish lateral line. Here, the authors show that mutating Wnt pathway genes (wnt11f1, fzd7a/b, and gpc4) causes concentric hair cell patterns not regulated by PCP, thus showing PCP/Wnt pathway genes have different consequences on hair cell orientation.
Collapse
|
35
|
Mi XX, Yan J, Li Y, Shi JP. Wnt/β-catenin signaling was activated in supporting cells during exposure of the zebrafish lateral line to cisplatin. Ann Anat 2019; 226:48-56. [PMID: 31330310 DOI: 10.1016/j.aanat.2019.07.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2018] [Revised: 02/05/2019] [Accepted: 07/03/2019] [Indexed: 11/16/2022]
Abstract
Zebrafish lateral line neuromasts are composed of central hair cells surrounded by supporting cells. Cisplatin is a common anticancer drug, with hair cell disruption being a frequent side effect of this drug. In our study, we observed complete functional hair cell loss after six hours of cisplatin insult in neuromasts, as demonstrated by anti-parvalbumin 3 immunofluorescence staining or YO-PRO1 vital dye staining. Time course analysis of neuromast hair cell regeneration showed that regenerated hair cells first appeared between 12 and 24h after damage, and the abundance of these cells increased stepwise with recovery time. After 72h, 90% of the hair cells were regenerated, and after 84h, the number of regenerated hair cells was comparable to the number of neuromast hair cells before treatment. The expression pattern of slc17a8 also showed that hair cells were regenerated after cisplatin exposure. Meanwhile, peripheral supporting cells moved toward the center of the neuromasts, as shown by the in situ expression pattern of sox21a. Increased hair cell progenitor formation was also observed, as demonstrated by the in situ expression pattern of atoh1a. Furthermore, we detected increased expression of wnt2, wnt3a, and ctnnb1 in sorted supporting cells from the sqet10 transgenic line, which labels neuromast supporting cells specifically. In situ hybridization analysis also showed decreased expression of dkk1a and dkk2. Regenerated hair cells were inhibited by early inhibition of Wnt/β-catenin signaling. Taken together, the results presented here showed that Wnt/β-catenin signaling was activated in supporting cells during cisplatin exposure earlier than expected. Our results also indicated that supporting cells enabled hair cell regeneration via Wnt/β-catenin signaling during cisplatin exposure.
Collapse
Affiliation(s)
- Xiao-Xiao Mi
- Institute of Translational Medicine, The Affiliated Hospital of Hangzhou Normal University, Hangzhou, Zhejiang, China.
| | - Jian Yan
- Institute of Translational Medicine, The Affiliated Hospital of Hangzhou Normal University, Hangzhou, Zhejiang, China.
| | - Yuan Li
- Department of Otolaryngology-Head and Neck Surgery, The Affiliated Hospital of Hangzhou Normal University, Hangzhou, Zhejiang, China.
| | - Jun-Ping Shi
- Institute of Translational Medicine, The Affiliated Hospital of Hangzhou Normal University, Hangzhou, Zhejiang, China.
| |
Collapse
|
36
|
Minhas R, Paterek A, Łapiński M, Bazała M, Korzh V, Winata CL. A novel conserved enhancer at zebrafish zic3 and zic6 loci drives neural expression. Dev Dyn 2019; 248:837-849. [PMID: 31194899 PMCID: PMC6771876 DOI: 10.1002/dvdy.69] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Revised: 05/28/2019] [Accepted: 06/07/2019] [Indexed: 01/15/2023] Open
Abstract
Background Identifying enhancers and deciphering their putative roles represent a major step to better understand the mechanism of metazoan gene regulation, development, and the role of regulatory elements in disease. Comparative genomics and transgenic assays have been used with some success to identify critical regions that are involved in regulating the spatiotemporal expression of genes during embryogenesis. Results We identified two novel tetrapod‐teleost conserved noncoding elements within the vicinity of the zic3 and zic6 loci in the zebrafish genome and demonstrated their ability to drive tissue‐specific expression in a transgenic zebrafish assay. The syntenic analysis and robust green fluorescent expression in the developing habenula in the stable transgenic line were correlated with known sites of endogenous zic3 and zic6 expression. Conclusion This transgenic line that expresses green fluorescent protein in the habenula is a valuable resource for studying a specific population of cells in the zebrafish central nervous system. Our observations indicate that a genomic sequence that is conserved between humans and zebrafish acts as an enhancer that likely controls zic3 and zic6 expression. Identified a novel enhancer near zebrafish zic3/zic6 locus. The novel enhancer drives tissue‐specific expression in the habenula. Zebrafish transgenic line generated in this study can be a useful resource for studying development of habenula.
Collapse
Affiliation(s)
- Rashid Minhas
- International Institute of Molecular and Cell Biology, Warsaw, Poland.,Randall Centre of Cell and Molecular Biophysics, Faculty of Life Sciences and Medicine, King's College London, London, UK
| | - Aleksandra Paterek
- International Institute of Molecular and Cell Biology, Warsaw, Poland.,Department of Clinical Physiology, Centre of Postgraduate Medical Education, Warsaw, Poland
| | - Maciej Łapiński
- International Institute of Molecular and Cell Biology, Warsaw, Poland
| | - Michał Bazała
- International Institute of Molecular and Cell Biology, Warsaw, Poland
| | - Vladimir Korzh
- International Institute of Molecular and Cell Biology, Warsaw, Poland
| | - Cecilia L Winata
- International Institute of Molecular and Cell Biology, Warsaw, Poland.,Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| |
Collapse
|
37
|
Filipponi D, Emelyanov A, Muller J, Molina C, Nichols J, Bulavin DV. DNA Damage Signaling-Induced Cancer Cell Reprogramming as a Driver of Tumor Relapse. Mol Cell 2019; 74:651-663.e8. [DOI: 10.1016/j.molcel.2019.03.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Revised: 01/23/2019] [Accepted: 02/28/2019] [Indexed: 12/14/2022]
|
38
|
Chan S, Shen D, Sang Y, Wang S, Wang Y, Chen C, Gao B, Song C. Development of enhancer-trapping and -detection vectors mediated by the Tol2 transposon in zebrafish. PeerJ 2019; 7:e6862. [PMID: 31106068 PMCID: PMC6499061 DOI: 10.7717/peerj.6862] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Accepted: 03/28/2019] [Indexed: 01/02/2023] Open
Abstract
Enhancers are key transcriptional drivers of gene expression. The identification of enhancers in the genome is central for understanding gene-expression programs. Although transposon-mediated enhancer trapping (ET) is a powerful approach to the identification of enhancers in zebrafish, its efficiency varies considerably. To improve the ET efficiency, we constructed Tol2-mediated ET vectors with a reporter gene (mCherry) expression box driven by four minimal promoters (Gata, Myc, Krt4 and Oct4), respectively. The ET efficiency and expression background were compared among the four promoters by zebrafish embryo injection at the one-cell stage. The results showed that the Gata minimal promoter yielded the lowest basic expression and the second-highest trapping efficiency (44.6% at 12 hpf (hour post-fertilization) and 23.1% at 72 hpf, n = 305 and n = 307). The Krt4 promoter had the highest trapping efficiency (64% at 12 hpf and 67.1% at 72 hpf, n = 302 and n = 301) and the strongest basic expression. To detect enhancer activity, chicken 5′HS4 double insulators were cloned into the two ET vectors with the Gata or Krt4 minimal promoter, flanking the mCherry expression box. The resulting detection vectors were injected into zebrafish embryos. mCherry expression driven by the Gata promoter (about 5%, n = 301) was decreased significantly compared with that observed for embryos injected with the ET vectors (23% at 72 hpf, n = 308). These results suggest that the insulators block the genome-position effects and that this vector is fit for enhancer-activity evaluation. To assess the compatibility between the enhancers and the minimal promoters, four enhancers (CNS1, Z48, Hand2 and Hs769) were cloned upstream of the Gata or Beta-globin minimal promoter in the enhancer-activity-detection vectors. The resulting recombinant vectors were assayed by zebrafish embryo injection. We found that Z48 and CNS1 responded to the Gata minimal promoter, and that Hand2 only responded to the Beta-globin minimal promoter. In contrast, Hs769 did not respond to either the Gata or Beta-globin minimal promoters. These results suggest the existence of compatibility between enhancers and minimal promoters. This study represents a systematic approach to the discovery of optional ET and enhancer-detection vectors. We are eager to provide a superior tool for understanding functional genomics.
Collapse
Affiliation(s)
- Shuheng Chan
- Yangzhou University, Institute of Animal Mobilome and Genome, College of Animal Science & Technology, Yangzhou, Jiangsu, China
| | - Dan Shen
- Yangzhou University, Institute of Animal Mobilome and Genome, College of Animal Science & Technology, Yangzhou, Jiangsu, China
| | - Yatong Sang
- Yangzhou University, Institute of Animal Mobilome and Genome, College of Animal Science & Technology, Yangzhou, Jiangsu, China
| | - Saisai Wang
- Yangzhou University, Institute of Animal Mobilome and Genome, College of Animal Science & Technology, Yangzhou, Jiangsu, China
| | - Yali Wang
- Yangzhou University, Institute of Animal Mobilome and Genome, College of Animal Science & Technology, Yangzhou, Jiangsu, China
| | - Cai Chen
- Yangzhou University, Institute of Animal Mobilome and Genome, College of Animal Science & Technology, Yangzhou, Jiangsu, China
| | - Bo Gao
- Yangzhou University, Institute of Animal Mobilome and Genome, College of Animal Science & Technology, Yangzhou, Jiangsu, China
| | - Chengyi Song
- Yangzhou University, Institute of Animal Mobilome and Genome, College of Animal Science & Technology, Yangzhou, Jiangsu, China
| |
Collapse
|
39
|
DeSmidt AA, Zou B, Grati M, Yan D, Mittal R, Yao Q, Richmond MT, Denyer S, Liu XZ, Lu Z. Zebrafish Model for Nonsyndromic X-Linked Sensorineural Deafness, DFNX1. Anat Rec (Hoboken) 2019; 303:544-555. [PMID: 30874365 DOI: 10.1002/ar.24115] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Revised: 08/23/2018] [Accepted: 09/05/2018] [Indexed: 11/08/2022]
Abstract
Hereditary deafness is often a neurosensory disorder and affects the quality of life of humans. Only three X-linked genes (POU class 3 homeobox 4 (POU3F4), phosphoribosyl pyrophosphate synthetase 1 (PRPS1), and small muscle protein X-linked (SMPX)) are known to be involved in nonsyndromic hearing loss. Four PRPS1 missense mutations have been found to associate with X-linked nonsyndromic sensorineural deafness (DFNX1/DFN2) in humans. However, a causative relationship between PRPS1 mutations and hearing loss in humans has not been well studied in any animal model. Phosphoribosyl pyrophosphate synthetase 1 (PRS-I) is highly conserved in vertebrate taxa. In this study, we used the zebrafish as a model to investigate the auditory role of zebrafish orthologs (prps1a and prps1b) of the human PRPS1 gene with whole mount in situ hybridization, reverse transcription polymerase chain reaction, phenotypic screening, confocal imaging, and electrophysiological methods. We found that both prps1a and prps1b genes were expressed in the inner ear of zebrafish. Splice-blocking antisense morpholino oligonucleotides (MO1 and MO2) caused exon-2 skip and intron-2 retention of prps1a and exon-2 skip and intron-1 retention of prps1b to knock down functions of the genes, respectively. MO1 and MO2 morphants had smaller otic vesicles and otoliths, fewer inner ear hair cells, and lower microphonic response amplitude and sensitivity than control zebrafish. Therefore, knockdown of either prps1a or prps1b resulted in significant sensorineural hearing loss in zebrafish. We conclude that the prps1 genes are essential for hearing in zebrafish, which has the potential to help us understand the biology of human deafness DFNX1/DFN2. Anat Rec, 303:544-555, 2020. © 2019 American Association for Anatomy.
Collapse
Affiliation(s)
| | - Bing Zou
- Department of Biology, University of Miami, Coral Gables, Florida.,Department of Otolaryngology, University of Miami Miller School of Medicine, Miami, Florida
| | - M'hamed Grati
- Department of Otolaryngology, University of Miami Miller School of Medicine, Miami, Florida
| | - Denise Yan
- Department of Otolaryngology, University of Miami Miller School of Medicine, Miami, Florida
| | - Rahul Mittal
- Department of Otolaryngology, University of Miami Miller School of Medicine, Miami, Florida
| | - Qi Yao
- Department of Biology, University of Miami, Coral Gables, Florida.,Department of Otolaryngology, University of Miami Miller School of Medicine, Miami, Florida
| | | | - Steven Denyer
- Department of Biology, University of Miami, Coral Gables, Florida
| | - Xue Zhong Liu
- Department of Otolaryngology, University of Miami Miller School of Medicine, Miami, Florida.,Department of Otolaryngology, Xiangya Hospital, Central South University, Changsha, People's Republic of China
| | - Zhongmin Lu
- Department of Biology, University of Miami, Coral Gables, Florida.,Department of Otolaryngology, University of Miami Miller School of Medicine, Miami, Florida.,Neuroscience Program, University of Miami, Miami, Florida.,International Center for Marine Studies, Shanghai Ocean University, Shanghai, People's Republic of China
| |
Collapse
|
40
|
Lush ME, Diaz DC, Koenecke N, Baek S, Boldt H, St Peter MK, Gaitan-Escudero T, Romero-Carvajal A, Busch-Nentwich EM, Perera AG, Hall KE, Peak A, Haug JS, Piotrowski T. scRNA-Seq reveals distinct stem cell populations that drive hair cell regeneration after loss of Fgf and Notch signaling. eLife 2019; 8:e44431. [PMID: 30681411 PMCID: PMC6363392 DOI: 10.7554/elife.44431] [Citation(s) in RCA: 88] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2018] [Accepted: 01/24/2019] [Indexed: 12/25/2022] Open
Abstract
Loss of sensory hair cells leads to deafness and balance deficiencies. In contrast to mammalian hair cells, zebrafish ear and lateral line hair cells regenerate from poorly characterized support cells. Equally ill-defined is the gene regulatory network underlying the progression of support cells to differentiated hair cells. scRNA-Seq of lateral line organs uncovered five different support cell types, including quiescent and activated stem cells. Ordering of support cells along a developmental trajectory identified self-renewing cells and genes required for hair cell differentiation. scRNA-Seq analyses of fgf3 mutants, in which hair cell regeneration is increased, demonstrates that Fgf and Notch signaling inhibit proliferation of support cells in parallel by inhibiting Wnt signaling. Our scRNA-Seq analyses set the foundation for mechanistic studies of sensory organ regeneration and is crucial for identifying factors to trigger hair cell production in mammals. The data is searchable and publicly accessible via a web-based interface.
Collapse
Affiliation(s)
- Mark E Lush
- Stowers Institute for Medical ResearchKansas CityUnited States
| | - Daniel C Diaz
- Stowers Institute for Medical ResearchKansas CityUnited States
| | - Nina Koenecke
- Stowers Institute for Medical ResearchKansas CityUnited States
| | - Sungmin Baek
- Stowers Institute for Medical ResearchKansas CityUnited States
| | - Helena Boldt
- Stowers Institute for Medical ResearchKansas CityUnited States
| | | | | | - Andres Romero-Carvajal
- Stowers Institute for Medical ResearchKansas CityUnited States
- Pontificia Universidad Catolica del EcuadorCiencias BiologicasQuitoEcuador
| | - Elisabeth M Busch-Nentwich
- Wellcome Sanger Institute, Wellcome Genome CampusHinxtonUnited Kingdom
- Department of MedicineUniversity of CambridgeCambridgeUnited Kingdom
| | - Anoja G Perera
- Stowers Institute for Medical ResearchKansas CityUnited States
| | - Kathryn E Hall
- Stowers Institute for Medical ResearchKansas CityUnited States
| | - Allison Peak
- Stowers Institute for Medical ResearchKansas CityUnited States
| | - Jeffrey S Haug
- Stowers Institute for Medical ResearchKansas CityUnited States
| | | |
Collapse
|
41
|
Korzh V, Kondrychyn I, Winata C. The Zebrafish as a New Model System for Experimental Biology. CYTOL GENET+ 2018. [DOI: 10.3103/s009545271806004x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
42
|
Enhancer Trapping and Annotation in Zebrafish Mediated with Sleeping Beauty, piggyBac and Tol2 Transposons. Genes (Basel) 2018; 9:genes9120630. [PMID: 30551672 PMCID: PMC6316676 DOI: 10.3390/genes9120630] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Revised: 12/07/2018] [Accepted: 12/10/2018] [Indexed: 12/18/2022] Open
Abstract
Although transposon-mediated enhancer trapping (ET) is successfully applied in diverse models, the efficiency of various transposon systems varies significantly, and little information is available regarding efficiency of enhancer trapping by various transposons in zebrafish. Most potential enhancers (Ens) still lack evidence of actual En activity. Here, we compared the differences in ET efficiency between sleeping beauty (SB), piggyBac (PB) and Tol2 transposons. Tol2 represented the highest germline transfer efficiencies at 55.56% (NF0 = 165), followed by SB (38.36%, NF0 = 151) and PB (32.65%, NF0 = 149). ET lines generated by the Tol2 transposon tended to produce offspring with a single expression pattern per line, while PB and SB tended to generate embryos with multiple expression patterns. In our tests, 10 putative Ens (En1–10) were identified by splinkerette PCR and comparative genomic analysis. Combining the GFP expression profiles and mRNA expression patterns revealed that En1 and En2 may be involved in regulation of the expression of dlx1a and dlx2a, while En6 may be involved in regulation of the expression of line TK4 transgene and rps26, and En7 may be involved in the regulation of the expression of wnt1 and wnt10b. Most identified Ens were found to be transcribed in zebrafish embryos, and their regulatory function may involve eRNAs.
Collapse
|
43
|
Live imaging of collagen deposition during skin development and repair in a collagen I - GFP fusion transgenic zebrafish line. Dev Biol 2018; 441:4-11. [PMID: 29883658 PMCID: PMC6080847 DOI: 10.1016/j.ydbio.2018.06.001] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Revised: 05/30/2018] [Accepted: 06/04/2018] [Indexed: 12/29/2022]
Abstract
Fibrillar collagen is a major component of many tissues but has been difficult to image in vivo using transgenic approaches because of problems associated with establishing cells and organisms that generate GFP-fusion collagens that can polymerise into functional fibrils. Here we have developed and characterised GFP and mCherry collagen-I fusion zebrafish lines with basal epidermal-specific expression. We use these lines to reveal the dynamic nature of collagen-I fibril deposition beneath the developing embryonic epidermis, as well as the repair of this collagen meshwork following wounding. Transmission electron microscope studies show that these transgenic lines faithfully reproduce the collagen ultrastructure present in wild type larval skin. During skin development we show that collagen I is deposited by basal epidermal cells initially in fine filaments that are largely randomly orientated but are subsequently aligned into a cross-hatch, orthogonal sub-epithelial network by embryonic day 4. Following skin wounding, we see that sub-epidermal collagen is re-established in the denuded domain, initially as randomly orientated wisps that subsequently become bonded to the undamaged collagen and aligned in a way that recapitulates developmental deposition of sub-epidermal collagen. Crossing our GFP-collagen line against one with tdTomato marking basal epidermal cell membranes reveals how much more rapidly wound re-epithelialisation occurs compared to the re-deposition of collagen beneath the healed epidermis. By use of other tissue specific drivers it will be possible to establish zebrafish lines to enable live imaging of collagen deposition and its remodelling in various other organs in health and disease. A GFP-collagen I transgenic zebrafish has been generated for live, in vivo, imaging. Collagen fibrils are initially deposited randomly beneath the developing epidermis. This random collagen array subsequently becomes orthogonally aligned. Collagen I deposition following larval wounding recapitulates developmental deposition. Expression of GFP-collagen enables study of collagen dynamics in health and disease.
Collapse
|
44
|
Affiliation(s)
- Jason R. Meyers
- Department of Biology and Program in Neuroscience, Colgate University; Hamilton New York
| |
Collapse
|
45
|
Viader-Llargués O, Lupperger V, Pola-Morell L, Marr C, López-Schier H. Live cell-lineage tracing and machine learning reveal patterns of organ regeneration. eLife 2018; 7:30823. [PMID: 29595471 PMCID: PMC5903862 DOI: 10.7554/elife.30823] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Accepted: 03/28/2018] [Indexed: 12/28/2022] Open
Abstract
Despite the intrinsically stochastic nature of damage, sensory organs recapitulate normal architecture during repair to maintain function. Here we present a quantitative approach that combines live cell-lineage tracing and multifactorial classification by machine learning to reveal how cell identity and localization are coordinated during organ regeneration. We use the superficial neuromasts in larval zebrafish, which contain three cell classes organized in radial symmetry and a single planar-polarity axis. Visualization of cell-fate transitions at high temporal resolution shows that neuromasts regenerate isotropically to recover geometric order, proportions and polarity with exceptional accuracy. We identify mediolateral position within the growing tissue as the best predictor of cell-fate acquisition. We propose a self-regulatory mechanism that guides the regenerative process to identical outcome with minimal extrinsic information. The integrated approach that we have developed is simple and broadly applicable, and should help define predictive signatures of cellular behavior during the construction of complex tissues.
Collapse
Affiliation(s)
- Oriol Viader-Llargués
- Unit Sensory Biology & Organogenesis, Helmholtz Zentrum München, Neuherberg, Germany.,Laboratory of Sensory Cell Biology, Centre for Genomic Regulation, Barcelona, Spain
| | - Valerio Lupperger
- Institute of Computational Biology, Helmholtz Zentrum München, Neuherberg, Germany
| | - Laura Pola-Morell
- Unit Sensory Biology & Organogenesis, Helmholtz Zentrum München, Neuherberg, Germany.,Laboratory of Sensory Cell Biology, Centre for Genomic Regulation, Barcelona, Spain
| | - Carsten Marr
- Institute of Computational Biology, Helmholtz Zentrum München, Neuherberg, Germany
| | - Hernán López-Schier
- Unit Sensory Biology & Organogenesis, Helmholtz Zentrum München, Neuherberg, Germany.,Laboratory of Sensory Cell Biology, Centre for Genomic Regulation, Barcelona, Spain
| |
Collapse
|
46
|
Zebrafish Zic Genes Mediate Developmental Signaling. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1046:157-177. [PMID: 29442322 DOI: 10.1007/978-981-10-7311-3_9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
The introduction of genomics into the field of developmental biology led to a vast expansion of knowledge about developmental genes and signaling mechanisms they are involved in. Unlike mammals, the zebrafish features seven Zic genes. This provides an interesting insight into Zic gene evolution. In addition, an unprecedented bioimaging capability of semitransparent zebrafish embryos turns to be a crucial factor in medium- to large-scale analysis of the activity of potential regulatory elements. The Zic family of zinc finger proteins plays an important, relatively well-established, role in the regulation of stem cells and neural development and, in particular, during neural fate commitment and determination. At the same time, some Zic genes are expressed in mesodermal lineages, and their deficiency causes a number of developmental defects in axis formation, establishing body symmetry and cardiac morphogenesis. In stem cells, Zic genes are required to maintain pluripotency by binding to the proximal promoters of pluripotency genes (Oct4, Nanog, Sox2, etc.). During embryogenesis, the dynamic nature of Zic transcriptional regulation is manifested by the interaction of these factors with distal enhancers and other regulatory elements associated with the control of gene transcription and, in particular, with the Nodal and Wnt signaling pathways that play a role in establishing basic organization of the vertebrate body. Zic transcription factors may regulate development through acting alone as well as in combination with other transcription factors. This is achieved due to Zic binding to sites adjacent to the binding sites of other transcription factors, including Gli. This probably leads to the formation of multi-transcription factor complexes associated with enhancers.
Collapse
|
47
|
García-Lecea M, Gasanov E, Jedrychowska J, Kondrychyn I, Teh C, You MS, Korzh V. Development of Circumventricular Organs in the Mirror of Zebrafish Enhancer-Trap Transgenics. Front Neuroanat 2017; 11:114. [PMID: 29375325 PMCID: PMC5770639 DOI: 10.3389/fnana.2017.00114] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2017] [Accepted: 11/22/2017] [Indexed: 11/18/2022] Open
Abstract
The circumventricular organs (CVOs) are small structures lining the cavities of brain ventricular system. They are associated with the semitransparent regions of the blood-brain barrier (BBB). Hence it is thought that CVOs mediate biochemical signaling and cell exchange between the brain and systemic blood. Their classification is still controversial and development not fully understood largely due to an absence of tissue-specific molecular markers. In a search for molecular determinants of CVOs we studied the green fluorescent protein (GFP) expression pattern in several zebrafish enhancer trap transgenics including Gateways (ET33-E20) that has been instrumental in defining the development of choroid plexus. In Gateways the GFP is expressed in regions of the developing brain outside the choroid plexus, which remain to be characterized. The neuroanatomical and histological analysis suggested that some previously unassigned domains of GFP expression may correspond to at least six other CVOs–the organum vasculosum laminae terminalis (OVLT), subfornical organ (SFO), paraventricular organ (PVO), pineal (epiphysis), area postrema (AP) and median eminence (ME). Two other CVOs, parapineal and subcommissural organ (SCO) were detected in other enhancer-trap transgenics. Hence enhancer-trap transgenic lines could be instrumental for developmental studies of CVOs in zebrafish and understanding of the molecular mechanism of disease such a hydrocephalus in human. Their future analysis may shed light on general and specific molecular mechanisms that regulate development of CVOs.
Collapse
Affiliation(s)
- Marta García-Lecea
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore, Singapore.,Department of Basic Biomedical Sciences, Universidad Europea de Madrid, Madrid, Spain
| | - Evgeny Gasanov
- International Institute of Molecular and Cell Biology in Warsaw, Warsaw, Poland
| | - Justyna Jedrychowska
- International Institute of Molecular and Cell Biology in Warsaw, Warsaw, Poland.,Postgraduate School of Molecular Medicine, Medical University of Warsaw, Warsaw, Poland
| | - Igor Kondrychyn
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore, Singapore.,RIKEN Center for Developmental Biology, Kobe, Japan
| | - Cathleen Teh
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore, Singapore
| | - May-Su You
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore, Singapore.,National Health Research Institutes (NHRI), Zhunan, Taiwan
| | - Vladimir Korzh
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore, Singapore.,International Institute of Molecular and Cell Biology in Warsaw, Warsaw, Poland
| |
Collapse
|
48
|
Preclinical and clinical advances in transposon-based gene therapy. Biosci Rep 2017; 37:BSR20160614. [PMID: 29089466 PMCID: PMC5715130 DOI: 10.1042/bsr20160614] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Revised: 10/26/2017] [Accepted: 10/30/2017] [Indexed: 02/08/2023] Open
Abstract
Transposons derived from Sleeping Beauty (SB), piggyBac (PB), or Tol2 typically require cotransfection of transposon DNA with a transposase either as an expression plasmid or mRNA. Consequently, this results in genomic integration of the potentially therapeutic gene into chromosomes of the desired target cells, and thus conferring stable expression. Non-viral transfection methods are typically preferred to deliver the transposon components into the target cells. However, these methods do not match the efficacy typically attained with viral vectors and are sometimes associated with cellular toxicity evoked by the DNA itself. In recent years, the overall transposition efficacy has gradually increased by codon optimization of the transposase, generation of hyperactive transposases, and/or introduction of specific mutations in the transposon terminal repeats. Their versatility enabled the stable genetic engineering in many different primary cell types, including stem/progenitor cells and differentiated cell types. This prompted numerous preclinical proof-of-concept studies in disease models that demonstrated the potential of DNA transposons for ex vivo and in vivo gene therapy. One of the merits of transposon systems relates to their ability to deliver relatively large therapeutic transgenes that cannot readily be accommodated in viral vectors such as full-length dystrophin cDNA. These emerging insights paved the way toward the first transposon-based phase I/II clinical trials to treat hematologic cancer and other diseases. Though encouraging results were obtained, controlled pivotal clinical trials are needed to corroborate the efficacy and safety of transposon-based therapies.
Collapse
|
49
|
Geisler R, Borel N, Ferg M, Maier JV, Strähle U. Maintenance of Zebrafish Lines at the European Zebrafish Resource Center. Zebrafish 2017; 13 Suppl 1:S19-23. [PMID: 27351617 PMCID: PMC4931740 DOI: 10.1089/zeb.2015.1205] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
We have established a European Zebrafish Resource Center (EZRC) at the KIT. This center not only maintains and distributes a large number of existing mutant and transgenic zebrafish lines but also gives zebrafish researchers access to screening services and technologies such as imaging and high-throughput sequencing, provided by the Institute of Toxicology and Genetics (ITG). The EZRC maintains and distributes the stock collection of the Nüsslein-Volhard laboratory, comprising over 2000 publicly released mutations, as frozen sperm samples. Within the framework of the ZF-HEALTH EU project, the EZRC distributes over 10,000 knockout mutations from the Sanger Institute (United Kingdom), as well as over 100 mutant and transgenic lines from other sources. In this article, we detail the measures we have taken to ensure the health of our fish, including hygiene, quarantine, and veterinary inspections.
Collapse
Affiliation(s)
- Robert Geisler
- Institute of Toxicology and Genetics, Karlsruhe Institute of Technology (KIT) , Eggenstein-Leopoldshafen, Germany
| | - Nadine Borel
- Institute of Toxicology and Genetics, Karlsruhe Institute of Technology (KIT) , Eggenstein-Leopoldshafen, Germany
| | - Marco Ferg
- Institute of Toxicology and Genetics, Karlsruhe Institute of Technology (KIT) , Eggenstein-Leopoldshafen, Germany
| | - Jana Viktoria Maier
- Institute of Toxicology and Genetics, Karlsruhe Institute of Technology (KIT) , Eggenstein-Leopoldshafen, Germany
| | - Uwe Strähle
- Institute of Toxicology and Genetics, Karlsruhe Institute of Technology (KIT) , Eggenstein-Leopoldshafen, Germany
| |
Collapse
|
50
|
Kawakami K, Largaespada DA, Ivics Z. Transposons As Tools for Functional Genomics in Vertebrate Models. Trends Genet 2017; 33:784-801. [PMID: 28888423 DOI: 10.1016/j.tig.2017.07.006] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Revised: 07/14/2017] [Accepted: 07/18/2017] [Indexed: 02/06/2023]
Abstract
Genetic tools and mutagenesis strategies based on transposable elements are currently under development with a vision to link primary DNA sequence information to gene functions in vertebrate models. By virtue of their inherent capacity to insert into DNA, transposons can be developed into powerful tools for chromosomal manipulations. Transposon-based forward mutagenesis screens have numerous advantages including high throughput, easy identification of mutated alleles, and providing insight into genetic networks and pathways based on phenotypes. For example, the Sleeping Beauty transposon has become highly instrumental to induce tumors in experimental animals in a tissue-specific manner with the aim of uncovering the genetic basis of diverse cancers. Here, we describe a battery of mutagenic cassettes that can be applied in conjunction with transposon vectors to mutagenize genes, and highlight versatile experimental strategies for the generation of engineered chromosomes for loss-of-function as well as gain-of-function mutagenesis for functional gene annotation in vertebrate models, including zebrafish, mice, and rats.
Collapse
Affiliation(s)
- Koichi Kawakami
- Division of Molecular and Developmental Biology, National Institute of Genetics, Mishima, Japan; These authors contributed equally to this work
| | - David A Largaespada
- Department of Genetics, Cell Biology and Development, University of Minnesota, MN, USA; These authors contributed equally to this work
| | - Zoltán Ivics
- Division of Medical Biotechnology, Paul Ehrlich Institute, Langen, Germany; These authors contributed equally to this work..
| |
Collapse
|