1
|
Constanty F, Wu B, Wei KH, Lin IT, Dallmann J, Guenther S, Lautenschlaeger T, Priya R, Lai SL, Stainier DYR, Beisaw A. Border-zone cardiomyocytes and macrophages regulate extracellular matrix remodeling to promote cardiomyocyte protrusion during cardiac regeneration. Nat Commun 2025; 16:3823. [PMID: 40268967 PMCID: PMC12019606 DOI: 10.1038/s41467-025-59169-4] [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: 05/24/2024] [Accepted: 04/14/2025] [Indexed: 04/25/2025] Open
Abstract
Despite numerous advances in our understanding of zebrafish cardiac regeneration, an aspect that remains less studied is how regenerating cardiomyocytes invade and replace the collagen-containing injured tissue. Here, we provide an in-depth analysis of the process of cardiomyocyte invasion. We observe close interactions between protruding border-zone cardiomyocytes and macrophages, and show that macrophages are essential for extracellular matrix remodeling at the wound border zone and cardiomyocyte protrusion into the injured area. Single-cell RNA-sequencing reveals the expression of mmp14b, encoding a membrane-anchored matrix metalloproteinase, in several cell types at the border zone. Genetic mmp14b mutation leads to decreased macrophage recruitment, collagen degradation, and subsequent cardiomyocyte protrusion into injured tissue. Furthermore, cardiomyocyte-specific overexpression of mmp14b is sufficient to enhance cardiomyocyte invasion into the injured tissue and along the apical surface of the wound. Altogether, our data provide important insights into the mechanisms underlying cardiomyocyte invasion of the collagen-containing injured tissue during cardiac regeneration.
Collapse
Affiliation(s)
- Florian Constanty
- Mechanisms of Cardiac Regeneration and Repair Lab, Institute of Experimental Cardiology, Heidelberg University, Heidelberg, Germany
- Helmholtz-Institute for Translational AngioCardioScience (HI-TAC) of the Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC) at Heidelberg University, Heidelberg, Germany
- DZHK (German Centre for Cardiovascular Research), Partner site Heidelberg/Mannheim, Heidelberg, Germany
| | - Bailin Wu
- Mechanisms of Cardiac Regeneration and Repair Lab, Institute of Experimental Cardiology, Heidelberg University, Heidelberg, Germany
- DZHK (German Centre for Cardiovascular Research), Partner site Heidelberg/Mannheim, Heidelberg, Germany
| | - Ke-Hsuan Wei
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - I-Ting Lin
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Julia Dallmann
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Stefan Guenther
- Deep Sequencing Platform, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
- DZHK (German Centre for Cardiovascular Research), Partner site Rhein/Main, Rhein/Main, Germany
| | - Till Lautenschlaeger
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Rashmi Priya
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
- Cardio-Pulmonary Institute, Bad Nauheim, Germany
- The Francis Crick Institute, London, UK
| | - Shih-Lei Lai
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Didier Y R Stainier
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
- DZHK (German Centre for Cardiovascular Research), Partner site Rhein/Main, Rhein/Main, Germany
- Cardio-Pulmonary Institute, Bad Nauheim, Germany
| | - Arica Beisaw
- Mechanisms of Cardiac Regeneration and Repair Lab, Institute of Experimental Cardiology, Heidelberg University, Heidelberg, Germany.
- Helmholtz-Institute for Translational AngioCardioScience (HI-TAC) of the Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC) at Heidelberg University, Heidelberg, Germany.
- DZHK (German Centre for Cardiovascular Research), Partner site Heidelberg/Mannheim, Heidelberg, Germany.
| |
Collapse
|
2
|
Kanai SM, Garcia CR, Augustus MR, Sharafeldeen SA, Brooks EP, Sucharov J, Lencer ES, Nichols JT, Clouthier DE. The Gq/11 family of Gα subunits is necessary and sufficient for lower jaw development. Development 2025; 152:dev204396. [PMID: 40171762 PMCID: PMC12045641 DOI: 10.1242/dev.204396] [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: 09/16/2024] [Accepted: 03/18/2025] [Indexed: 04/04/2025]
Abstract
Vertebrate jaw development is coordinated by highly conserved ligand-receptor systems such as the peptide ligand Endothelin 1 (Edn1) and Endothelin receptor type A (Ednra), which are required for patterning of lower jaw structures. The Edn1/Ednra signaling pathway establishes the identity of lower jaw progenitor cells by regulating expression of numerous patterning genes, but the intracellular signaling mechanisms linking receptor activation to gene regulation remain poorly understood. As a first step towards elucidating this mechanism, we examined the function of the Gq/11 family of Gα subunits in zebrafish using pharmacological inhibition and genetic ablation of Gq/11 activity, and transgenic induction of a constitutively active Gq protein in edn1-/- embryos. Genetic loss of Gq/11 activity fully recapitulated the edn1-/- phenotype, with genes encoding G11 being most essential. Furthermore, inducing Gq activity in edn1-/- embryos not only restored Edn1/Ednra-dependent jaw structures and gene expression signatures but also caused homeosis of the upper jaw structure into a lower jaw-like structure. These results indicate that Gq/11 is necessary and sufficient to mediate the lower jaw patterning mechanism for Ednra in zebrafish.
Collapse
Affiliation(s)
- Stanley M. Kanai
- Department of Craniofacial Biology, School of Dental Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80108, USA
| | - Chloe R. Garcia
- Department of Craniofacial Biology, School of Dental Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80108, USA
| | - MaCalia R. Augustus
- Department of Craniofacial Biology, School of Dental Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80108, USA
| | - Shujan A. Sharafeldeen
- Department of Craniofacial Biology, School of Dental Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80108, USA
| | - Elliott P. Brooks
- Department of Craniofacial Biology, School of Dental Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80108, USA
| | - Juliana Sucharov
- Department of Craniofacial Biology, School of Dental Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80108, USA
| | - Ezra S. Lencer
- Department of Biology, Lafayette College, Easton, PA 18042, USA
| | - James T. Nichols
- Department of Craniofacial Biology, School of Dental Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80108, USA
| | - David E. Clouthier
- Department of Craniofacial Biology, School of Dental Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80108, USA
| |
Collapse
|
3
|
Benman W, Iyengar P, Mumford TR, Huang Z, Kapoor M, Liu G, Bugaj LJ. Multiplexed dynamic control of temperature to probe and observe mammalian cells. Cell Syst 2025; 16:101234. [PMID: 40081372 DOI: 10.1016/j.cels.2025.101234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Revised: 09/03/2024] [Accepted: 02/19/2025] [Indexed: 03/16/2025]
Abstract
Temperature is an important biological stimulus, yet there is a lack of approaches to modulate the temperature of biological samples in a dynamic and high-throughput manner. The thermoPlate is a device for programmable control of temperature in a 96-well plate, compatible with cell culture and microscopy. The thermoPlate maintains feedback control of temperature independently in each well, with minutes-scale heating and cooling through ΔT = 15-20°C. We first used the thermoPlate to characterize the rapid temperature-dependent phase separation of a synthetic elastin-like polypeptide (ELP53). We then examined stress granule (SG) formation in response to dynamic heat stress, revealing adaptation of SGs to persistent heat and formation of a memory of stress that prevented SG formation in response to subsequent heat shocks. The capabilities and open-source nature of the thermoPlate will empower the study and engineering of a wide range of thermoresponsive phenomena. A record of this paper's transparent peer review process is included in the Supplemental information.
Collapse
Affiliation(s)
- William Benman
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Pavan Iyengar
- Department of Chemistry, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Thomas R Mumford
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Zikang Huang
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Manya Kapoor
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Grace Liu
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Lukasz J Bugaj
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA; Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA 19104, USA; Institute of Regenerative Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
4
|
Burkhardt DS, Leyden C, Thomas C, Brysch C, Dehmelt FA, Arrenberg AB. Behavioral and neurophysiological effects of electrical stunning on zebrafish larvae. Lab Anim (NY) 2025; 54:50-58. [PMID: 39870879 PMCID: PMC11790490 DOI: 10.1038/s41684-024-01505-0] [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: 03/06/2024] [Accepted: 12/16/2024] [Indexed: 01/29/2025]
Abstract
Two methods dominate the way that zebrafish larvae are euthanized after experimental procedures: anesthetic overdose and rapid cooling. Although MS-222 is easy to apply, this anesthetic takes about a minute to act and fish show aversive reactions and interindividual differences, limiting its reliability. Rapid cooling kills larvae after several hours and is not listed as an approved method in the relevant European Union directive. Electrical stunning is a promising alternative euthanasia method for zebrafish but has not yet been fully established. Here we characterize both behavioral and neurophysiological effects of electrical stunning in 4-day-old zebrafish larvae. We identified the electric field characteristics and stimulus duration (50 V/cm alternating current for 32 s) that reliably euthanizes free-swimming larvae and agarose-embedded larvae with an easy-to-implement protocol. Behavioral analysis and calcium neurophysiology show that larvae lose consciousness and stop responding to touch and visual stimuli very quickly (<1 s). Electrically stunned larvae no longer show coordinated brain activity. Their brains instead undergo a series of concerted whole-brain calcium waves over the course of many minutes before the cessation of all brain signals. Consistent with the need to implement the 3R at all stages of animal experimentation, the rapid and reliable euthanasia achieved by electrical stunning has potential for refinement of the welfare of more than 5 million zebrafish used annually in biomedical research worldwide.
Collapse
Affiliation(s)
- David-Samuel Burkhardt
- Werner Reichardt Centre for Integrative Neuroscience and Institute for Neurobiology, University of Tuebingen, Tuebingen, Germany
| | - Claire Leyden
- Werner Reichardt Centre for Integrative Neuroscience and Institute for Neurobiology, University of Tuebingen, Tuebingen, Germany
| | - Carina Thomas
- Werner Reichardt Centre for Integrative Neuroscience and Institute for Neurobiology, University of Tuebingen, Tuebingen, Germany
| | - Christian Brysch
- Werner Reichardt Centre for Integrative Neuroscience and Institute for Neurobiology, University of Tuebingen, Tuebingen, Germany
| | - Florian Alexander Dehmelt
- Werner Reichardt Centre for Integrative Neuroscience and Institute for Neurobiology, University of Tuebingen, Tuebingen, Germany
| | - Aristides B Arrenberg
- Werner Reichardt Centre for Integrative Neuroscience and Institute for Neurobiology, University of Tuebingen, Tuebingen, Germany.
| |
Collapse
|
5
|
Weissenboeck FP, Pieper M, Schepers H, Hötte S, Klöcker N, Hüwel S, van Impel A, Schulte-Merker S, Rentmeister A. Spatiotemporal control of translation in live zebrafish embryos via photoprotected mRNAs. Commun Chem 2025; 8:16. [PMID: 39828804 PMCID: PMC11743775 DOI: 10.1038/s42004-025-01411-7] [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: 05/20/2024] [Accepted: 01/07/2025] [Indexed: 01/22/2025] Open
Abstract
Translation of mRNA into protein is a fundamental process and tightly controlled during development. Several mechanisms acting on the mRNA level regulate when and where an mRNA is expressed. To explore the effects of conditional and transient gene expression in a developing organism, it is vital to experimentally enable abrogation and restoration of translation. We recently developed the FlashCaps technology allowing preparation of translationally muted mRNAs and their controlled activation by light. Here, we validate its functionality in vivo. We demonstrate that translation of FlashCap-eGFP-mRNA can be triggered in zebrafish embryos with spatiotemporal control. The injected FlashCap-mRNA is stable for hours and remains muted. Light-mediated activation up to 24 h post fertilization produces visible amounts of eGFP and can be restricted to distinct parts of the embryo. This methodology extends the toolbox for vertebrate models by enabling researchers to locally activate mRNA translation at different timepoints during development.
Collapse
Affiliation(s)
| | - Melissa Pieper
- Institute of Biochemistry, University of Münster, Münster, Germany
| | - Helena Schepers
- Institute of Biochemistry, University of Münster, Münster, Germany
| | - Sophie Hötte
- Institute for Cardiovascular Organogenesis and Regeneration, Faculty of Medicine, University of Münster, Münster, Germany
- Multiscale Imaging Center, University of Münster, Münster, Germany
| | - Nils Klöcker
- Institute of Biochemistry, University of Münster, Münster, Germany
| | - Sabine Hüwel
- Institute of Biochemistry, University of Münster, Münster, Germany
| | - Andreas van Impel
- Institute for Cardiovascular Organogenesis and Regeneration, Faculty of Medicine, University of Münster, Münster, Germany
- Multiscale Imaging Center, University of Münster, Münster, Germany
| | - Stefan Schulte-Merker
- Institute for Cardiovascular Organogenesis and Regeneration, Faculty of Medicine, University of Münster, Münster, Germany
- Multiscale Imaging Center, University of Münster, Münster, Germany
| | - Andrea Rentmeister
- Institute of Biochemistry, University of Münster, Münster, Germany.
- Department of Chemistry, Ludwig-Maximilians-Universität München, Butenandtstr. 5-13, 81377, Munich, Germany.
| |
Collapse
|
6
|
Mead AF, Wood NB, Nelson SR, Palmer BM, Yang L, Previs SB, Ploysangngam A, Kennedy GG, McAdow JF, Tremble SM, Zimmermann MA, Cipolla MJ, Ebert AM, Johnson AN, Gurnett CA, Previs MJ, Warshaw DM. Functional role of myosin-binding protein H in thick filaments of developing vertebrate fast-twitch skeletal muscle. J Gen Physiol 2024; 156:e202413604. [PMID: 39373654 PMCID: PMC11461142 DOI: 10.1085/jgp.202413604] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 08/01/2024] [Accepted: 09/20/2024] [Indexed: 10/08/2024] Open
Abstract
Myosin-binding protein H (MyBP-H) is a component of the vertebrate skeletal muscle sarcomere with sequence and domain homology to myosin-binding protein C (MyBP-C). Whereas skeletal muscle isoforms of MyBP-C (fMyBP-C, sMyBP-C) modulate muscle contractility via interactions with actin thin filaments and myosin motors within the muscle sarcomere "C-zone," MyBP-H has no known function. This is in part due to MyBP-H having limited expression in adult fast-twitch muscle and no known involvement in muscle disease. Quantitative proteomics reported here reveal that MyBP-H is highly expressed in prenatal rat fast-twitch muscles and larval zebrafish, suggesting a conserved role in muscle development and prompting studies to define its function. We take advantage of the genetic control of the zebrafish model and a combination of structural, functional, and biophysical techniques to interrogate the role of MyBP-H. Transgenic, FLAG-tagged MyBP-H or fMyBP-C both localize to the C-zones in larval myofibers, whereas genetic depletion of endogenous MyBP-H or fMyBP-C leads to increased accumulation of the other, suggesting competition for C-zone binding sites. Does MyBP-H modulate contractility in the C-zone? Globular domains critical to MyBP-C's modulatory functions are absent from MyBP-H, suggesting that MyBP-H may be functionally silent. However, our results suggest an active role. In vitro motility experiments indicate MyBP-H shares MyBP-C's capacity as a molecular "brake." These results provide new insights and raise questions about the role of the C-zone during muscle development.
Collapse
Affiliation(s)
- Andrew F. Mead
- Department of Molecular Physiology and Biophysics, Larner College of Medicine, University of Vermont, Burlington, VT, USA
- Cardiovascular Research Institute, University of Vermont, Burlington, VT, USA
| | - Neil B. Wood
- Department of Molecular Physiology and Biophysics, Larner College of Medicine, University of Vermont, Burlington, VT, USA
| | - Shane R. Nelson
- Department of Molecular Physiology and Biophysics, Larner College of Medicine, University of Vermont, Burlington, VT, USA
- Cardiovascular Research Institute, University of Vermont, Burlington, VT, USA
| | - Bradley M. Palmer
- Department of Molecular Physiology and Biophysics, Larner College of Medicine, University of Vermont, Burlington, VT, USA
- Cardiovascular Research Institute, University of Vermont, Burlington, VT, USA
| | - Lin Yang
- National Synchrotron Light Source II, Brookhaven National Laboratory, Upton, NY, USA
| | - Samantha Beck Previs
- Department of Molecular Physiology and Biophysics, Larner College of Medicine, University of Vermont, Burlington, VT, USA
- Cardiovascular Research Institute, University of Vermont, Burlington, VT, USA
| | - Angela Ploysangngam
- Department of Molecular Physiology and Biophysics, Larner College of Medicine, University of Vermont, Burlington, VT, USA
| | - Guy G. Kennedy
- Department of Molecular Physiology and Biophysics, Larner College of Medicine, University of Vermont, Burlington, VT, USA
| | - Jennifer F. McAdow
- Department of Neurlogical Sciences, Larner College of Medicine, University of Vermont, Burlington, VT, USA
| | - Sarah M. Tremble
- Department of Electrical and Biomedical Engineering, College of Engineering and Mathematical Sciences, University of Vermont, Burlington, VT, USA
| | - Marcus A. Zimmermann
- Department of Molecular Physiology and Biophysics, Larner College of Medicine, University of Vermont, Burlington, VT, USA
- Cardiovascular Research Institute, University of Vermont, Burlington, VT, USA
| | - Marilyn J. Cipolla
- Department of Electrical and Biomedical Engineering, College of Engineering and Mathematical Sciences, University of Vermont, Burlington, VT, USA
- Department of Neurology, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
| | - Alicia M. Ebert
- Department of Biology, College of Arts and Sciences, University of Vermont, Burlington, VT, USA
| | - Aaron N. Johnson
- Department of Developmental Biology, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
| | - Christina A. Gurnett
- Department of Developmental Biology, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
| | - Michael J. Previs
- Department of Molecular Physiology and Biophysics, Larner College of Medicine, University of Vermont, Burlington, VT, USA
- Cardiovascular Research Institute, University of Vermont, Burlington, VT, USA
| | - David M. Warshaw
- Department of Molecular Physiology and Biophysics, Larner College of Medicine, University of Vermont, Burlington, VT, USA
- Cardiovascular Research Institute, University of Vermont, Burlington, VT, USA
| |
Collapse
|
7
|
Kanai SM, Garcia CR, Augustus MR, Sharafeldeen SA, Brooks EP, Sucharov J, Lencer ES, Nichols JT, Clouthier DE. The Gq/11 family of Gα subunits is necessary and sufficient for lower jaw development. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.17.611698. [PMID: 39345358 PMCID: PMC11430119 DOI: 10.1101/2024.09.17.611698] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
Vertebrate jaw development is coordinated by highly conserved ligand-receptor systems such as the peptide ligand Endothelin 1 (Edn1) and Endothelin receptor type A (Ednra), which are required for patterning of lower jaw structures. The Edn1/Ednra signaling pathway establishes the identity of lower jaw progenitor cells by regulating expression of numerous patterning genes, but the intracellular signaling mechanisms linking receptor activation to gene regulation remain poorly understood. As a first step towards elucidating this mechanism, we examined the function of the Gq/11 family of Gα subunits in zebrafish using pharmacological inhibition and genetic ablation of Gq/11 activity and transgenic induction of a constitutively active Gq protein in edn1 -/- embryos. Genetic loss of Gq/11 activity fully recapitulated the edn1 -/- phenotype, with genes encoding G11 being most essential. Furthermore, inducing Gq activity in edn1 -/- embryos not only restored Edn1/Ednra-dependent jaw structures and gene expression signatures but also caused homeosis of the upper jaw structure into a lower jaw-like structure. These results indicate that Gq/11 is necessary and sufficient to mediate the lower jaw patterning mechanism for Ednra in zebrafish.
Collapse
Affiliation(s)
- Stanley M. Kanai
- Department of Craniofacial Biology, School of Dental Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO USA
| | - Chloe R. Garcia
- Department of Craniofacial Biology, School of Dental Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO USA
| | - MaCalia R. Augustus
- Department of Craniofacial Biology, School of Dental Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO USA
| | - Shujan A. Sharafeldeen
- Department of Craniofacial Biology, School of Dental Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO USA
| | - Elliott P. Brooks
- Department of Craniofacial Biology, School of Dental Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO USA
| | - Juliana Sucharov
- Department of Craniofacial Biology, School of Dental Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO USA
| | - Ezra S. Lencer
- Department of Biology, Lafayette College, Easton, PA USA
| | - James T. Nichols
- Department of Craniofacial Biology, School of Dental Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO USA
| | - David E. Clouthier
- Department of Craniofacial Biology, School of Dental Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO USA
| |
Collapse
|
8
|
Maekawa M, Saito S, Isobe D, Takemoto K, Miura Y, Dobashi Y, Yamasu K. The Oct4-related PouV gene, pou5f3, mediates isthmus development in zebrafish by directly and dynamically regulating pax2a. Cells Dev 2024; 179:203933. [PMID: 38908828 DOI: 10.1016/j.cdev.2024.203933] [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: 04/14/2024] [Revised: 05/24/2024] [Accepted: 06/13/2024] [Indexed: 06/24/2024]
Abstract
Using a transgenic zebrafish line harboring a heat-inducible dominant-interference pou5f3 gene (en-pou5f3), we reported that this PouV gene is involved in isthmus development at the midbrain-hindbrain boundary (MHB), which patterns the midbrain and cerebellum. Importantly, the functions of pou5f3 reportedly differ before and after the end of gastrulation. In the present study, we examined in detail the effects of en-pou5f3 induction on isthmus development during embryogenesis. When en-pou5f3 was induced around the end of gastrulation (bud stage), the isthmus was abrogated or deformed by the end of somitogenesis (24 hours post-fertilization). At this stage, the expression of MHB markers -- such as pax2a, fgf8a, wnt1, and gbx2 -- was absent in embryos lacking the isthmus structure, whereas it was present, although severely distorted, in embryos with a deformed isthmus. We further found that, after en-pou5f3 induction at late gastrulation, pax2a, fgf8a, and wnt1 were immediately and irreversibly downregulated, whereas the expression of en2a and gbx2 was reduced only weakly and slowly. Induction of en-pou5f3 at early somite stages also immediately downregulated MHB genes, particularly pax2a, but their expression was restored later. Overall, the data suggested that pou5f3 directly upregulates at least pax2a and possibly fgf8a and wnt1, which function in parallel in establishing the MHB, and that the role of pou5f3 dynamically changes around the end of gastrulation. We next examined the transcriptional regulation of pax2a using both in vitro and in vivo reporter analyses; the results showed that two upstream 1.0-kb regions with sequences conserved among vertebrates specifically drove transcription at the MHB. These reporter analyses confirmed that development of the isthmic organizer is regulated by PouV through direct regulation of pax2/pax2a in vertebrate embryos.
Collapse
Affiliation(s)
- Masato Maekawa
- Division of Life Science, Graduate School of Science and Engineering, Saitama University, Shimo-Okubo, Sakura-ku, Saitama City, Saitama 338-8570, Japan
| | - Shinji Saito
- Division of Life Science, Graduate School of Science and Engineering, Saitama University, Shimo-Okubo, Sakura-ku, Saitama City, Saitama 338-8570, Japan; Institute for Vaccine Research and Development, Hokkaido University, N21, W11, Kita-ku, Sapporo, Hokkaido 001-0021, Japan
| | - Daiki Isobe
- Division of Life Science, Graduate School of Science and Engineering, Saitama University, Shimo-Okubo, Sakura-ku, Saitama City, Saitama 338-8570, Japan
| | - Kazumasa Takemoto
- Division of Life Science, Graduate School of Science and Engineering, Saitama University, Shimo-Okubo, Sakura-ku, Saitama City, Saitama 338-8570, Japan; Department of Physiology and Neurobiology, University of Connecticut, 75 North Eagleville Road, U3156, Storrs, CT 06269, USA
| | - Yuhei Miura
- Division of Life Science, Graduate School of Science and Engineering, Saitama University, Shimo-Okubo, Sakura-ku, Saitama City, Saitama 338-8570, Japan
| | - Yurie Dobashi
- Division of Life Science, Graduate School of Science and Engineering, Saitama University, Shimo-Okubo, Sakura-ku, Saitama City, Saitama 338-8570, Japan
| | - Kyo Yamasu
- Division of Life Science, Graduate School of Science and Engineering, Saitama University, Shimo-Okubo, Sakura-ku, Saitama City, Saitama 338-8570, Japan.
| |
Collapse
|
9
|
Gillespie W, Zhang Y, Ruiz OE, Cerda J, Ortiz-Guzman J, Turner WD, Largoza G, Sherman M, Mosser LE, Fujimoto E, Chien CB, Kwan KM, Arenkiel BR, Devine WP, Wythe JD. Multisite Assembly of Gateway Induced Clones (MAGIC): a flexible cloning toolbox with diverse applications in vertebrate model systems. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.13.603267. [PMID: 39026881 PMCID: PMC11257631 DOI: 10.1101/2024.07.13.603267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/20/2024]
Abstract
Here we present the Multisite Assembly of Gateway Induced Clones (MAGIC) system, which harnesses site-specific recombination-based cloning via Gateway technology for rapid, modular assembly of between 1 and 3 "Entry" vector components, all into a fourth, standard high copy "Destination" plasmid backbone. The MAGIC toolkit spans a range of in vitro and in vivo uses, from directing tunable gene expression, to driving simultaneous expression of microRNAs and fluorescent reporters, to enabling site-specific recombinase-dependent gene expression. All MAGIC system components are directly compatible with existing multisite gateway Tol2 systems currently used in zebrafish, as well as existing eukaryotic cell culture expression Destination plasmids, and available mammalian lentiviral and adenoviral Destination vectors, allowing rapid cross-species experimentation. Moreover, herein we describe novel vectors with flanking piggyBac transposon elements for stable genomic integration in vitro or in vivo when used with piggyBac transposase. Collectively, the MAGIC system facilitates transgenesis in cultured mammalian cells, electroporated mouse and chick embryos, as well as in injected zebrafish embryos, enabling the rapid generation of innovative DNA constructs for biological research due to a shared, common plasmid platform.
Collapse
|
10
|
Shaw DK, Saraswathy VM, McAdow AR, Zhou L, Park D, Mote R, Johnson AN, Mokalled MH. Elevated phagocytic capacity directs innate spinal cord repair. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.11.598515. [PMID: 38915507 PMCID: PMC11195157 DOI: 10.1101/2024.06.11.598515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/26/2024]
Abstract
Immune cells elicit a continuum of transcriptional and functional states after spinal cord injury (SCI). In mammals, inefficient debris clearance and chronic inflammation impede recovery and overshadow pro-regenerative immune functions. We found that, unlike mammals, zebrafish SCI elicits transient immune activation and efficient debris clearance, without causing chronic inflammation. Single-cell transcriptomics and inducible genetic ablation showed zebrafish macrophages are highly phagocytic and required for regeneration. Cross-species comparisons between zebrafish and mammalian macrophages identified transcription and immune response regulator ( tcim ) as a macrophage-enriched zebrafish gene. Genetic deletion of zebrafish tcim impairs phagocytosis and regeneration, causes aberrant and chronic immune activation, and can be rescued by transplanting wild-type immune precursors into tcim mutants. Conversely, genetic expression of human TCIM accelerates debris clearance and regeneration by reprogramming myeloid precursors into activated phagocytes. This study establishes a central requirement for elevated phagocytic capacity to achieve innate spinal cord repair.
Collapse
|
11
|
Mead AF, Wood NB, Nelson SR, Palmer BM, Yang L, Previs SB, Ploysangngam A, Kennedy GG, McAdow JF, Tremble SM, Cipolla MJ, Ebert AM, Johnson AN, Gurnett CA, Previs MJ, Warshaw DM. Functional role of myosin-binding protein H in thick filaments of developing vertebrate fast-twitch skeletal muscle. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.10.593199. [PMID: 38798399 PMCID: PMC11118323 DOI: 10.1101/2024.05.10.593199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
Myosin-binding protein H (MyBP-H) is a component of the vertebrate skeletal muscle sarcomere with sequence and domain homology to myosin-binding protein C (MyBP-C). Whereas skeletal muscle isoforms of MyBP-C (fMyBP-C, sMyBP-C) modulate muscle contractility via interactions with actin thin filaments and myosin motors within the muscle sarcomere "C-zone," MyBP-H has no known function. This is in part due to MyBP-H having limited expression in adult fast-twitch muscle and no known involvement in muscle disease. Quantitative proteomics reported here reveal MyBP-H is highly expressed in prenatal rat fast-twitch muscles and larval zebrafish, suggesting a conserved role in muscle development, and promoting studies to define its function. We take advantage of the genetic control of the zebrafish model and a combination of structural, functional, and biophysical techniques to interrogate the role of MyBP-H. Transgenic, FLAG-tagged MyBP-H or fMyBP-C both localize to the C-zones in larval myofibers, whereas genetic depletion of endogenous MyBP-H or fMyBP-C leads to increased accumulation of the other, suggesting competition for C-zone binding sites. Does MyBP-H modulate contractility from the C-zone? Globular domains critical to MyBP-C's modulatory functions are absent from MyBP-H, suggesting MyBP-H may be functionally silent. However, our results suggest an active role. Small angle x-ray diffraction of intact larval tails revealed MyBP-H contributes to the compression of the myofilament lattice accompanying stretch or contraction, while in vitro motility experiments indicate MyBP-H shares MyBP-C's capacity as a molecular "brake". These results provide new insights and raise questions about the role of the C-zone during muscle development.
Collapse
Affiliation(s)
- Andrew F. Mead
- Department of Molecular Physiology and Biophysics, Larner College of Medicine, University of Vermont, Burlington, VT 05405
- Cardiovascular Research Institute, University of Vermont, Burlington, VT 05405
| | - Neil B. Wood
- Department of Molecular Physiology and Biophysics, Larner College of Medicine, University of Vermont, Burlington, VT 05405
| | - Shane R. Nelson
- Department of Molecular Physiology and Biophysics, Larner College of Medicine, University of Vermont, Burlington, VT 05405
- Cardiovascular Research Institute, University of Vermont, Burlington, VT 05405
| | - Bradley M. Palmer
- Department of Molecular Physiology and Biophysics, Larner College of Medicine, University of Vermont, Burlington, VT 05405
- Cardiovascular Research Institute, University of Vermont, Burlington, VT 05405
| | - Lin Yang
- National Synchrotron Light Source II, Brookhaven National Laboratory, Upton, NY 11973
| | - Samantha Beck Previs
- Department of Molecular Physiology and Biophysics, Larner College of Medicine, University of Vermont, Burlington, VT 05405
- Cardiovascular Research Institute, University of Vermont, Burlington, VT 05405
| | - Angela Ploysangngam
- Department of Molecular Physiology and Biophysics, Larner College of Medicine, University of Vermont, Burlington, VT 05405
| | - Guy G. Kennedy
- Department of Molecular Physiology and Biophysics, Larner College of Medicine, University of Vermont, Burlington, VT 05405
| | - Jennifer F. McAdow
- Department of Neurlogical Sciences, Larner College of Medicine, University of Vermont, Burlington, VT 05405
| | - Sarah M. Tremble
- Department of Electrical and Biomedical Engineering, College of Engineering and Mathematical Sciences, University of Vermont, Burlington, VT 05405
| | - Marilyn J. Cipolla
- Department of Electrical and Biomedical Engineering, College of Engineering and Mathematical Sciences, University of Vermont, Burlington, VT 05405
- Department of Neurology, Washington University School of Medicine in St. Louis, St. Louis, MO 63110
| | - Alicia M. Ebert
- Department of Biology, College of Arts and Sciences, University of Vermont, Burlington, VT 05405
| | - Aaron N. Johnson
- Department of Developmental Biology, Washington University School of Medicine in St. Louis, St. Louis, MO 63110
| | - Christina A. Gurnett
- Department of Developmental Biology, Washington University School of Medicine in St. Louis, St. Louis, MO 63110
| | - Michael J. Previs
- Department of Molecular Physiology and Biophysics, Larner College of Medicine, University of Vermont, Burlington, VT 05405
- Cardiovascular Research Institute, University of Vermont, Burlington, VT 05405
| | - David M. Warshaw
- Department of Molecular Physiology and Biophysics, Larner College of Medicine, University of Vermont, Burlington, VT 05405
- Cardiovascular Research Institute, University of Vermont, Burlington, VT 05405
| |
Collapse
|
12
|
Chen H, Xie Y, Zhang M, Huang J, Jiang W, Zhang R, Li C, Du X, Chen H, Nie Q, Liang S, Tan Q, Yang J, Jin M, Huang S, Kuang L, Su N, Qi H, Luo X, Xu X, Deng C, Chen L, Luo F. An Hsp70 promoter-based mouse for heat shock-induced gene modulation. J Mol Med (Berl) 2024; 102:693-707. [PMID: 38492027 DOI: 10.1007/s00109-024-02433-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 02/11/2024] [Accepted: 02/13/2024] [Indexed: 03/18/2024]
Abstract
Physical therapy is extensively employed in clinical settings. Nevertheless, the absence of suitable animal models has resulted in an incomplete understanding of the in vivo mechanisms and cellular distribution that respond to physical stimuli. The objective of this research was to create a mouse model capable of indicating the cells affected by physical stimuli. In this study, we successfully established a mouse line based on the heat shock protein 70 (Hsp70) promoter, wherein the expression of CreERT2 can be induced by physical stimuli. Following stimulation of the mouse tail, ear, or cultured calvarias with heat shock (generated by heating, ultrasound, or laser), a distinct Cre-mediated excision was observed in cells stimulated by these physical factors with minimal occurrence of leaky reporter expression. The application of heat shock to Hsp70-CreERT2; FGFR2-P253R double transgenic mice or Hsp70-CreERT2 mice infected with AAV-BMP4 at calvarias induced the activation of Cre-dependent mutant FGFR2-P253R or BMP4 respectively, thereby facilitating the premature closure of cranial sutures or the repair of calvarial defects. This novel mouse line holds significant potential for investigating the underlying mechanisms of physical therapy, tissue repair and regeneration, lineage tracing, and targeted modulation of gene expression of cells in local tissue stimulated by physical factor at the interested time points. KEY MESSAGES: In the study, an Hsp70-CreERT2 transgenic mouse was generated for heat shock-induced gene modulation. Heat shock, ultrasound, and laser stimulation effectively activated Cre expression in Hsp70-CreERT2; reporter mice, which leads to deletion of floxed DNA sequence in the tail, ear, and cultured calvaria tissues of mice. Local laser stimuli on cultured calvarias effectively induce Fgfr2-P253R expression in Hsp70-mTmG-Fgfr2-P253R mice and result in accelerated premature closure of cranial suture. Heat shock activated AAV9-FLEX-BMP4 expression and subsequently promoted the repair of calvarial defect of Hsp70-CreERT2; Rosa26-mTmG mice.
Collapse
Affiliation(s)
- Hangang Chen
- Laboratory of Wound Repair and Rehabilitation Medicine, Daping Hospital, Third Military Medical University, Chongqing, 400042, China
| | - Yangli Xie
- Laboratory of Wound Repair and Rehabilitation Medicine, Daping Hospital, Third Military Medical University, Chongqing, 400042, China
- State Key Laboratory of Trauma, Burn and Combined Injury, Third Military Medical University, Chongqing, 400038, China
| | - Mei Zhang
- Laboratory of Wound Repair and Rehabilitation Medicine, Daping Hospital, Third Military Medical University, Chongqing, 400042, China
| | - Junlan Huang
- Laboratory of Wound Repair and Rehabilitation Medicine, Daping Hospital, Third Military Medical University, Chongqing, 400042, China
| | - Wanling Jiang
- Department of Chinese Medicine Rehabilitation, Chongqing Emergency Medical Center, Chongqing University Central Hospital), Chongqing, 400042, China
| | - Ruobin Zhang
- Laboratory of Wound Repair and Rehabilitation Medicine, Daping Hospital, Third Military Medical University, Chongqing, 400042, China
| | - Can Li
- Laboratory of Wound Repair and Rehabilitation Medicine, Daping Hospital, Third Military Medical University, Chongqing, 400042, China
| | - Xiaolan Du
- Laboratory of Wound Repair and Rehabilitation Medicine, Daping Hospital, Third Military Medical University, Chongqing, 400042, China
- State Key Laboratory of Trauma, Burn and Combined Injury, Third Military Medical University, Chongqing, 400038, China
| | - Hua Chen
- Laboratory of Wound Repair and Rehabilitation Medicine, Daping Hospital, Third Military Medical University, Chongqing, 400042, China
| | - Qiang Nie
- Laboratory of Wound Repair and Rehabilitation Medicine, Daping Hospital, Third Military Medical University, Chongqing, 400042, China
| | - Sen Liang
- Laboratory of Wound Repair and Rehabilitation Medicine, Daping Hospital, Third Military Medical University, Chongqing, 400042, China
| | - Qiaoyan Tan
- Laboratory of Wound Repair and Rehabilitation Medicine, Daping Hospital, Third Military Medical University, Chongqing, 400042, China
- State Key Laboratory of Trauma, Burn and Combined Injury, Third Military Medical University, Chongqing, 400038, China
| | - Jing Yang
- Laboratory of Wound Repair and Rehabilitation Medicine, Daping Hospital, Third Military Medical University, Chongqing, 400042, China
- State Key Laboratory of Trauma, Burn and Combined Injury, Third Military Medical University, Chongqing, 400038, China
| | - Min Jin
- Laboratory of Wound Repair and Rehabilitation Medicine, Daping Hospital, Third Military Medical University, Chongqing, 400042, China
- State Key Laboratory of Trauma, Burn and Combined Injury, Third Military Medical University, Chongqing, 400038, China
| | - Shuo Huang
- Laboratory of Wound Repair and Rehabilitation Medicine, Daping Hospital, Third Military Medical University, Chongqing, 400042, China
| | - Liang Kuang
- Laboratory of Wound Repair and Rehabilitation Medicine, Daping Hospital, Third Military Medical University, Chongqing, 400042, China
| | - Nan Su
- Laboratory of Wound Repair and Rehabilitation Medicine, Daping Hospital, Third Military Medical University, Chongqing, 400042, China
- State Key Laboratory of Trauma, Burn and Combined Injury, Third Military Medical University, Chongqing, 400038, China
| | - Huabing Qi
- Laboratory of Wound Repair and Rehabilitation Medicine, Daping Hospital, Third Military Medical University, Chongqing, 400042, China
- State Key Laboratory of Trauma, Burn and Combined Injury, Third Military Medical University, Chongqing, 400038, China
| | - Xiaoqing Luo
- Laboratory of Wound Repair and Rehabilitation Medicine, Daping Hospital, Third Military Medical University, Chongqing, 400042, China
| | - Xiaoling Xu
- Faculty of Health Sciences, University of Macau, Macau SAR, China
| | - Chuxia Deng
- Faculty of Health Sciences, University of Macau, Macau SAR, China
| | - Lin Chen
- Laboratory of Wound Repair and Rehabilitation Medicine, Daping Hospital, Third Military Medical University, Chongqing, 400042, China.
- State Key Laboratory of Trauma, Burn and Combined Injury, Third Military Medical University, Chongqing, 400038, China.
| | - Fengtao Luo
- Laboratory of Wound Repair and Rehabilitation Medicine, Daping Hospital, Third Military Medical University, Chongqing, 400042, China.
- State Key Laboratory of Trauma, Burn and Combined Injury, Third Military Medical University, Chongqing, 400038, China.
| |
Collapse
|
13
|
Benman W, Iyengar P, Mumford T, Huang Z, Bugaj LJ. Multiplexed dynamic control of temperature to probe and observe mammalian cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.18.580877. [PMID: 38562729 PMCID: PMC10983861 DOI: 10.1101/2024.02.18.580877] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
Temperature is a critical parameter for biological function, yet there is a lack of approaches to modulate the temperature of biological specimens in a dynamic and high-throughput manner. We present the thermoPlate, a device for programmable control of temperature in each well of a 96-well plate, in a manner compatible with mammalian cell culture and live cell imaging. The thermoPlate maintains precise feedback control of temperature patterns independently in each well, with minutes-scale heating and cooling through ΔT ~15-20°C. A computational model that predicts thermal diffusion guides optimal design of heating protocols. The thermoPlate allowed systematic characterization of both synthetic and natural thermo-responsive systems. We first used the thermoPlate in conjunction with live-cell microscopy to characterize the rapid temperature-dependent phase separation of a synthetic elastin-like polypeptide (ELP53). We then measured stress granule (SG) formation in response to heat stress, observing differences in SG dynamics with each increasing degree of stress. We observed adaptive formation of SGs, whereby SGs formed but then dissolved in response to persistent heat stress (≥ 42°C). SG adaptation revealed a biochemical memory of stress that depended on both the time and temperature of heat shock. Stress memories continued to form even after the removal of heat and persisted for 6-9 hours before dissipating. The capabilities and open-source nature of the thermoPlate will empower the study and engineering of a wide range of thermoresponsive phenomena.
Collapse
Affiliation(s)
- William Benman
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Pavan Iyengar
- Department of Chemistry, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Thomas Mumford
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Zikang Huang
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Lukasz J. Bugaj
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Institute of Regenerative Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| |
Collapse
|
14
|
Constanty F, Wu B, Wei KH, Lin IT, Dallmann J, Guenther S, Lautenschlaeger T, Priya R, Lai SL, Stainier DYR, Beisaw A. Border-zone cardiomyocytes and macrophages contribute to remodeling of the extracellular matrix to promote cardiomyocyte invasion during zebrafish cardiac regeneration. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.12.584570. [PMID: 38559277 PMCID: PMC10980021 DOI: 10.1101/2024.03.12.584570] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
Despite numerous advances in our understanding of zebrafish cardiac regeneration, an aspect that remains less studied is how regenerating cardiomyocytes invade, and eventually replace, the collagen-containing fibrotic tissue following injury. Here, we provide an in-depth analysis of the process of cardiomyocyte invasion using live-imaging and histological approaches. We observed close interactions between protruding cardiomyocytes and macrophages at the wound border zone, and macrophage-deficient irf8 mutant zebrafish exhibited defects in extracellular matrix (ECM) remodeling and cardiomyocyte protrusion into the injured area. Using a resident macrophage ablation model, we show that defects in ECM remodeling at the border zone and subsequent cardiomyocyte protrusion can be partly attributed to a population of resident macrophages. Single-cell RNA-sequencing analysis of cells at the wound border revealed a population of cardiomyocytes and macrophages with fibroblast-like gene expression signatures, including the expression of genes encoding ECM structural proteins and ECM-remodeling proteins. The expression of mmp14b , which encodes a membrane-anchored matrix metalloproteinase, was restricted to cells in the border zone, including cardiomyocytes, macrophages, fibroblasts, and endocardial/endothelial cells. Genetic deletion of mmp14b led to a decrease in 1) macrophage recruitment to the border zone, 2) collagen degradation at the border zone, and 3) subsequent cardiomyocyte invasion. Furthermore, cardiomyocyte-specific overexpression of mmp14b was sufficient to enhance cardiomyocyte invasion into the injured tissue and along the apical surface of the wound. Altogether, our data shed important insights into the process of cardiomyocyte invasion of the collagen-containing injured tissue during cardiac regeneration. They further suggest that cardiomyocytes and resident macrophages contribute to ECM remodeling at the border zone to promote cardiomyocyte replenishment of the fibrotic injured tissue.
Collapse
|
15
|
Zhang A, Lu J, Feng S, Yu H, Yu T, Zhao S, Chen K, Huang Z, Xu J, Qu JY, Wen Z. Fli1 acts in parallel with Pu.1 to control macrophage and neutrophil fate in zebrafish. J Genet Genomics 2024; 51:359-362. [PMID: 37944856 DOI: 10.1016/j.jgg.2023.11.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 11/03/2023] [Accepted: 11/04/2023] [Indexed: 11/12/2023]
Affiliation(s)
- Ao Zhang
- Division of Life Science, Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China
| | - Jingao Lu
- Division of Cell, Developmental and Integrative Biology, School of Medicine, South China University of Technology, Guangzhou, Guangdong 510006, China
| | - Shachuan Feng
- Department of Immunology and Microbiology, School of Life Science, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
| | - Huapeng Yu
- Department of Immunology and Microbiology, School of Life Science, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
| | - Tao Yu
- Biomedical Research Institute, Shenzhen Peking University-Hong Kong University of Science and Technology Medical Center, Shenzhen, Guangdong 518036, China
| | - Shizheng Zhao
- Biomedical Research Institute, Shenzhen Peking University-Hong Kong University of Science and Technology Medical Center, Shenzhen, Guangdong 518036, China
| | - Kemin Chen
- Division of Cell, Developmental and Integrative Biology, School of Medicine, South China University of Technology, Guangzhou, Guangdong 510006, China
| | - Zhibin Huang
- Division of Cell, Developmental and Integrative Biology, School of Medicine, South China University of Technology, Guangzhou, Guangdong 510006, China
| | - Jin Xu
- Division of Cell, Developmental and Integrative Biology, School of Medicine, South China University of Technology, Guangzhou, Guangdong 510006, China
| | - Jianan Y Qu
- Department of Electronic and Computer Engineering, Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China
| | - Zilong Wen
- Division of Life Science, Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China; Department of Immunology and Microbiology, School of Life Science, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China; Greater Bay Biomedical Innocenter, Shenzhen Bay Laboratory, Shenzhen, Guangdong 518067, China.
| |
Collapse
|
16
|
Nguyen LTM, Hassan S, Pan H, Wu S, Wen Z. Interplay of Zeb2a, Id2a and Batf3 regulates microglia and dendritic cell development in the zebrafish brain. Development 2024; 151:dev201829. [PMID: 38240311 DOI: 10.1242/dev.201829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Accepted: 01/08/2024] [Indexed: 02/06/2024]
Abstract
In vertebrates, the central nervous system (CNS) harbours various immune cells, including parenchymal microglia, perivascular macrophages and dendritic cells, which act in coordination to establish an immune network to regulate neurogenesis and neural function, and to maintain the homeostasis of the CNS. Recent single cell transcriptomic profiling has revealed that the adult zebrafish CNS contains microglia, plasmacytoid dendritic cells (pDCs) and two conventional dendritic cells (cDCs), ccl35+ cDCs and cnn3a+cDCs. However, how these distinct myeloid cells are established in the adult zebrafish CNS remains incompletely defined. Here, we show that the Inhibitor of DNA binding 2a (Id2a) is essential for the development of pDCs and cDCs but is dispensable for the formation of microglia, whereas the Basic leucine zipper transcription factor ATF-like 3 (Batf3) acts downstream of id2a and is required exclusively for the formation of the cnn3a+ cDC subset. In contrast, the Zinc finger E-box-binding homeobox 2a (Zeb2a) promotes the expansion of microglia and inhibits the DC specification, possibly through repressing id2a expression. Our study unravels the genetic networks that govern the development of microglia and brain-associated DCs in the zebrafish CNS.
Collapse
Affiliation(s)
- Linh Thi My Nguyen
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, the Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China
| | - Shaoli Hassan
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, the Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China
| | - Hongru Pan
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, the Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China
| | - Shuting Wu
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Zilong Wen
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, the Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China
- Greater Bay Biomedical Innocenter, Shenzhen Bay Laboratory, Shenzhen 518055, China
- Department of Immunology and Microbiology, School of Life Science, the Southern University of Science and Technology, Shenzhen 518000, China
| |
Collapse
|
17
|
Yaganoglu S, Kalyviotis K, Vagena-Pantoula C, Jülich D, Gaub BM, Welling M, Lopes T, Lachowski D, Tang SS, Del Rio Hernandez A, Salem V, Müller DJ, Holley SA, Vermot J, Shi J, Helassa N, Török K, Pantazis P. Highly specific and non-invasive imaging of Piezo1-dependent activity across scales using GenEPi. Nat Commun 2023; 14:4352. [PMID: 37468521 PMCID: PMC10356793 DOI: 10.1038/s41467-023-40134-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Accepted: 07/11/2023] [Indexed: 07/21/2023] Open
Abstract
Mechanosensing is a ubiquitous process to translate external mechanical stimuli into biological responses. Piezo1 ion channels are directly gated by mechanical forces and play an essential role in cellular mechanotransduction. However, readouts of Piezo1 activity are mainly examined by invasive or indirect techniques, such as electrophysiological analyses and cytosolic calcium imaging. Here, we introduce GenEPi, a genetically-encoded fluorescent reporter for non-invasive optical monitoring of Piezo1-dependent activity. We demonstrate that GenEPi has high spatiotemporal resolution for Piezo1-dependent stimuli from the single-cell level to that of the entire organism. GenEPi reveals transient, local mechanical stimuli in the plasma membrane of single cells, resolves repetitive contraction-triggered stimulation of beating cardiomyocytes within microtissues, and allows for robust and reliable monitoring of Piezo1-dependent activity in vivo. GenEPi will enable non-invasive optical monitoring of Piezo1 activity in mechanochemical feedback loops during development, homeostatic regulation, and disease.
Collapse
Affiliation(s)
- Sine Yaganoglu
- Department of Biosystems Science and Engineering, Eidgenössische Technische Hochschule (ETH) Zurich, Basel, Switzerland
| | | | | | - Dörthe Jülich
- Department of Molecular, Cellular and Developmental Biology, Yale University, New Haven, CT, USA
| | - Benjamin M Gaub
- Department of Biosystems Science and Engineering, Eidgenössische Technische Hochschule (ETH) Zurich, Basel, Switzerland
| | - Maaike Welling
- Department of Biosystems Science and Engineering, Eidgenössische Technische Hochschule (ETH) Zurich, Basel, Switzerland
- Department of Bioengineering, Imperial College London, London, UK
| | - Tatiana Lopes
- Section of Investigative Medicine, Department of Metabolism, Digestion, and Reproduction, Imperial College London, London, UK
| | | | - See Swee Tang
- Department of Bioengineering, Imperial College London, London, UK
| | | | - Victoria Salem
- Department of Bioengineering, Imperial College London, London, UK
| | - Daniel J Müller
- Department of Biosystems Science and Engineering, Eidgenössische Technische Hochschule (ETH) Zurich, Basel, Switzerland
| | - Scott A Holley
- Department of Molecular, Cellular and Developmental Biology, Yale University, New Haven, CT, USA
| | - Julien Vermot
- Department of Bioengineering, Imperial College London, London, UK
| | - Jian Shi
- Leeds Institute of Cardiovascular and Metabolic Medicine, LIGHT Laboratories, University of Leeds, Leeds, UK
| | - Nordine Helassa
- Molecular and Clinical Sciences Research Institute, St. George's, University of London, London, UK
- Department of Biochemistry, Cell and Systems Biology, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, UK
| | - Katalin Török
- Molecular and Clinical Sciences Research Institute, St. George's, University of London, London, UK
| | - Periklis Pantazis
- Department of Biosystems Science and Engineering, Eidgenössische Technische Hochschule (ETH) Zurich, Basel, Switzerland.
- Department of Bioengineering, Imperial College London, London, UK.
| |
Collapse
|
18
|
Gupta K, Datta A. An activity-based fluorescent sensor with a penta-coordinate N-donor binding site detects Cu ions in living systems. Chem Commun (Camb) 2023; 59:8282-8285. [PMID: 37318277 DOI: 10.1039/d3cc02201c] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
An activity-based sensor afforded a 63 times fluorescence-enhancement with Cu2+/Cu+ ions and could image Cu2+/Cu+ in living cells and in a multicellular organism. The sensor functioned only in the presence of ambient dioxygen and glutathione, and the characterization of intermediates and products hinted toward a sensing mechanism involving a CuII hydroperoxo species.
Collapse
Affiliation(s)
- Kunika Gupta
- Department of Chemical Sciences, Tata Institute of Fundamental Research, 1 Homi Bhabha Road, Colaba, Mumbai-400005, India.
| | - Ankona Datta
- Department of Chemical Sciences, Tata Institute of Fundamental Research, 1 Homi Bhabha Road, Colaba, Mumbai-400005, India.
| |
Collapse
|
19
|
Blum N, Harris MP. Localized heterochrony integrates overgrowth potential of oncogenic clones. Dis Model Mech 2023; 16:dmm049793. [PMID: 36621776 PMCID: PMC9932785 DOI: 10.1242/dmm.049793] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Accepted: 12/23/2022] [Indexed: 01/10/2023] Open
Abstract
Somatic mutations occur frequently and can arise during embryogenesis, resulting in the formation of a patchwork of mutant clones. Such mosaicism has been implicated in a broad range of developmental anomalies; however, their etiology is poorly understood. Patients carrying a common somatic oncogenic mutation in either PIK3CA or AKT1 can present with disproportionally large digits or limbs. How mutant clones, carrying an oncogenic mutation that often drives unchecked proliferation, can lead to controlled and coordinated overgrowth is unknown. We use zebrafish to explore the growth dynamics of oncogenic clones during development. Here, in a subset of clones, we observed a local increase in proportion of the fin skeleton closely resembling overgrowth phenotypes in patients. We unravel the cellular and developmental mechanisms of these overgrowths, and pinpoint the cell type and timing of clonal expansion. Coordinated overgrowth is associated with rapid clone expansion during early pre-chondrogenic phase of bone development, inducing a heterochronic shift that drives the change in bone size. Our study details how development integrates and translates growth potential of oncogenic clones, thereby shaping the phenotypic consequences of somatic mutations.
Collapse
Affiliation(s)
- Nicola Blum
- Department of Orthopaedics, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA 02115, USA
- Department of Genetics, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA
| | - Matthew P. Harris
- Department of Orthopaedics, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA 02115, USA
- Department of Genetics, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA
| |
Collapse
|
20
|
Henke K, Farmer DT, Niu X, Kraus JM, Galloway JL, Youngstrom DW. Genetically engineered zebrafish as models of skeletal development and regeneration. Bone 2023; 167:116611. [PMID: 36395960 PMCID: PMC11080330 DOI: 10.1016/j.bone.2022.116611] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 11/01/2022] [Accepted: 11/08/2022] [Indexed: 11/16/2022]
Abstract
Zebrafish (Danio rerio) are aquatic vertebrates with significant homology to their terrestrial counterparts. While zebrafish have a centuries-long track record in developmental and regenerative biology, their utility has grown exponentially with the onset of modern genetics. This is exemplified in studies focused on skeletal development and repair. Herein, the numerous contributions of zebrafish to our understanding of the basic science of cartilage, bone, tendon/ligament, and other skeletal tissues are described, with a particular focus on applications to development and regeneration. We summarize the genetic strengths that have made the zebrafish a powerful model to understand skeletal biology. We also highlight the large body of existing tools and techniques available to understand skeletal development and repair in the zebrafish and introduce emerging methods that will aid in novel discoveries in skeletal biology. Finally, we review the unique contributions of zebrafish to our understanding of regeneration and highlight diverse routes of repair in different contexts of injury. We conclude that zebrafish will continue to fill a niche of increasing breadth and depth in the study of basic cellular mechanisms of skeletal biology.
Collapse
Affiliation(s)
- Katrin Henke
- Department of Orthopaedics, Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322, USA.
| | - D'Juan T Farmer
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, CA 90095, USA; Department of Orthopaedic Surgery, University of California, Los Angeles, CA 90095, USA.
| | - Xubo Niu
- Center for Regenerative Medicine, Department of Orthopaedic Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA.
| | - Jessica M Kraus
- Department of Orthopaedic Surgery, University of Connecticut Health Center, Farmington, CT 06030, USA.
| | - Jenna L Galloway
- Center for Regenerative Medicine, Department of Orthopaedic Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA.
| | - Daniel W Youngstrom
- Department of Orthopaedic Surgery, University of Connecticut Health Center, Farmington, CT 06030, USA.
| |
Collapse
|
21
|
Leiba J, Özbilgiç R, Hernández L, Demou M, Lutfalla G, Yatime L, Nguyen-Chi M. Molecular Actors of Inflammation and Their Signaling Pathways: Mechanistic Insights from Zebrafish. BIOLOGY 2023; 12:153. [PMID: 36829432 PMCID: PMC9952950 DOI: 10.3390/biology12020153] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 01/12/2023] [Accepted: 01/16/2023] [Indexed: 01/21/2023]
Abstract
Inflammation is a hallmark of the physiological response to aggressions. It is orchestrated by a plethora of molecules that detect the danger, signal intracellularly, and activate immune mechanisms to fight the threat. Understanding these processes at a level that allows to modulate their fate in a pathological context strongly relies on in vivo studies, as these can capture the complexity of the whole process and integrate the intricate interplay between the cellular and molecular actors of inflammation. Over the years, zebrafish has proven to be a well-recognized model to study immune responses linked to human physiopathology. We here provide a systematic review of the molecular effectors of inflammation known in this vertebrate and recapitulate their modes of action, as inferred from sterile or infection-based inflammatory models. We present a comprehensive analysis of their sequence, expression, and tissue distribution and summarize the tools that have been developed to study their function. We further highlight how these tools helped gain insights into the mechanisms of immune cell activation, induction, or resolution of inflammation, by uncovering downstream receptors and signaling pathways. These progresses pave the way for more refined models of inflammation, mimicking human diseases and enabling drug development using zebrafish models.
Collapse
|
22
|
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
|
23
|
Zhao S, Zhang A, Zhu H, Wen Z. The ETS transcription factor Spi2 regulates hematopoietic cell development in zebrafish. Development 2022; 149:276980. [DOI: 10.1242/dev.200881] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Accepted: 08/26/2022] [Indexed: 11/20/2022]
Abstract
ABSTRACT
The E26 transformation-specific or E-twenty-six (ETS) genes encode a superfamily of transcription factors involved in diverse biological processes. Here, we report the identification and characterization of a previously unidentified member of the ETS transcription factors, Spi2, that is found exclusively in the ray-finned fish kingdom. We show that the expression of spi2 is restricted to hemogenic endothelial cells (HECs) and to hematopoietic stem and progenitor cells (HSPCs) in zebrafish. Using bacteria artificial chromosome transgenesis, we generate a spi2 reporter line, TgBAC(spi2:P2a-GFP), which manifests the GFP pattern recapitulating the endogenous spi2 expression. Genetic ablation of spi2 has little effect on HEC formation and the endothelial-to-hematopoietic transition, but results in compromised proliferation of HSPCs in the caudal hematopoietic tissue (CHT) during early development and in severe myeloid lineage defect in adulthood. Epistatic analysis shows that spi2 acts downstream of runx1 in regulating HSPC development in the CHT. Our study identifies Spi2 as an essential regulator for definitive hematopoietic cell development and creates a TgBAC(spi2:P2a-GFP) reporter line for tracking HECs, HSPCs, myeloid cells and thrombocytes from early development to adulthood.
Collapse
Affiliation(s)
- Shizheng Zhao
- State Key Laboratory of Molecular Neuroscience, Hong Kong University of Science and Technology 1 Division of Life Science , , Clear Water Bay, Hong Kong , China
| | - Ao Zhang
- State Key Laboratory of Molecular Neuroscience, Hong Kong University of Science and Technology 1 Division of Life Science , , Clear Water Bay, Hong Kong , China
| | - Hao Zhu
- State Key Laboratory of Molecular Neuroscience, Hong Kong University of Science and Technology 1 Division of Life Science , , Clear Water Bay, Hong Kong , China
| | - Zilong Wen
- State Key Laboratory of Molecular Neuroscience, Hong Kong University of Science and Technology 1 Division of Life Science , , Clear Water Bay, Hong Kong , China
- Greater Bay Biomedical Innocenter, Shenzhen Bay Laboratory, Shenzhen Peking University−Hong Kong University of Science and Technology Medical Center 2 , Shenzhen 518055 , China
| |
Collapse
|
24
|
Hemogenic and aortic endothelium arise from a common hemogenic angioblast precursor and are specified by the Etv2 dosage. Proc Natl Acad Sci U S A 2022; 119:e2119051119. [PMID: 35333649 PMCID: PMC9060440 DOI: 10.1073/pnas.2119051119] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
SignificanceHematopoietic stem cells (HSCs) are generated from specialized endothelial cells, called hemogenic endothelial cells (HECs). It has been debated whether HECs and non-HSC-forming conventional endothelial cells (cECs) arise from a common precursor or represent distinct lineages. Moreover, the molecular basis underlying their distinct fate determination is poorly understood. We use photoconvertible labeling, time-lapse imaging, and single-cell RNA-sequencing analysis to trace the lineage of HECs. We discovered that HECs and cECs arise from a common hemogenic angioblast precursor, and their distinct fate is determined by high or low dosage of Etv2, respectively. Our results illuminate the lineage origin and a mechanism on the fate determination of HECs, which may enhance the understanding on the ontogeny of HECs in vertebrates.
Collapse
|
25
|
Ahmed M, Kojima Y, Masai I. Strip1 regulates retinal ganglion cell survival by suppressing Jun-mediated apoptosis to promote retinal neural circuit formation. eLife 2022; 11:74650. [PMID: 35314028 PMCID: PMC8940179 DOI: 10.7554/elife.74650] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Accepted: 02/04/2022] [Indexed: 12/13/2022] Open
Abstract
In the vertebrate retina, an interplay between retinal ganglion cells (RGCs), amacrine (AC), and bipolar (BP) cells establishes a synaptic layer called the inner plexiform layer (IPL). This circuit conveys signals from photoreceptors to visual centers in the brain. However, the molecular mechanisms involved in its development remain poorly understood. Striatin-interacting protein 1 (Strip1) is a core component of the striatin-interacting phosphatases and kinases (STRIPAK) complex, and it has shown emerging roles in embryonic morphogenesis. Here, we uncover the importance of Strip1 in inner retina development. Using zebrafish, we show that loss of Strip1 causes defects in IPL formation. In strip1 mutants, RGCs undergo dramatic cell death shortly after birth. AC and BP cells subsequently invade the degenerating RGC layer, leading to a disorganized IPL. Mechanistically, zebrafish Strip1 interacts with its STRIPAK partner, Striatin 3 (Strn3), and both show overlapping functions in RGC survival. Furthermore, loss of Strip1 or Strn3 leads to activation of the proapoptotic marker, Jun, within RGCs, and Jun knockdown rescues RGC survival in strip1 mutants. In addition to its function in RGC maintenance, Strip1 is required for RGC dendritic patterning, which likely contributes to proper IPL formation. Taken together, we propose that a series of Strip1-mediated regulatory events coordinates inner retinal circuit formation by maintaining RGCs during development, which ensures proper positioning and neurite patterning of inner retinal neurons. The back of the eye is lined with an intricate tissue known as the retina, which consists of carefully stacked neurons connecting to each other in well-defined ‘synaptic’ layers. Near the surface, photoreceptors cells detect changes in light levels, before passing this information through the inner plexiform layer to retinal ganglion cells (or RGCs) below. These neurons will then relay the visual signals to the brain. Despite the importance of this inner retinal circuit, little is known about how it is created as an organism develops. As a response, Ahmed et al. sought to identify which genes are essential to establish the inner retinal circuit, and how their absence affects retinal structure. To do this, they introduced random errors in the genetic code of zebrafish and visualised the resulting retinal circuits in these fast-growing, translucent fish. Initial screening studies found fish with mutations in a gene encoding a protein called Strip1 had irregular layering of the inner retina. Further imaging experiments to pinpoint the individual neurons affected showed that in zebrafish without Strip1, RGCs died in the first few days of development. Consequently, other neurons moved into the RGC layer to replace the lost cells, leading to layering defects. Ahmed et al. concluded that Strip1 promotes RGC survival and thereby coordinates proper positioning of neurons in the inner retina. In summary, these findings help to understand how the inner retina is wired; they could also shed light on the way other layered structures are established in the nervous system. Moreover, this study paves the way for future research investigating Strip1 as a potential therapeutic target to slow down the death of RGCs in conditions such as glaucoma.
Collapse
Affiliation(s)
- Mai Ahmed
- Developmental Neurobiology Unit, Okinawa Institute of Science and Technology Graduate University
| | - Yutaka Kojima
- Developmental Neurobiology Unit, Okinawa Institute of Science and Technology Graduate University
| | - Ichiro Masai
- Developmental Neurobiology Unit, Okinawa Institute of Science and Technology Graduate University
| |
Collapse
|
26
|
Feng C, Nita-Lazar M, González-Montalbán N, Wang J, Mancini J, Wang S, Ravindran C, Ahmed H, Vasta GR. Manipulating Galectin Expression in Zebrafish (Danio rerio). Methods Mol Biol 2022; 2442:425-443. [PMID: 35320539 DOI: 10.1007/978-1-0716-2055-7_23] [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: 06/14/2023]
Abstract
Techniques for disrupting gene expression are invaluable tools for the analysis of the biological role of a gene product. Because of its genetic tractability and multiple advantages over conventional mammalian models, the zebrafish (Danio rerio) is recognized as a powerful system for gaining new insight into diverse aspects of human health and disease. Among the multiple mammalian gene families for which the zebrafish has shown promise as an invaluable model for functional studies, the galectins have attracted great interest due to their participation in early development, regulation of immune homeostasis, and recognition of microbial pathogens. Galectins are β-galactosyl-binding lectins with a characteristic sequence motif in their carbohydrate recognition domains (CRDs), that constitute an evolutionary conserved family ubiquitous in eukaryotic taxa. Galectins are emerging as key players in the modulation of many important pathological processes, which include acute and chronic inflammatory diseases, autoimmunity and cancer, thus making them potential molecular targets for innovative drug discovery. Here, we provide a review of the current methods available for the manipulation of gene expression in the zebrafish, with a focus on gene knockdown [morpholino (MO)-derived antisense oligonucleotides] and knockout (CRISPR-Cas) technologies.
Collapse
Affiliation(s)
- Chiguang Feng
- Department of Microbiology and Immunology, Institute of Marine and Environmental Technology, University of Maryland Baltimore, Baltimore, MD, USA
| | - Mihai Nita-Lazar
- Department of Microbiology and Immunology, Institute of Marine and Environmental Technology, University of Maryland Baltimore, Baltimore, MD, USA
| | - Nuria González-Montalbán
- Department of Microbiology and Immunology, Institute of Marine and Environmental Technology, University of Maryland Baltimore, Baltimore, MD, USA
| | - Jingyu Wang
- Department of Microbiology and Immunology, Institute of Marine and Environmental Technology, University of Maryland Baltimore, Baltimore, MD, USA
| | - Justin Mancini
- Department of Microbiology and Immunology, Institute of Marine and Environmental Technology, University of Maryland Baltimore, Baltimore, MD, USA
| | - Sheng Wang
- Department of Microbiology and Immunology, Institute of Marine and Environmental Technology, University of Maryland Baltimore, Baltimore, MD, USA
- State Key Laboratory for Biocontrol, School of Marine Sciences, Sun Yat-Sen University, Guangzhou, People's Republic of China
| | - Chinnarajan Ravindran
- Department of Microbiology and Immunology, Institute of Marine and Environmental Technology, University of Maryland Baltimore, Baltimore, MD, USA
- Department of Marine Biotechnology, National Institute of Oceanography (CSIR), Dona Paula, Goa, India
| | - Hafiz Ahmed
- Department of Biochemistry, School of Medicine, Institute of Marine and Environmental Technology, University of Maryland Baltimore, Baltimore, MD, USA
| | - Gerardo R Vasta
- Department of Microbiology and Immunology, Institute of Marine and Environmental Technology, University of Maryland Baltimore, Baltimore, MD, USA.
| |
Collapse
|
27
|
Wnt/β-catenin signaling acts cell-autonomously to promote cardiomyocyte regeneration in the zebrafish heart. Dev Biol 2021; 481:226-237. [PMID: 34748730 DOI: 10.1016/j.ydbio.2021.11.001] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Revised: 10/15/2021] [Accepted: 11/02/2021] [Indexed: 12/22/2022]
Abstract
Zebrafish can achieve scar-free healing of heart injuries, and robustly replace all cardiomyocytes lost to injury via dedifferentiation and proliferation of mature cardiomyocytes. Previous studies suggested that Wnt/β-catenin signaling is active in the injured zebrafish heart, where it induces fibrosis and prevents cardiomyocyte cell cycling. Here, via targeting the destruction complex of the Wnt/β-catenin pathway with pharmacological and genetic tools, we demonstrate that Wnt/β-catenin activity is required for cardiomyocyte proliferation and dedifferentiation, as well as for maturation of the scar during regeneration. Using cardiomyocyte-specific conditional inhibition of the pathway, we show that Wnt/β-catenin signaling acts cell-autonomously to promote cardiomyocyte proliferation. Our results stand in contrast to previous reports and rather support a model in which Wnt/β-catenin signaling plays a positive role during heart regeneration in zebrafish.
Collapse
|
28
|
Yoshimatsu T, Bartel P, Schröder C, Janiak FK, St-Pierre F, Berens P, Baden T. Ancestral circuits for vertebrate color vision emerge at the first retinal synapse. SCIENCE ADVANCES 2021; 7:eabj6815. [PMID: 34644120 PMCID: PMC8514090 DOI: 10.1126/sciadv.abj6815] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
For color vision, retinal circuits separate information about intensity and wavelength. In vertebrates that use the full complement of four “ancestral” cone types, the nature and implementation of this computation remain poorly understood. Here, we establish the complete circuit architecture of outer retinal circuits underlying color processing in larval zebrafish. We find that the synaptic outputs of red and green cones efficiently rotate the encoding of natural daylight in a principal components analysis–like manner to yield primary achromatic and spectrally opponent axes, respectively. Blue cones are tuned to capture most remaining variance when opposed to green cones, while UV cone present a UV achromatic axis for prey capture. We note that fruitflies use essentially the same strategy. Therefore, rotating color space into primary achromatic and chromatic axes at the eye’s first synapse may thus be a fundamental principle of color vision when using more than two spectrally well-separated photoreceptor types.
Collapse
Affiliation(s)
| | - Philipp Bartel
- School of Life Sciences, University of Sussex, Brighton, UK
| | - Cornelius Schröder
- Institute of Ophthalmic Research, University of Tübingen, Tübingen, Germany
- Centre for Integrative Neuroscience, University of Tübingen, Tübingen, Germany
| | | | - François St-Pierre
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA
- Department of Electrical and Computer Engineering, Rice University, Houston, TX, USA
- Systems, Synthetic, and Physical Biology Program, Rice University, Houston, TX, USA
| | - Philipp Berens
- Institute of Ophthalmic Research, University of Tübingen, Tübingen, Germany
- Centre for Integrative Neuroscience, University of Tübingen, Tübingen, Germany
- Institute for Bioinformatics and Medical Informatics, University of Tübingen, Tübingen, Germany
| | - Tom Baden
- School of Life Sciences, University of Sussex, Brighton, UK
- Institute of Ophthalmic Research, University of Tübingen, Tübingen, Germany
- Corresponding author.
| |
Collapse
|
29
|
Hans S, Zöller D, Hammer J, Stucke J, Spieß S, Kesavan G, Kroehne V, Eguiguren JS, Ezhkova D, Petzold A, Dahl A, Brand M. Cre-Controlled CRISPR mutagenesis provides fast and easy conditional gene inactivation in zebrafish. Nat Commun 2021; 12:1125. [PMID: 33602923 PMCID: PMC7893016 DOI: 10.1038/s41467-021-21427-6] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2020] [Accepted: 01/22/2021] [Indexed: 12/12/2022] Open
Abstract
Conditional gene inactivation is a powerful tool to determine gene function when constitutive mutations result in detrimental effects. The most commonly used technique to achieve conditional gene inactivation employs the Cre/loxP system and its ability to delete DNA sequences flanked by two loxP sites. However, targeting a gene with two loxP sites is time and labor consuming. Here, we show Cre-Controlled CRISPR (3C) mutagenesis to circumvent these issues. 3C relies on gRNA and Cre-dependent Cas9-GFP expression from the same transgene. Exogenous or transgenic supply of Cre results in Cas9-GFP expression and subsequent mutagenesis of the gene of interest. The recombined cells become fluorescently visible enabling their isolation and subjection to various omics techniques. Hence, 3C mutagenesis provides a valuable alternative to the production of loxP-flanked alleles. It might even enable the conditional inactivation of multiple genes simultaneously and should be applicable to other model organisms amenable to single integration transgenesis.
Collapse
Affiliation(s)
- Stefan Hans
- Center for Molecular and Cellular Bioengineering (CMCB), Center for Regenerative Therapies Dresden (CRTD), Technische Universität Dresden, Dresden, Germany.
| | - Daniela Zöller
- Center for Molecular and Cellular Bioengineering (CMCB), Center for Regenerative Therapies Dresden (CRTD), Technische Universität Dresden, Dresden, Germany
| | - Juliane Hammer
- Center for Molecular and Cellular Bioengineering (CMCB), Center for Regenerative Therapies Dresden (CRTD), Technische Universität Dresden, Dresden, Germany
| | - Johanna Stucke
- Center for Molecular and Cellular Bioengineering (CMCB), Center for Regenerative Therapies Dresden (CRTD), Technische Universität Dresden, Dresden, Germany
| | - Sandra Spieß
- Center for Molecular and Cellular Bioengineering (CMCB), Center for Regenerative Therapies Dresden (CRTD), Technische Universität Dresden, Dresden, Germany
| | - Gokul Kesavan
- Center for Molecular and Cellular Bioengineering (CMCB), Center for Regenerative Therapies Dresden (CRTD), Technische Universität Dresden, Dresden, Germany
| | - Volker Kroehne
- Center for Molecular and Cellular Bioengineering (CMCB), Center for Regenerative Therapies Dresden (CRTD), Technische Universität Dresden, Dresden, Germany
| | - Juan Sebastian Eguiguren
- Center for Molecular and Cellular Bioengineering (CMCB), Center for Regenerative Therapies Dresden (CRTD), Technische Universität Dresden, Dresden, Germany
| | - Diana Ezhkova
- Center for Molecular and Cellular Bioengineering (CMCB), Center for Regenerative Therapies Dresden (CRTD), Technische Universität Dresden, Dresden, Germany
| | - Andreas Petzold
- Center for Molecular and Cellular Bioengineering (CMCB), DRESDEN-Concept Genome Center, Technische Universität Dresden, Dresden, Germany
| | - Andreas Dahl
- Center for Molecular and Cellular Bioengineering (CMCB), DRESDEN-Concept Genome Center, Technische Universität Dresden, Dresden, Germany
| | - Michael Brand
- Center for Molecular and Cellular Bioengineering (CMCB), Center for Regenerative Therapies Dresden (CRTD), Technische Universität Dresden, Dresden, Germany.
| |
Collapse
|
30
|
LaBelle J, Ramos-Martinez A, Shen K, Motta-Mena LB, Gardner KH, Materna SC, Woo S. TAEL 2.0: An Improved Optogenetic Expression System for Zebrafish. Zebrafish 2021; 18:20-28. [PMID: 33555975 DOI: 10.1089/zeb.2020.1951] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Inducible gene expression systems are valuable tools for studying biological processes. We previously developed an optogenetic gene expression system called TAEL that is optimized for use in zebrafish. When illuminated with blue light, TAEL transcription factors dimerize and activate gene expression downstream of the TAEL-responsive C120 promoter. By using light as the inducing agent, the TAEL/C120 system overcomes limitations of traditional inducible expression systems by enabling fine spatial and temporal regulation of gene expression. In this study, we describe ongoing efforts to improve the TAEL/C120 system. We made modifications to both the TAEL transcriptional activator and the C120 regulatory element, collectively referred to as TAEL 2.0. We demonstrate that TAEL 2.0 consistently induces higher levels of reporter gene expression and at a faster rate, but with comparable background and toxicity as the original TAEL system. With these improvements, we were able to create functional stable transgenic lines to express the TAEL 2.0 transcription factor either ubiquitously or with a tissue-specific promoter. We demonstrate that the ubiquitous line in particular can be used to induce expression at late embryonic and larval stages, addressing a major deficiency of the original TAEL system. This improved optogenetic expression system will be a broadly useful resource for the zebrafish community.
Collapse
Affiliation(s)
- Jesselynn LaBelle
- Department of Molecular Cell Biology, University of California Merced, Merced, California, USA
| | - Adela Ramos-Martinez
- Department of Molecular Cell Biology, University of California Merced, Merced, California, USA
| | - Kyle Shen
- Department of Molecular Cell Biology, University of California Merced, Merced, California, USA
| | | | - Kevin H Gardner
- Structural Biology Initiative, CUNY Advanced Science Research Center, New York, New York, USA
| | - Stefan C Materna
- Department of Molecular Cell Biology, University of California Merced, Merced, California, USA
| | - Stephanie Woo
- Department of Molecular Cell Biology, University of California Merced, Merced, California, USA
| |
Collapse
|
31
|
Tsata V, Möllmert S, Schweitzer C, Kolb J, Möckel C, Böhm B, Rosso G, Lange C, Lesche M, Hammer J, Kesavan G, Beis D, Guck J, Brand M, Wehner D. A switch in pdgfrb + cell-derived ECM composition prevents inhibitory scarring and promotes axon regeneration in the zebrafish spinal cord. Dev Cell 2021; 56:509-524.e9. [PMID: 33412105 DOI: 10.1016/j.devcel.2020.12.009] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2020] [Revised: 10/12/2020] [Accepted: 12/07/2020] [Indexed: 12/13/2022]
Abstract
In mammals, perivascular cell-derived scarring after spinal cord injury impedes axonal regrowth. In contrast, the extracellular matrix (ECM) in the spinal lesion site of zebrafish is permissive and required for axon regeneration. However, the cellular mechanisms underlying this interspecies difference have not been investigated. Here, we show that an injury to the zebrafish spinal cord triggers recruitment of pdgfrb+ myoseptal and perivascular cells in a PDGFR signaling-dependent manner. Interference with pdgfrb+ cell recruitment or depletion of pdgfrb+ cells inhibits axonal regrowth and recovery of locomotor function. Transcriptional profiling and functional experiments reveal that pdgfrb+ cells upregulate expression of axon growth-promoting ECM genes (cthrc1a and col12a1a/b) and concomitantly reduce synthesis of matrix molecules that are detrimental to regeneration (lum and mfap2). Our data demonstrate that a switch in ECM composition is critical for axon regeneration after spinal cord injury and identify the cellular source and components of the growth-promoting lesion ECM.
Collapse
Affiliation(s)
- Vasiliki Tsata
- Center for Regenerative Therapies TU Dresden, Technische Universität Dresden, 01307 Dresden, Germany; Developmental Biology, Clinical, Experimental Surgery and Translational Research Center, Biomedical Research Foundation Academy of Athens, 11527 Athens, Greece
| | - Stephanie Möllmert
- Max Planck Institute for the Science of Light, 91058 Erlangen, Germany; Max-Planck-Zentrum für Physik und Medizin, 91058 Erlangen, Germany
| | - Christine Schweitzer
- Max Planck Institute for the Science of Light, 91058 Erlangen, Germany; Max-Planck-Zentrum für Physik und Medizin, 91058 Erlangen, Germany
| | - Julia Kolb
- Max Planck Institute for the Science of Light, 91058 Erlangen, Germany; Max-Planck-Zentrum für Physik und Medizin, 91058 Erlangen, Germany
| | - Conrad Möckel
- Max Planck Institute for the Science of Light, 91058 Erlangen, Germany; Max-Planck-Zentrum für Physik und Medizin, 91058 Erlangen, Germany
| | - Benjamin Böhm
- Max Planck Institute for the Science of Light, 91058 Erlangen, Germany; Max-Planck-Zentrum für Physik und Medizin, 91058 Erlangen, Germany
| | - Gonzalo Rosso
- Max Planck Institute for the Science of Light, 91058 Erlangen, Germany; Max-Planck-Zentrum für Physik und Medizin, 91058 Erlangen, Germany; Institute of Physiology II, University of Münster, 48149 Münster, Germany
| | - Christian Lange
- Center for Regenerative Therapies TU Dresden, Technische Universität Dresden, 01307 Dresden, Germany
| | - Mathias Lesche
- DRESDEN-concept Genome Center c/o Center for Molecular and Cellular Bioengineering (CMCB), Technische Universität Dresden, 01307 Dresden, Germany
| | - Juliane Hammer
- Center for Regenerative Therapies TU Dresden, Technische Universität Dresden, 01307 Dresden, Germany
| | - Gokul Kesavan
- Center for Regenerative Therapies TU Dresden, Technische Universität Dresden, 01307 Dresden, Germany
| | - Dimitris Beis
- Developmental Biology, Clinical, Experimental Surgery and Translational Research Center, Biomedical Research Foundation Academy of Athens, 11527 Athens, Greece
| | - Jochen Guck
- Max Planck Institute for the Science of Light, 91058 Erlangen, Germany; Max-Planck-Zentrum für Physik und Medizin, 91058 Erlangen, Germany; Biotechnology Center, Center for Molecular and Cellular Bioengineering, Technische Universität Dresden, 01307 Dresden, Germany
| | - Michael Brand
- Center for Regenerative Therapies TU Dresden, Technische Universität Dresden, 01307 Dresden, Germany; Biotechnology Center, Center for Molecular and Cellular Bioengineering, Technische Universität Dresden, 01307 Dresden, Germany
| | - Daniel Wehner
- Center for Regenerative Therapies TU Dresden, Technische Universität Dresden, 01307 Dresden, Germany; Max Planck Institute for the Science of Light, 91058 Erlangen, Germany; Max-Planck-Zentrum für Physik und Medizin, 91058 Erlangen, Germany.
| |
Collapse
|
32
|
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: 2.0] [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
|
33
|
He S, Xu J, Qu JY, Wen Z. Lightening the way of hematopoiesis: Infrared laser-mediated lineage tracing with high spatial-temporal resolution. Exp Hematol 2020; 85:3-7. [PMID: 32437907 DOI: 10.1016/j.exphem.2020.04.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Revised: 04/29/2020] [Accepted: 04/29/2020] [Indexed: 12/13/2022]
Abstract
Hematopoiesis refers to the developmental process generating all blood lineages. In vertebrates, there are multiple waves of hematopoiesis, which emerge in distinct anatomic locations at different times and give rise to different blood lineages. In the last decade, numerous lineage-tracing studies have been conducted to investigate the hierarchical structure of the hematopoietic system. Yet, the majority of these lineage-tracing studies are not able to integrate the spatial-temporal information with the developmental potential of hematopoietic cells. With the newly developed infrared laser-evoked gene operator (IR-LEGO) microscope heating system, it is now possible to improve our understanding of hematopoiesis to spatial-temporal-controlled single-cell resolution. Here, we discuss the recent development of the IR-LEGO system and its applications in hematopoietic lineage tracing in vivo.
Collapse
Affiliation(s)
- Sicong He
- Department of Electronic and Computer Engineering, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China
| | - Jin Xu
- Division of Cell, Developmental and Integrative Biology, School of Medicine, South China University of Technology, Guangzhou, China
| | - Jianan Y Qu
- Department of Electronic and Computer Engineering, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China.
| | - Zilong Wen
- Division of Life Science, State Key Laboratory of Molecular Neuroscience and Center of Systems Biology and Human Health, Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China; Greater Bay Biomedical Innocenter, Shenzhen Bay Laboratory, Shenzhen Peking University-Hong Kong University of Science and Technology Medical Center, Shenzhen, China.
| |
Collapse
|
34
|
Lu H, Shagirova A, Goggi JL, Yeo HL, Roy S. Reissner fibre-induced urotensin signalling from cerebrospinal fluid-contacting neurons prevents scoliosis of the vertebrate spine. Biol Open 2020; 9:9/5/bio052027. [PMID: 32409296 PMCID: PMC7240301 DOI: 10.1242/bio.052027] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Reissner fibre (RF), discovered by the 19th-century German anatomist Ernst Reissner, is a filamentous structure present in cerebrospinal fluid (CSF). RF forms by aggregation of a glycoprotein called SCO-spondin (Sspo), but its function has remained enigmatic. Recent studies have shown that zebrafish sspo mutants develop a curved embryonic body axis. Zebrafish embryos with impaired cilia motility also develop curved bodies, which arises from failure of expression of urotensin related peptide (urp) genes in CSF-contacting neurons (CSF-cNs), impairing downstream signalling in trunk muscles. Here, we show that sspo mutants can survive into adulthood, but display severe curvatures of the vertebral column, resembling the common human spine disorder idiopathic scoliosis (IS). sspo mutants also exhibit significant reduction of urp gene expression from CSF-cNs. Consistent with epinephrine in CSF being bound by RF and required for urp expression, treating sspo mutants with this catecholamine rescued expression of the urp genes and axial defects. More strikingly, providing Urp2, specifically in the CSF-cNs, rescued body curvature of sspo homozygotes during larval stages as well as in the adult. These findings bridge existing gaps in our knowledge between cilia motility, RF, Urp signalling and spine deformities, and suggest that targeting the Urotensin pathway could provide novel therapeutic avenues for IS. Summary: Reissner fibre (RF) is a glycoprotein filament suspended in cerebrospinal fluid (CSF). We show that RF regulates zebrafish spine morphogenesis by regulating Urotensin signalling from CSF-contacting neurons of the spinal cord.
Collapse
Affiliation(s)
- Hao Lu
- Institute of Molecular and Cell Biology, Proteos, 61 Biopolis Drive, Singapore 138673
| | - Aidana Shagirova
- Institute of Molecular and Cell Biology, Proteos, 61 Biopolis Drive, Singapore 138673.,Department of Biological Sciences, National University of Singapore, 14 Science Drive 4, Singapore 117543
| | - Julian L Goggi
- Singapore Bioimaging Consortium, Helios, 11 Biopolis Way, Singapore 138667
| | - Hui Li Yeo
- Institute of Molecular and Cell Biology, Proteos, 61 Biopolis Drive, Singapore 138673
| | - Sudipto Roy
- Institute of Molecular and Cell Biology, Proteos, 61 Biopolis Drive, Singapore 138673 .,Department of Biological Sciences, National University of Singapore, 14 Science Drive 4, Singapore 117543.,Department of Pediatrics, Yong Loo Lin School of Medicine, National University of Singapore, 1E Kent Ridge Road, Singapore 119288
| |
Collapse
|
35
|
Heat-shock-induced tyrosinase gene ablation with CRISPR in zebrafish. Mol Genet Genomics 2020; 295:911-922. [PMID: 32367255 DOI: 10.1007/s00438-020-01681-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Accepted: 04/23/2020] [Indexed: 01/05/2023]
Abstract
Tyrosinase (TYR) converts L-tyrosine into 3,4-dihydroxyphenylalanine (L-DOPA) and L-DOPA into L-dopaquinone, which can produce melanin pigment. The abrogation of the functional activity of TYR can result in albino skin and eye diseases because of a deficiency in melanin pigment production. In this study, we developed and characterized an inducible knockout TYR platform comprising the heat-inducible heat-shock-promoter-70-driving CRISPR/Cas9 system and a zU6-promoter-driving tyr single guide RNA (sgRNA) system to investigate the temporal expression of TYR genes. To overcome the difficulty of identifying zebrafish germline integrations and facilitate the observation of Cas9 expression, heart-specific cmlc2:enhanced green fluorescent protein (EGFP; used to confirm tyr sgRNA expression) and two selectable markers (P2A-mCherry and internal ribosomal entry site-EGFP) were applied in our system. Heat shock treatment administered to Cas9 transgenic embryos induced mCherry or EGFP fluorescence expression throughout the embryos' bodies, and Cas9 protein was detected 1 h after heat shock treatment. Mutations were created by direct injection and line crossing, which led to mosaic and complete depigmentation phenotypes in approximately 50% and 100% of the embryos, respectively. Using our system, conditional TYR knockout in zebrafish was achieved efficiently and simply.
Collapse
|
36
|
Lee DA, Liu J, Hong Y, Lane JM, Hill AJ, Hou SL, Wang H, Oikonomou G, Pham U, Engle J, Saxena R, Prober DA. Evolutionarily conserved regulation of sleep by epidermal growth factor receptor signaling. SCIENCE ADVANCES 2019; 5:eaax4249. [PMID: 31763451 PMCID: PMC6853770 DOI: 10.1126/sciadv.aax4249] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Accepted: 09/17/2019] [Indexed: 05/03/2023]
Abstract
The genetic bases for most human sleep disorders and for variation in human sleep quantity and quality are largely unknown. Using the zebrafish, a diurnal vertebrate, to investigate the genetic regulation of sleep, we found that epidermal growth factor receptor (EGFR) signaling is necessary and sufficient for normal sleep levels and is required for the normal homeostatic response to sleep deprivation. We observed that EGFR signaling promotes sleep via mitogen-activated protein kinase/extracellular signal-regulated kinase and RFamide neuropeptide signaling and that it regulates RFamide neuropeptide expression and neuronal activity. Consistent with these findings, analysis of a large cohort of human genetic data from participants of European ancestry revealed that common variants in genes within the EGFR signaling pathway are associated with variation in human sleep quantity and quality. These results indicate that EGFR signaling and its downstream pathways play a central and ancient role in regulating sleep and provide new therapeutic targets for sleep disorders.
Collapse
Affiliation(s)
- Daniel A. Lee
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Justin Liu
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Young Hong
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Jacqueline M. Lane
- Center for Genomic Medicine and Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
- Program in Medical and Population Genetics, Broad Institute, Cambridge, MA 02142, USA
| | - Andrew J. Hill
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Sarah L. Hou
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Heming Wang
- Center for Genomic Medicine and Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
- Program in Medical and Population Genetics, Broad Institute, Cambridge, MA 02142, USA
| | - Grigorios Oikonomou
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Uyen Pham
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Jae Engle
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Richa Saxena
- Center for Genomic Medicine and Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
- Program in Medical and Population Genetics, Broad Institute, Cambridge, MA 02142, USA
- Division of Sleep and Circadian Disorders, Department of Medicine, Brigham and Women’s Hospital and Division of Sleep Medicine, Harvard Medical School, Boston, MA 02115, USA
| | - David A. Prober
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| |
Collapse
|
37
|
Allen JR, Skeath JB, Johnson SL. Maintenance of Melanocyte Stem Cell Quiescence by GABA-A Signaling in Larval Zebrafish. Genetics 2019; 213:555-566. [PMID: 31444245 PMCID: PMC6781893 DOI: 10.1534/genetics.119.302416] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Accepted: 08/12/2019] [Indexed: 12/22/2022] Open
Abstract
In larval zebrafish, melanocyte stem cells (MSCs) are quiescent, but can be recruited to regenerate the larval pigment pattern following melanocyte ablation. Through pharmacological experiments, we found that inhibition of γ-aminobutyric acid (GABA)-A receptor function, specifically the GABA-A ρ subtype, induces excessive melanocyte production in larval zebrafish. Conversely, pharmacological activation of GABA-A inhibited melanocyte regeneration. We used clustered regularly interspaced short palindromic repeats/Cas9 to generate two mutant alleles of gabrr1, a subtype of GABA-A receptors. Both alleles exhibited robust melanocyte overproduction, while conditional overexpression of gabrr1 inhibited larval melanocyte regeneration. Our data suggest that gabrr1 signaling is necessary to maintain MSC quiescence and sufficient to reduce, but not eliminate, melanocyte regeneration in larval zebrafish.
Collapse
Affiliation(s)
- James R Allen
- Department of Genetics, Washington University School of Medicine, St. Louis, Missouri 63110
| | - James B Skeath
- Department of Genetics, Washington University School of Medicine, St. Louis, Missouri 63110
| | - Stephen L Johnson
- Department of Genetics, Washington University School of Medicine, St. Louis, Missouri 63110
| |
Collapse
|
38
|
Cook ZT, Brockway NL, Tobias ZJC, Pajarla J, Boardman IS, Ippolito H, Nkombo Nkoula S, Weissman TA. Combining near-infrared fluorescence with Brainbow to visualize expression of specific genes within a multicolor context. Mol Biol Cell 2019; 30:491-505. [PMID: 30586321 PMCID: PMC6594444 DOI: 10.1091/mbc.e18-06-0340] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Revised: 12/13/2018] [Accepted: 12/18/2018] [Indexed: 12/18/2022] Open
Abstract
Fluorescent proteins are a powerful experimental tool, allowing the visualization of gene expression and cellular behaviors in a variety of systems. Multicolor combinations of fluorescent proteins, such as Brainbow, have expanded the range of possible research questions and are useful for distinguishing and tracking cells. The addition of a separately driven color, however, would allow researchers to report expression of a manipulated gene within the multicolor context to investigate mechanistic effects. A far-red or near-infrared protein could be particularly suitable in this context, as these can be distinguished spectrally from Brainbow. We investigated five far-red/near-infrared proteins in zebrafish: TagRFP657, mCardinal, miRFP670, iRFP670, and mIFP. Our results show that both mCardinal and iRFP670 are useful fluorescent proteins for zebrafish expression. We also introduce a new transgenic zebrafish line that expresses Brainbow under the control of the neuroD promoter. We demonstrate that mCardinal can be used to track the expression of a manipulated bone morphogenetic protein receptor within the Brainbow context. The overlay of near-infrared fluorescence onto a Brainbow background defines a clear strategy for future research questions that aim to manipulate or track the effects of specific genes within a population of cells that are delineated using multicolor approaches.
Collapse
Affiliation(s)
- Zoe T. Cook
- Biology Department, Lewis and Clark College, Portland, OR 97219
| | | | | | - Joy Pajarla
- Biology Department, Lewis and Clark College, Portland, OR 97219
| | | | - Helen Ippolito
- Biology Department, Lewis and Clark College, Portland, OR 97219
| | | | | |
Collapse
|
39
|
Yamaguchi N, Colak-Champollion T, Knaut H. zGrad is a nanobody-based degron system that inactivates proteins in zebrafish. eLife 2019; 8:43125. [PMID: 30735119 PMCID: PMC6384026 DOI: 10.7554/elife.43125] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Accepted: 02/07/2019] [Indexed: 12/28/2022] Open
Abstract
The analysis of protein function is essential to modern biology. While protein function has mostly been studied through gene or RNA interference, more recent approaches to degrade proteins directly have been developed. Here, we adapted the anti-GFP nanobody-based system deGradFP from flies to zebrafish. We named this system zGrad and show that zGrad efficiently degrades transmembrane, cytosolic and nuclear GFP-tagged proteins in zebrafish in an inducible and reversible manner. Using tissue-specific and inducible promoters in combination with functional GFP-fusion proteins, we demonstrate that zGrad can inactivate transmembrane and cytosolic proteins globally, locally and temporally with different consequences. Global protein depletion results in phenotypes similar to loss of gene activity, while local and temporal protein inactivation yields more restricted and novel phenotypes. Thus, zGrad is a versatile tool to study the spatial and temporal requirement of proteins in zebrafish.
Collapse
Affiliation(s)
- Naoya Yamaguchi
- Skirball Institute of Biomolecular Medicine, New York University School of Medicine, New York, United States
| | - Tugba Colak-Champollion
- Skirball Institute of Biomolecular Medicine, New York University School of Medicine, New York, United States
| | - Holger Knaut
- Skirball Institute of Biomolecular Medicine, New York University School of Medicine, New York, United States
| |
Collapse
|
40
|
Podlasz P, Jakimiuk A, Kasica-Jarosz N, Czaja K, Wasowicz K. Neuroanatomical Localization of Galanin in Zebrafish Telencephalon and Anticonvulsant Effect of Galanin Overexpression. ACS Chem Neurosci 2018; 9:3049-3059. [PMID: 30095254 DOI: 10.1021/acschemneuro.8b00239] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Galanin is a neuropeptide widely expressed in the nervous system, but it is also present in non-neuronal locations. In the brain, galanin may function as an inhibitory neurotransmitter. Several studies have shown that galanin is involved in seizure regulation and can modulate epileptic activity in the brain. The overall goal of the study was to establish zebrafish as a model to study the antiepileptic effect of galanin. The goal of this study was achieved by (1) determining neuroanatomical localization of galanin in zebrafish lateral pallium, which is considered to be the zebrafish homologue of the mammalian hippocampus, the brain region essential for initiation of seizures, and (2) testing the anticonvulsant effect of galanin overexpression. Whole mount immunofluorescence staining and pentylenotetrazole (PTZ)-seizure model in larval zebrafish using automated analysis of motor function and qPCR were used in the study. Immunohistochemical staining of zebrafish larvae revealed numerous galanin-IR fibers innervating the subpallium, but only scarce fibers reaching the dorsal parts of telencephalon, including lateral pallium. In three-month old zebrafish, galanin-IR innervation of the telencephalon was similar; however, many more galanin-IR fibers reached the dorsal telencephalon, but in the lateral pallium only scarce galanin-IR fibers were visible. qRT-PCR revealed, as expected, a strong increase in the expression of galanin in the Tg(hsp70l:galn) line after heat shock; however, also without heat shock, the galanin expression was several-fold higher than in the control animals. Galanin overexpression resulted in downregulation of c-fos after PTZ treatment. Behavioral analysis showed that galanin overexpression inhibited locomotor activity in PTZ-treated and control larvae. The obtained results show that galanin overexpression reduced the incidence of seizure-like behavior episodes and their intensity but had no significant effect on their duration. The findings indicate that in addition to antiepileptic action, galanin modulates arousal behavior and demonstrates a sedative effect. The current study showed that galanin overexpression correlated with a potent anticonvulsant effect in the zebrafish PTZ-seizure model.
Collapse
Affiliation(s)
- Piotr Podlasz
- Department of Pathophysiology, Forensic Veterinary and Administration, Faculty of Veterinary Medicine, University of Warmia and Mazury in Olsztyn, Olsztyn, Poland
| | - Anna Jakimiuk
- Department of Pathophysiology, Forensic Veterinary and Administration, Faculty of Veterinary Medicine, University of Warmia and Mazury in Olsztyn, Olsztyn, Poland
| | - Natalia Kasica-Jarosz
- Department of Animal Anatomy, Faculty of Veterinary Medicine, University of Warmia and Mazury in Olsztyn, Olsztyn, Poland
| | - Krzysztof Czaja
- Department of Veterinary Biosciences and Diagnostic Imaging, University of Georgia, Athens, Georgia, United States
| | - Krzysztof Wasowicz
- Department of Pathophysiology, Forensic Veterinary and Administration, Faculty of Veterinary Medicine, University of Warmia and Mazury in Olsztyn, Olsztyn, Poland
| |
Collapse
|
41
|
Chai A, Mateus AM, Oozeer F, Sousa-Nunes R. Spatiotemporally controlled genetic perturbation for efficient large-scale studies of cell non-autonomous effects. eLife 2018; 7:e38393. [PMID: 30479273 PMCID: PMC6320068 DOI: 10.7554/elife.38393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Accepted: 11/26/2018] [Indexed: 11/13/2022] Open
Abstract
Studies in genetic model organisms have revealed much about the development and pathology of complex tissues. Most have focused on cell-intrinsic gene functions and mechanisms. Much less is known about how transformed, or otherwise functionally disrupted, cells interact with healthy ones toward a favorable or pathological outcome. This is largely due to technical limitations. We developed new genetic tools in Drosophila melanogaster that permit efficient multiplexed gain- and loss-of-function genetic perturbations with separable spatial and temporal control. Importantly, our novel tool-set is independent of the commonly used GAL4/UAS system, freeing the latter for additional, non-autonomous, genetic manipulations; and is built into a single strain, allowing one-generation interrogation of non-autonomous effects. Altogether, our design opens up efficient genome-wide screens on any deleterious phenotype, once plasmid or genome engineering is used to place the desired miRNA(s) or ORF(s) into our genotype. Specifically, we developed tools to study extrinsic effects on neural tumor growth but the strategy presented has endless applications within and beyond neurobiology, and in other model organisms.
Collapse
Affiliation(s)
- Andrea Chai
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and NeuroscienceKing’s College LondonLondonUnited Kingdom
| | - Ana M Mateus
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and NeuroscienceKing’s College LondonLondonUnited Kingdom
| | - Fazal Oozeer
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and NeuroscienceKing’s College LondonLondonUnited Kingdom
| | - Rita Sousa-Nunes
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and NeuroscienceKing’s College LondonLondonUnited Kingdom
| |
Collapse
|
42
|
Pierpont ME, Brueckner M, Chung WK, Garg V, Lacro RV, McGuire AL, Mital S, Priest JR, Pu WT, Roberts A, Ware SM, Gelb BD, Russell MW. Genetic Basis for Congenital Heart Disease: Revisited: A Scientific Statement From the American Heart Association. Circulation 2018; 138:e653-e711. [PMID: 30571578 PMCID: PMC6555769 DOI: 10.1161/cir.0000000000000606] [Citation(s) in RCA: 387] [Impact Index Per Article: 55.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
This review provides an updated summary of the state of our knowledge of the genetic contributions to the pathogenesis of congenital heart disease. Since 2007, when the initial American Heart Association scientific statement on the genetic basis of congenital heart disease was published, new genomic techniques have become widely available that have dramatically changed our understanding of the causes of congenital heart disease and, clinically, have allowed more accurate definition of the pathogeneses of congenital heart disease in patients of all ages and even prenatally. Information is presented on new molecular testing techniques and their application to congenital heart disease, both isolated and associated with other congenital anomalies or syndromes. Recent advances in the understanding of copy number variants, syndromes, RASopathies, and heterotaxy/ciliopathies are provided. Insights into new research with congenital heart disease models, including genetically manipulated animals such as mice, chicks, and zebrafish, as well as human induced pluripotent stem cell-based approaches are provided to allow an understanding of how future research breakthroughs for congenital heart disease are likely to happen. It is anticipated that this review will provide a large range of health care-related personnel, including pediatric cardiologists, pediatricians, adult cardiologists, thoracic surgeons, obstetricians, geneticists, genetic counselors, and other related clinicians, timely information on the genetic aspects of congenital heart disease. The objective is to provide a comprehensive basis for interdisciplinary care for those with congenital heart disease.
Collapse
|
43
|
Wang J, Yin Y, Lau S, Sankaran J, Rothenberg E, Wohland T, Meier-Schellersheim M, Knaut H. Anosmin1 Shuttles Fgf to Facilitate Its Diffusion, Increase Its Local Concentration, and Induce Sensory Organs. Dev Cell 2018; 46:751-766.e12. [PMID: 30122631 DOI: 10.1016/j.devcel.2018.07.015] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2018] [Revised: 05/26/2018] [Accepted: 07/18/2018] [Indexed: 02/08/2023]
Abstract
Growth factors induce and pattern sensory organs, but how their distribution is regulated by the extracellular matrix (ECM) is largely unclear. To address this question, we analyzed the diffusion behavior of Fgf10 molecules during sensory organ formation in the zebrafish posterior lateral line primordium. In this tissue, secreted Fgf10 induces organ formation at a distance from its source. We find that most Fgf10 molecules are highly diffusive and move rapidly through the ECM. We identify Anosmin1, which when mutated in humans causes Kallmann Syndrome, as an ECM protein that binds to Fgf10 and facilitates its diffusivity by increasing the pool of fast-moving Fgf10 molecules. In the absence of Anosmin1, Fgf10 levels are reduced and organ formation is impaired. Global overexpression of Anosmin1 slows the fast-moving Fgf10 molecules and results in Fgf10 dispersal. These results suggest that Anosmin1 liberates ECM-bound Fgf10 and shuttles it to increase its signaling range.
Collapse
Affiliation(s)
- John Wang
- Skirball Institute of Biomolecular Medicine, New York University School of Medicine, 540 First Avenue, New York, NY 10016, USA
| | - Yandong Yin
- Department of Biochemistry and Molecular Pharmacology, New York University School of Medicine, 540 First Avenue, New York, NY 10016, USA
| | - Stephanie Lau
- Skirball Institute of Biomolecular Medicine, New York University School of Medicine, 540 First Avenue, New York, NY 10016, USA
| | - Jagadish Sankaran
- Department of Chemistry, National University of Singapore, Singapore, Singapore
| | - Eli Rothenberg
- Department of Biochemistry and Molecular Pharmacology, New York University School of Medicine, 540 First Avenue, New York, NY 10016, USA
| | - Thorsten Wohland
- Department of Chemistry, National University of Singapore, Singapore, Singapore
| | - Martin Meier-Schellersheim
- Laboratory of Systems Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Holger Knaut
- Skirball Institute of Biomolecular Medicine, New York University School of Medicine, 540 First Avenue, New York, NY 10016, USA.
| |
Collapse
|
44
|
A Collection of Transgenic Medaka Strains for Efficient Site-Directed Transgenesis Mediated by phiC31 Integrase. G3-GENES GENOMES GENETICS 2018; 8:2585-2593. [PMID: 29848622 PMCID: PMC6071608 DOI: 10.1534/g3.118.200130] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Genetic analysis is facilitated by the efficient production of transgenic strains expressing a DNA of interest as a single copy at a designated chromosomal location. However, technical progress toward this goal in medaka fish (Oryzias latipes), a vertebrate model organism, has been slow. It is well known that phiC31 integrase enables efficient site-directed transgenesis by catalyzing the recombination of an attP DNA motif in a host genome with an attB motif in a targeting vector. This system was pioneered in medaka using the Sleeping Beauty transposon system, and the attP site was established at three chromosomal locations. However, this number appeared insufficient with regard to genetic linkage between the attP-landing site and a genetically modified locus of interest. Here, to establish a collection of transgenic strains of medaka, we introduced an attP motif into the medaka genome using the Ac/Ds maize transposon system and established 12 independent transgenic strains harboring a single copy of the attP motif in at least 11 of the 24 medaka chromosomes. We designed an attB-targeting vector that was integrated efficiently and precisely into the attP-landing site, and with which the DNA of interest was efficiently transmitted to germline cells. Extraneous sequences in the integrants derived from the bacterial backbone of the attB-targeting vector as well as a transgenic fluorescence marker present in the attP-landing site were removable through flippase-mediated recombination. Further, an advanced targeting vector with a heart-specific recombination marker served as a useful tool for easily screening phiC31 integrase-mediated recombinant G0 embryos, leading to the efficient establishment of transgenic strains. Thus, our resources advance genetic research in medaka.
Collapse
|
45
|
Linsley JW, Hsu IU, Wang W, Kuwada JY. Transport of the alpha subunit of the voltage gated L-type calcium channel through the sarcoplasmic reticulum occurs prior to localization to triads and requires the beta subunit but not Stac3 in skeletal muscles. Traffic 2018; 18:622-632. [PMID: 28697281 DOI: 10.1111/tra.12502] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Revised: 07/06/2017] [Accepted: 07/06/2017] [Indexed: 12/20/2022]
Abstract
Contraction of skeletal muscle is initiated by excitation-contraction (EC) coupling during which membrane voltage is transduced to intracellular Ca2+ release. EC coupling requires L-type voltage gated Ca2+ channels (the dihydropyridine receptor or DHPR) located at triads, which are junctions between the transverse (T) tubule and sarcoplasmic reticulum (SR) membranes, that sense membrane depolarization in the T tubule membrane. Reduced EC coupling is associated with ageing, and disruptions of EC coupling result in congenital myopathies for which there are few therapies. The precise localization of DHPRs to triads is critical for EC coupling, yet trafficking of the DHPR to triads is not well understood. Using dynamic imaging of zebrafish muscle fibers, we find that DHPR is transported along the longitudinal SR in a microtubule-independent mechanism. Furthermore, transport of DHPR in the SR membrane is differentially affected in null mutants of Stac3 or DHPRβ, two essential components of EC coupling. These findings reveal previously unappreciated features of DHPR motility within the SR prior to assembly at triads.
Collapse
Affiliation(s)
- Jeremy W Linsley
- Cell and Molecular Biology Program, University of Michigan, Ann Arbor, Michigan.,Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, Michigan
| | - I-Uen Hsu
- Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, Michigan
| | - Wenjia Wang
- Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, Michigan
| | - John Y Kuwada
- Cell and Molecular Biology Program, University of Michigan, Ann Arbor, Michigan.,Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, Michigan
| |
Collapse
|
46
|
Functional Analysis of the Promoter Region of Japanese Flounder ( Paralichthys olivaceus) β-actin Gene: A Useful Tool for Gene Research in Marine Fish. Int J Mol Sci 2018; 19:ijms19051401. [PMID: 29738459 PMCID: PMC5983668 DOI: 10.3390/ijms19051401] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Revised: 04/21/2018] [Accepted: 05/02/2018] [Indexed: 12/16/2022] Open
Abstract
A newly isolated Japanese flounder (Paralichthys olivaceus) β-actin promoter and its derivative compact construct Poβ-actinΔ−1080/−801Δ−500/−201 have recently been demonstrated to promote ectopic gene expression in cell lines. Different Poβ-actin promoter deletion mutants were constructed and functionally characterized. Mutational analyses by dual-luciferase detected that three regulatory elements, including one enhancer (−1399/−1081) and two silencers (−1080/−801, −500/−201) in the first intron. The sequence located at −1399/−1081 was determined to significantly affect promoter activity. Additionally, the first exon (−1489/−1400) could also remarkably promote the β-actin promoter activity. In the following transduction application, we removed the two silencers and generated a compact reconstruct promoter/enhancer (Poβ-actinΔ−1080/−801Δ−500/−201), which exhibited relatively stronger promoter activity compared with Poβ-actin. Furthermore, the green fluorescent protein (GFP) transgenic stable flounder cell line was obtained by the reconstructed Poβ-actinΔ−1080/−801Δ−500/−201 promoter. Our study provided the potential application of Japanese flounder β-actin, particularly Poβ-actinΔ−1080/−801Δ−500/−201, in ectopic gene expression in the future.
Collapse
|
47
|
González-Rosa JM, Sharpe M, Field D, Soonpaa MH, Field LJ, Burns CE, Burns CG. Myocardial Polyploidization Creates a Barrier to Heart Regeneration in Zebrafish. Dev Cell 2018; 44:433-446.e7. [PMID: 29486195 PMCID: PMC5830170 DOI: 10.1016/j.devcel.2018.01.021] [Citation(s) in RCA: 205] [Impact Index Per Article: 29.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Revised: 12/11/2017] [Accepted: 01/26/2018] [Indexed: 01/07/2023]
Abstract
Correlative evidence suggests that polyploidization of heart muscle, which occurs naturally in post-natal mammals, creates a barrier to heart regeneration. Here, we move beyond a correlation by demonstrating that experimental polyploidization of zebrafish cardiomyocytes is sufficient to suppress their proliferative potential during regeneration. Initially, we determined that zebrafish myocardium becomes susceptible to polyploidization upon transient cytokinesis inhibition mediated by dominant-negative Ect2. Using a transgenic strategy, we generated adult animals containing mosaic hearts composed of differentially labeled diploid and polyploid-enriched cardiomyocyte populations. Diploid cardiomyocytes outcompeted their polyploid neighbors in producing regenerated heart muscle. Moreover, hearts composed of equivalent proportions of diploid and polyploid cardiomyocytes failed to regenerate altogether, demonstrating that a critical percentage of diploid cardiomyocytes is required to achieve heart regeneration. Our data identify cardiomyocyte polyploidization as a barrier to heart regeneration and suggest that mobilizing rare diploid cardiomyocytes in the human heart will improve its regenerative capacity.
Collapse
Affiliation(s)
- Juan Manuel González-Rosa
- Cardiovascular Research Center, Massachusetts General Hospital, Charlestown, MA 02129, USA; Harvard Medical School, Boston, MA 02115, USA
| | - Michka Sharpe
- Cardiovascular Research Center, Massachusetts General Hospital, Charlestown, MA 02129, USA; Harvard Medical School, Boston, MA 02115, USA
| | - Dorothy Field
- The Krannert Institute of Cardiology, the Wells Center for Pediatric Research, and Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Mark H Soonpaa
- The Krannert Institute of Cardiology, the Wells Center for Pediatric Research, and Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Loren J Field
- The Krannert Institute of Cardiology, the Wells Center for Pediatric Research, and Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Caroline E Burns
- Cardiovascular Research Center, Massachusetts General Hospital, Charlestown, MA 02129, USA; Harvard Medical School, Boston, MA 02115, USA; Harvard Stem Cell Institute, Cambridge, MA 02138, USA.
| | - C Geoffrey Burns
- Cardiovascular Research Center, Massachusetts General Hospital, Charlestown, MA 02129, USA; Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
48
|
Sorrells S, Nik S, Casey MJ, Cameron RC, Truong H, Toruno C, Gulfo M, Lowe A, Jette C, Stewart RA, Bowman TV. Spliceosomal components protect embryonic neurons from R-loop-mediated DNA damage and apoptosis. Dis Model Mech 2018; 11:dmm.031583. [PMID: 29419415 PMCID: PMC5894942 DOI: 10.1242/dmm.031583] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Accepted: 01/18/2018] [Indexed: 02/02/2023] Open
Abstract
RNA splicing factors are essential for the viability of all eukaryotic cells; however, in metazoans some cell types are exquisitely sensitive to disruption of splicing factors. Neuronal cells represent one such cell type, and defects in RNA splicing factors can lead to neurodegenerative diseases. The basis for this tissue selectivity is not well understood owing to difficulties in analyzing the consequences of splicing factor defects in whole-animal systems. Here, we use zebrafish mutants to show that loss of spliceosomal components, including splicing factor 3b, subunit 1 (sf3b1), causes increased DNA double-strand breaks and apoptosis in embryonic neurons. Moreover, these mutants show a concomitant accumulation of R-loops, which are non-canonical nucleic acid structures that promote genomic instability. Dampening R-loop formation by conditional induction of ribonuclease H1 in sf3b1 mutants reduced neuronal DNA damage and apoptosis. These findings show that splicing factor dysfunction leads to R-loop accumulation and DNA damage that sensitizes embryonic neurons to apoptosis. Our results suggest that diseases associated with splicing factor mutations could be susceptible to treatments that modulate R-loop levels. Summary: Loss of RNA splicing factors causes R-loop accumulation and DNA damage in embryonic neurons, sensitizing them to radiation-induced cell death. These findings suggest that diseased cells with mutations in splicing factors are vulnerable to radiotherapy.
Collapse
Affiliation(s)
- Shelly Sorrells
- Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112, USA
| | - Sara Nik
- Department of Developmental & Molecular Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA.,Gottesman Institute for Stem Cell Biology and Regenerative Medicine, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Mattie J Casey
- Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112, USA
| | - Rosannah C Cameron
- Department of Developmental & Molecular Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA.,Gottesman Institute for Stem Cell Biology and Regenerative Medicine, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Harold Truong
- Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112, USA
| | - Cristhian Toruno
- Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112, USA
| | - Michelle Gulfo
- Gottesman Institute for Stem Cell Biology and Regenerative Medicine, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Albert Lowe
- Gottesman Institute for Stem Cell Biology and Regenerative Medicine, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Cicely Jette
- Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112, USA
| | - Rodney A Stewart
- Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112, USA
| | - Teresa V Bowman
- Department of Developmental & Molecular Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA .,Gottesman Institute for Stem Cell Biology and Regenerative Medicine, Albert Einstein College of Medicine, Bronx, NY 10461, USA.,Department of Medicine (Oncology), Albert Einstein College of Medicine, Bronx, NY 10461, USA
| |
Collapse
|
49
|
Jeffrey EJ, Crawford BD. The epitope-mediated MMP activation assay: detection and quantification of the activation of Mmp2 in vivo in the zebrafish embryo. Histochem Cell Biol 2018; 149:277-286. [PMID: 29350268 DOI: 10.1007/s00418-018-1634-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/11/2018] [Indexed: 12/18/2022]
Abstract
Matrix remodeling is a consequence of tightly regulated matrix metalloproteinase (MMP) activity. MMPs are synthesized as inactive precursors with auto-inhibitory N-terminal propeptides, the proteolytic removal of which exposes the catalytic zinc ion, rendering the protease active. The regulation of MMP activation has been investigated primarily in tissue culture and biochemical assays that lack important biological context. Here we present the epitope-mediated MMP activation (EMMA) assay and use it to observe the activation of Mmp2 (gelatinase A) by endogenous mechanisms in the intact zebrafish embryo. The hemagglutinin (HA) and GFP-tagged reporter construct becomes activated on the surface of specific cells and this activation is abolished by broad-spectrum inhibition of metalloproteinase activity, consistent with existing models of gelatinase A activation. The mechanism(s) acting on the construct are spatially restricted, metalloproteinase-dependent and replacing the HA tag with mCherry abolishes activation, showing that the mechanism(s) are sensitive to the structure of the N-terminal domain. The construct is activated strongly in maturing myotome boundaries, but also intracellularly within myofibrils, consistent with reports implicating this protease in muscle development and function. In addition to general-purpose tools for the production of "EMMAed" MMPs and other proteins, we have established a transgenic line of zebrafish expressing EMMAedMmp2 under control of an inducible promoter to facilitate further investigation into the regulation of this ubiquitous ECM-remodeling protease in vivo.
Collapse
Affiliation(s)
- Emma J Jeffrey
- Matrix Dynamics Lab, Biology Department, University of New Brunswick, 10 Bailey Drive, Fredericton, NB, E3B 5A3, Canada
| | - Bryan D Crawford
- Matrix Dynamics Lab, Biology Department, University of New Brunswick, 10 Bailey Drive, Fredericton, NB, E3B 5A3, Canada.
| |
Collapse
|
50
|
Lee DA, Andreev A, Truong TV, Chen A, Hill AJ, Oikonomou G, Pham U, Hong YK, Tran S, Glass L, Sapin V, Engle J, Fraser SE, Prober DA. Genetic and neuronal regulation of sleep by neuropeptide VF. eLife 2017; 6:25727. [PMID: 29106375 PMCID: PMC5705210 DOI: 10.7554/elife.25727] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Accepted: 11/03/2017] [Indexed: 12/25/2022] Open
Abstract
Sleep is an essential and phylogenetically conserved behavioral state, but it remains unclear to what extent genes identified in invertebrates also regulate vertebrate sleep. RFamide-related neuropeptides have been shown to promote invertebrate sleep, and here we report that the vertebrate hypothalamic RFamide neuropeptide VF (NPVF) regulates sleep in the zebrafish, a diurnal vertebrate. We found that NPVF signaling and npvf-expressing neurons are both necessary and sufficient to promote sleep, that mature peptides derived from the NPVF preproprotein promote sleep in a synergistic manner, and that stimulation of npvf-expressing neurons induces neuronal activity levels consistent with normal sleep. These results identify NPVF signaling and npvf-expressing neurons as a novel vertebrate sleep-promoting system and suggest that RFamide neuropeptides participate in an ancient and central aspect of sleep control.
Collapse
Affiliation(s)
- Daniel A Lee
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, United States
| | - Andrey Andreev
- Department of Bioengineering, University of Southern California, Los Angeles, United States
| | - Thai V Truong
- Translational Imaging Center, University of Southern California, Los Angeles, United States
| | - Audrey Chen
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, United States
| | - Andrew J Hill
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, United States
| | - Grigorios Oikonomou
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, United States
| | - Uyen Pham
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, United States
| | - Young K Hong
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, United States
| | - Steven Tran
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, United States
| | - Laura Glass
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, United States
| | - Viveca Sapin
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, United States
| | - Jae Engle
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, United States
| | - Scott E Fraser
- Department of Bioengineering, University of Southern California, Los Angeles, United States.,Translational Imaging Center, University of Southern California, Los Angeles, United States
| | - David A Prober
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, United States
| |
Collapse
|