1
|
Till K, Borchers A. The Rho GEF Trio functions in contact inhibition of locomotion of neural crest cells by interacting with Ptk7. Development 2025; 152:dev204446. [PMID: 40326503 DOI: 10.1242/dev.204446] [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: 10/11/2024] [Accepted: 04/07/2025] [Indexed: 05/07/2025]
Abstract
Neural crest (NC) cells are highly migratory cells that contribute to a wide range of vertebrate tissues and must respond to a variety of external signals to precisely control directed cell migration. The RhoGEF Trio is particularly well suited to relay signals to the cytoskeleton because it contains two GEF domains that activate Rac1 and RhoA, respectively. Previously, we have shown that Trio is dynamically localized in Xenopus NC cells and required for their migration. However, how its distinct enzymatic functions are spatially controlled remains unclear. Here, we show that Trio is required for contact inhibition of locomotion (CIL), a phenomenon whereby NC cells change their polarity and directionality upon cell-cell contact. At cell-cell contacts, Trio interacts with Ptk7, a regulator of planar cell polarity that we have recently shown to be required for CIL. Our data suggest that Ptk7 inhibits the Rac1 activity of Trio, thereby limiting Trio activity to the activation of RhoA and promoting CIL.
Collapse
Affiliation(s)
- Katharina Till
- Department of Biology, Molecular Embryology, Philipps-University Marburg, Karl-von-Frisch-Straße 8, 35043 Marburg, Germany
| | - Annette Borchers
- Department of Biology, Molecular Embryology, Philipps-University Marburg, Karl-von-Frisch-Straße 8, 35043 Marburg, Germany
| |
Collapse
|
2
|
Gritti N, Power RM, Graves A, Huisken J. Image restoration of degraded time-lapse microscopy data mediated by near-infrared imaging. Nat Methods 2024; 21:311-321. [PMID: 38177507 PMCID: PMC10864180 DOI: 10.1038/s41592-023-02127-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Accepted: 11/10/2023] [Indexed: 01/06/2024]
Abstract
Time-lapse fluorescence microscopy is key to unraveling biological development and function; however, living systems, by their nature, permit only limited interrogation and contain untapped information that can only be captured by more invasive methods. Deep-tissue live imaging presents a particular challenge owing to the spectral range of live-cell imaging probes/fluorescent proteins, which offer only modest optical penetration into scattering tissues. Herein, we employ convolutional neural networks to augment live-imaging data with deep-tissue images taken on fixed samples. We demonstrate that convolutional neural networks may be used to restore deep-tissue contrast in GFP-based time-lapse imaging using paired final-state datasets acquired using near-infrared dyes, an approach termed InfraRed-mediated Image Restoration (IR2). Notably, the networks are remarkably robust over a wide range of developmental times. We employ IR2 to enhance the information content of green fluorescent protein time-lapse images of zebrafish and Drosophila embryo/larval development and demonstrate its quantitative potential in increasing the fidelity of cell tracking/lineaging in developing pescoids. Thus, IR2 is poised to extend live imaging to depths otherwise inaccessible.
Collapse
Affiliation(s)
- Nicola Gritti
- Morgridge Institute for Research, Madison, WI, USA
- Mesoscopic Imaging Facility, European Molecular Biology Laboratory Barcelona, Barcelona, Spain
| | - Rory M Power
- Morgridge Institute for Research, Madison, WI, USA
- EMBL Imaging Center, European Molecular Biology Laboratory Heidelberg, Heidelberg, Germany
| | | | - Jan Huisken
- Morgridge Institute for Research, Madison, WI, USA.
- Department of Integrative Biology, University of Wisconsin Madison, Madison, WI, USA.
- Department of Biology and Psychology, Georg-August-University Göttingen, Göttingen, Germany.
- Cluster of Excellence 'Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells' (MBExC), University of Göttingen, Göttingen, Germany.
| |
Collapse
|
3
|
Frommelt J, Liu E, Bhaidani A, Hu B, Gao Y, Ye D, Lin F. Flat mount preparation for whole-mount fluorescent imaging of zebrafish embryos. Biol Open 2023; 12:bio060048. [PMID: 37746815 PMCID: PMC10373579 DOI: 10.1242/bio.060048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Accepted: 06/23/2023] [Indexed: 09/26/2023] Open
Abstract
The zebrafish is a widely used model organism for biomedical research due to its ease of maintenance, external fertilization of embryos, rapid embryonic development, and availability of established genetic tools. One notable advantage of using zebrafish is the transparency of the embryos, which enables high-resolution imaging of specific cells, tissues, and structures through the use of transgenic and knock-in lines. However, as the embryo develops, multiple layers of tissue wrap around the lipid-enriched yolk, which can create a challenge to image tissues located deep within the embryo. While various methods are available, such as two-photon imaging, cryosectioning, vibratome sectioning, and micro-surgery, each of these has limitations. In this study, we present a novel deyolking method that allows for high-quality imaging of tissues that are obscured by other tissues and the yolk. Embryos are lightly fixed in 1% PFA to remove the yolk without damaging embryonic tissues and are then refixed in 4% PFA and mounted on custom-made bridged slides. This method offers a simple way to prepare imaging samples that can be subjected to further preparation, such as immunostaining. Furthermore, the bridged slides described in this study can be used for imaging tissue and organ preparations from various model organisms.
Collapse
Affiliation(s)
- Joseph Frommelt
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, IA, 52242, USA
| | - Emily Liu
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, IA, 52242, USA
| | - Afraz Bhaidani
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, IA, 52242, USA
| | - Bo Hu
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, IA, 52242, USA
| | - Yuanyuan Gao
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, IA, 52242, USA
| | - Ding Ye
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, IA, 52242, USA
| | - Fang Lin
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, IA, 52242, USA
| |
Collapse
|
4
|
Herchenröther A, Gossen S, Friedrich T, Reim A, Daus N, Diegmüller F, Leers J, Sani HM, Gerstner S, Schwarz L, Stellmacher I, Szymkowiak LV, Nist A, Stiewe T, Borggrefe T, Mann M, Mackay JP, Bartkuhn M, Borchers A, Lan J, Hake SB. The H2A.Z and NuRD associated protein HMG20A controls early head and heart developmental transcription programs. Nat Commun 2023; 14:472. [PMID: 36709316 PMCID: PMC9884267 DOI: 10.1038/s41467-023-36114-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Accepted: 01/17/2023] [Indexed: 01/30/2023] Open
Abstract
Specialized chromatin-binding proteins are required for DNA-based processes during development. We recently established PWWP2A as a direct histone variant H2A.Z interactor involved in mitosis and craniofacial development. Here, we identify the H2A.Z/PWWP2A-associated protein HMG20A as part of several chromatin-modifying complexes, including NuRD, and show that it localizes to distinct genomic regulatory regions. Hmg20a depletion causes severe head and heart developmental defects in Xenopus laevis. Our data indicate that craniofacial malformations are caused by defects in neural crest cell (NCC) migration and cartilage formation. These developmental failures are phenocopied in Hmg20a-depleted mESCs, which show inefficient differentiation into NCCs and cardiomyocytes (CM). Consequently, loss of HMG20A, which marks open promoters and enhancers, results in chromatin accessibility changes and a striking deregulation of transcription programs involved in epithelial-mesenchymal transition (EMT) and differentiation processes. Collectively, our findings implicate HMG20A as part of the H2A.Z/PWWP2A/NuRD-axis and reveal it as a key modulator of intricate developmental transcription programs that guide the differentiation of NCCs and CMs.
Collapse
Affiliation(s)
| | - Stefanie Gossen
- Department of Biology, Molecular Embryology, Philipps University Marburg, Marburg, Germany
| | - Tobias Friedrich
- Institute for Biochemistry, Justus-Liebig University Giessen, Giessen, Germany.,Biomedical Informatics and Systems Medicine, Science Unit for Basic and Clinical Medicine, Institute for lung health, Justus-Liebig University Giessen, Giessen, Germany
| | - Alexander Reim
- Department of Proteomics and Signal Transduction, Max-Planck Institute of Biochemistry, Martinsried, Germany
| | - Nadine Daus
- Institute for Genetics, Justus-Liebig University Giessen, Giessen, Germany
| | - Felix Diegmüller
- Institute for Genetics, Justus-Liebig University Giessen, Giessen, Germany
| | - Jörg Leers
- Institute for Genetics, Justus-Liebig University Giessen, Giessen, Germany
| | - Hakimeh Moghaddas Sani
- School of Life and Environmental Sciences, University of Sydney, New South Wales, Australia
| | - Sarah Gerstner
- Department of Biology, Molecular Embryology, Philipps University Marburg, Marburg, Germany
| | - Leah Schwarz
- Department of Biology, Molecular Embryology, Philipps University Marburg, Marburg, Germany
| | - Inga Stellmacher
- Institute for Genetics, Justus-Liebig University Giessen, Giessen, Germany
| | - Laura Victoria Szymkowiak
- Institute for Genetics, Justus-Liebig University Giessen, Giessen, Germany.,Institute for Physiological Chemistry, Technical University Dresden, Dresden, Germany
| | - Andrea Nist
- Genomics Core Facility, Institute of Molecular Oncology, Universities of Giessen and Marburg Lung Center, Member of the German Center for Lung Research (DZL), Philipps-University Marburg, Marburg, Germany
| | - Thorsten Stiewe
- Genomics Core Facility, Institute of Molecular Oncology, Universities of Giessen and Marburg Lung Center, Member of the German Center for Lung Research (DZL), Philipps-University Marburg, Marburg, Germany
| | - Tilman Borggrefe
- Institute for Biochemistry, Justus-Liebig University Giessen, Giessen, Germany
| | - Matthias Mann
- Department of Proteomics and Signal Transduction, Max-Planck Institute of Biochemistry, Martinsried, Germany
| | - Joel P Mackay
- School of Life and Environmental Sciences, University of Sydney, New South Wales, Australia
| | - Marek Bartkuhn
- Biomedical Informatics and Systems Medicine, Science Unit for Basic and Clinical Medicine, Institute for lung health, Justus-Liebig University Giessen, Giessen, Germany.
| | - Annette Borchers
- Department of Biology, Molecular Embryology, Philipps University Marburg, Marburg, Germany.
| | - Jie Lan
- Institute for Genetics, Justus-Liebig University Giessen, Giessen, Germany.
| | - Sandra B Hake
- Institute for Genetics, Justus-Liebig University Giessen, Giessen, Germany.
| |
Collapse
|
5
|
Zulueta-Coarasa T, Fadul J, Ahmed M, Rosenblatt J. Physical confinement promotes mesenchymal trans-differentiation of invading transformed cells in vivo. iScience 2022; 25:105330. [PMID: 36325066 PMCID: PMC9618776 DOI: 10.1016/j.isci.2022.105330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 09/29/2022] [Accepted: 10/10/2022] [Indexed: 11/19/2022] Open
Abstract
Metastasis is tightly linked with poor cancer prognosis, yet it is not clear how transformed cells become invasive carcinomas. We previously discovered that single KRasV12-transformed cells can invade directly from the epithelium by basal cell extrusion. During this process, cells de-differentiate by mechanically pinching off their epithelial determinants, but how they trans-differentiate into a migratory, mesenchymal phenotype is not known. Here, we demonstrate that basally extruded KRasV12-expressing cells become significantly deformed as they invade the zebrafish body. Decreasing the confinement that cells experience after they invade reduces the percentage of KRasV12 cells that trans-differentiate into mesenchymal cell types, while higher confinement increases this percentage. Additionally, increased confinement promotes accumulation of internal masses over time. Altogether, our results suggest that mechanical forces drive not only de-differentiation of KRasV12-transformed epithelial cells as they invade but also their re-differentiation into mesenchymal phenotypes that contribute to distant metastases.
Collapse
Affiliation(s)
- Teresa Zulueta-Coarasa
- The Randall Centre for Cell and Molecular Biophysics, School of Basic and Medical Biosciences, Faculty of Life Sciences and Medicine, School of Cancer and Pharmaceutical Sciences, King’s College London, London, SE1 1UL, UK
| | - John Fadul
- The Randall Centre for Cell and Molecular Biophysics, School of Basic and Medical Biosciences, Faculty of Life Sciences and Medicine, School of Cancer and Pharmaceutical Sciences, King’s College London, London, SE1 1UL, UK
| | - Marjana Ahmed
- The Randall Centre for Cell and Molecular Biophysics, School of Basic and Medical Biosciences, Faculty of Life Sciences and Medicine, School of Cancer and Pharmaceutical Sciences, King’s College London, London, SE1 1UL, UK
| | - Jody Rosenblatt
- The Randall Centre for Cell and Molecular Biophysics, School of Basic and Medical Biosciences, Faculty of Life Sciences and Medicine, School of Cancer and Pharmaceutical Sciences, King’s College London, London, SE1 1UL, UK
| |
Collapse
|
6
|
Moore RE, Pop S, Alleyne C, Clarke JDW. Microtubules are not required to generate a nascent axon in embryonic spinal neurons
in vivo. EMBO Rep 2022; 23:e52493. [DOI: 10.15252/embr.202152493] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Revised: 09/05/2022] [Accepted: 09/08/2022] [Indexed: 11/06/2022] Open
Affiliation(s)
- Rachel E Moore
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience King's College London London UK
| | - Sînziana Pop
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience King's College London London UK
- The Francis Crick Institute London UK
| | - Caché Alleyne
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience King's College London London UK
| | - Jonathan D W Clarke
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience King's College London London UK
| |
Collapse
|
7
|
Fulton T, Verd B, Steventon B. The unappreciated generative role of cell movements in pattern formation. ROYAL SOCIETY OPEN SCIENCE 2022; 9:211293. [PMID: 35601454 PMCID: PMC9043703 DOI: 10.1098/rsos.211293] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Accepted: 04/05/2022] [Indexed: 06/15/2023]
Abstract
The mechanisms underpinning the formation of patterned cellular landscapes has been the subject of extensive study as a fundamental problem of developmental biology. In most cases, attention has been given to situations in which cell movements are negligible, allowing researchers to focus on the cell-extrinsic signalling mechanisms, and intrinsic gene regulatory interactions that lead to pattern emergence at the tissue level. However, in many scenarios during development, cells rapidly change their neighbour relationships in order to drive tissue morphogenesis, while also undergoing patterning. To draw attention to the ubiquity of this problem and propose methodologies that will accommodate morphogenesis into the study of pattern formation, we review the current approaches to studying pattern formation in both static and motile cellular environments. We then consider how the cell movements themselves may contribute to the generation of pattern, rather than hinder it, with both a species specific and evolutionary viewpoint.
Collapse
Affiliation(s)
- Timothy Fulton
- Department of Genetics, University of Cambridge, Cambridge, UK
- School of Biological and Behavioural Sciences, Queen Mary University of London, London, UK
| | - Berta Verd
- Department of Genetics, University of Cambridge, Cambridge, UK
- Department of Zoology, University of Oxford, Oxford, UK
| | | |
Collapse
|
8
|
Crombez S, Leclerc P, Ray C, Ducros N. Computational hyperspectral light-sheet microscopy. OPTICS EXPRESS 2022; 30:4856-4866. [PMID: 35209458 DOI: 10.1364/oe.442043] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Accepted: 12/20/2021] [Indexed: 06/14/2023]
Abstract
We describe a computational light-sheet microscope designed for hyperspectral acquisition at high spectral resolution. The fluorescence light emitted from the full field-of-view is focused along the entrance slit of an imaging spectrometer using a cylindrical lens. To acquire the spatial dimension orthogonal to the slit of the spectrometer, we propose to illuminate the specimen with a sequence of structured light patterns and to solve the image reconstruction problem. Beam shaping is obtained simply using a digital micromirror device in conjunction with a traditional selective plane illumination microscopy setup. We demonstrate the feasibility of this method and report the first results in vivo in hydra specimens labeled using two fluorophores.
Collapse
|
9
|
Grund A, Till K, Giehl K, Borchers A. Ptk7 Is Dynamically Localized at Neural Crest Cell-Cell Contact Sites and Functions in Contact Inhibition of Locomotion. Int J Mol Sci 2021; 22:ijms22179324. [PMID: 34502237 PMCID: PMC8431534 DOI: 10.3390/ijms22179324] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 08/24/2021] [Accepted: 08/25/2021] [Indexed: 02/08/2023] Open
Abstract
Neural crest (NC) cells are highly migratory cells that contribute to various vertebrate tissues, and whose migratory behaviors resemble cancer cell migration and invasion. Information exchange via dynamic NC cell-cell contact is one mechanism by which the directionality of migrating NC cells is controlled. One transmembrane protein that is most likely involved in this process is protein tyrosine kinase 7 (PTK7), an evolutionary conserved Wnt co-receptor that is expressed in cranial NC cells and several tumor cells. In Xenopus, Ptk7 is required for NC migration. In this study, we show that the Ptk7 protein is dynamically localized at cell-cell contact zones of migrating Xenopus NC cells and required for contact inhibition of locomotion (CIL). Using deletion constructs of Ptk7, we determined that the extracellular immunoglobulin domains of Ptk7 are important for its transient accumulation and that they mediate homophilic binding. Conversely, we found that ectopic expression of Ptk7 in non-NC cells was able to prevent NC cell invasion. However, deletion of the extracellular domains of Ptk7 abolished this effect. Thus, Ptk7 is sufficient at protecting non-NC tissue from NC cell invasion, suggesting a common role of PTK7 in contact inhibition, cell invasion, and tissue integrity.
Collapse
Affiliation(s)
- Anita Grund
- Faculty of Biology, Molecular Embryology, Philipps-University Marburg, D-35032 Marburg, Germany; (A.G.); (K.T.)
| | - Katharina Till
- Faculty of Biology, Molecular Embryology, Philipps-University Marburg, D-35032 Marburg, Germany; (A.G.); (K.T.)
| | - Klaudia Giehl
- Faculty of Medicine, Signal Transduction of Cellular Motility, Internal Medicine V, Justus-Liebig University Giessen, D-35392 Giessen, Germany;
| | - Annette Borchers
- Faculty of Biology, Molecular Embryology, Philipps-University Marburg, D-35032 Marburg, Germany; (A.G.); (K.T.)
- Correspondence: ; Tel.: +49-6421-2826587
| |
Collapse
|
10
|
Iyer S, Mukherjee S, Kumar M. Watching the embryo: Evolution of the microscope for the study of embryogenesis. Bioessays 2021; 43:e2000238. [PMID: 33837551 DOI: 10.1002/bies.202000238] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 03/09/2021] [Accepted: 03/10/2021] [Indexed: 11/08/2022]
Abstract
Embryos and microscopes share a long, remarkable history and biologists have always been intrigued to watch how embryos develop under the microscope. Here we discuss the advances in microscopy which have greatly influenced our current understanding of embryogenesis. We highlight the evolution of microscopes and the optical technologies that have been instrumental in studying various developmental processes. These imaging modalities provide mechanistic insights into the dynamic cellular and molecular events which drive lineage commitment and morphogenetic changes in the developing embryo. We begin the journey with a brief history of microscopy to study embryos. First, we review the principles and optics of light, fluorescence, confocal, and electron microscopy which have been key techniques for imaging cellular and molecular events during embryonic development. Next, we discuss recent key imaging modalities such as light-sheet microscopy, which are suitable for whole embryo imaging. Further, we highlight imaging techniques like multiphoton and super resolution microscopy for beyond light diffraction limit, high resolution imaging. Lastly, we review some of the scattering-based imaging methods and techniques used for imaging human embryos.
Collapse
Affiliation(s)
- Sharada Iyer
- Academy of Scientific and Innovative Research (AcCSIR), CSIR-CCMB campus, Uppal road, Hyderabad, 500007, India.,CSIR - Centre for Cellular and Molecular Biology, Hyderabad, India
| | | | - Megha Kumar
- CSIR - Centre for Cellular and Molecular Biology, Hyderabad, India
| |
Collapse
|
11
|
Mariani RA, Paranjpe S, Dobrowolski R, Weber GF. 14-3-3 targets keratin intermediate filaments to mechanically sensitive cell-cell contacts. Mol Biol Cell 2020; 31:930-943. [PMID: 32074004 PMCID: PMC7185971 DOI: 10.1091/mbc.e18-06-0373] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Intermediate filament (IF) cytoskeletal networks simultaneously support mechanical integrity and influence signal transduction pathways. Marked remodeling of the keratin IF network accompanies collective cellular morphogenetic movements that occur during early embryonic development in the frog Xenopus laevis. While this reorganization of keratin is initiated by force transduction on cell–cell contacts mediated by C-cadherin, the mechanism by which keratin filament reorganization occurs remains poorly understood. In this work, we demonstrate that 14-3-3 proteins regulate keratin reorganization dynamics in embryonic mesendoderm cells from Xenopus gastrula. 14-3-3 colocalizes with keratin filaments near cell–cell junctions in migrating mesendoderm. Coimmunoprecipitation, mass spectrometry, and bioinformatic analyses indicate 14-3-3 is associated with Keratin 19 (K19) in the whole embryo and, more specifically, mesendoderm tissue. Inhibition of 14-3-3 results in both the decreased exchange of keratin subunits into filaments and blocks keratin filament recruitment toward cell–cell contacts. Synthetically coupling 14-3-3 to K19 through a unique fusion construct conversely induces the localization of this keratin population to the region of cell–cell contacts. Taken together, these findings indicate that 14-3-3 acts on keratin IFs and is involved in their reorganization to sites of cell adhesion.
Collapse
Affiliation(s)
- Richard A Mariani
- Department of Biological Sciences, Rutgers University-Newark, Newark, NJ 07102
| | - Shalaka Paranjpe
- Department of Biological Sciences, Rutgers University-Newark, Newark, NJ 07102
| | - Radek Dobrowolski
- Department of Biological Sciences, Rutgers University-Newark, Newark, NJ 07102.,Department of Biology, University of Indianapolis, Indianapolis, IN 46227
| | - Gregory F Weber
- Department of Biological Sciences, Rutgers University-Newark, Newark, NJ 07102.,Glenn Biggs Institute for Alzheimer's & Neurodegenerative Diseases, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229
| |
Collapse
|
12
|
Affiliation(s)
- John B Wallingford
- Department of Molecular Biosciences, University of Texas at Austin, Austin, TX, USA.
| |
Collapse
|
13
|
Hadjivasiliou Z, Moore RE, McIntosh R, Galea GL, Clarke JDW, Alexandre P. Basal Protrusions Mediate Spatiotemporal Patterns of Spinal Neuron Differentiation. Dev Cell 2020; 49:907-919.e10. [PMID: 31211994 PMCID: PMC6584357 DOI: 10.1016/j.devcel.2019.05.035] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Revised: 02/26/2019] [Accepted: 05/20/2019] [Indexed: 12/22/2022]
Abstract
During early spinal cord development, neurons of particular subtypes differentiate with a sparse periodic pattern while later neurons differentiate in the intervening space to eventually produce continuous columns of similar neurons. The mechanisms that regulate this spatiotemporal pattern are unknown. In vivo imaging in zebrafish reveals that differentiating spinal neurons transiently extend two long protrusions along the basal surface of the spinal cord before axon initiation. These protrusions express Delta protein, consistent with the hypothesis they influence Notch signaling at a distance of several cell diameters. Experimental reduction of Laminin expression leads to smaller protrusions and shorter distances between differentiating neurons. The experimental data and a theoretical model support the proposal that neuronal differentiation pattern is regulated by transient basal protrusions that deliver temporally controlled lateral inhibition mediated at a distance. This work uncovers a stereotyped protrusive activity of newborn neurons that organize long-distance spatiotemporal patterning of differentiation.
Collapse
Affiliation(s)
- Zena Hadjivasiliou
- Department of Biochemistry, Science II, University of Geneva, Geneva, Switzerland; Centre for Mathematics and Physics in the Life Sciences and Experimental Biology, University College London, Gower Street, London WC1N 1EH, UK
| | - Rachel E Moore
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology, and Neuroscience, King's College London, London SE1 1UL, UK
| | - Rebecca McIntosh
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology, and Neuroscience, King's College London, London SE1 1UL, UK; Developmental Biology and Cancer, UCL GOS Institute of Child Health, London WC1N 1EH, UK
| | - Gabriel L Galea
- Developmental Biology and Cancer, UCL GOS Institute of Child Health, London WC1N 1EH, UK
| | - Jonathan D W Clarke
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology, and Neuroscience, King's College London, London SE1 1UL, UK.
| | - Paula Alexandre
- Developmental Biology and Cancer, UCL GOS Institute of Child Health, London WC1N 1EH, UK.
| |
Collapse
|
14
|
Kleinhans DS, Lecaudey V. Standardized mounting method of (zebrafish) embryos using a 3D-printed stamp for high-content, semi-automated confocal imaging. BMC Biotechnol 2019; 19:68. [PMID: 31640669 PMCID: PMC6805687 DOI: 10.1186/s12896-019-0558-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Accepted: 09/04/2019] [Indexed: 01/24/2023] Open
Abstract
Background Developmental biology relies to a large extent on the observation and comparison of phenotypic traits through time using high resolution microscopes. In this context, transparent model organisms such as the zebrafish Danio rerio in which developing tissues and organs can be easily observed and imaged using fluorescent proteins have become very popular. One limiting factor however is the acquisition of a sufficient amount of data, in standardized and reproducible conditions, to allow robust quantitative analysis. One way to improve this is by developing mounting methods to increase the number of embryos that can be imaged simultaneously in near-to-identical orientation. Results Here we present an improved mounting method allowing semi-automated and high-content imaging of zebrafish embryos. It is based on a 3D-printed stamp which is used to create a 2D coordinate system of multiple μ-wells in an agarose cast. Each μ-well models a negative of the average zebrafish embryo morphology between 22 and 96 h-post-fertilization. Due to this standardized and reproducible arrangement, it is possible to define a custom well plate in the respective imaging software that allows for a semi-automated imaging process. Furthermore, the improvement in Z-orientation significantly reduces post-processing and improves comparability of volumetric data while reducing light exposure and thus photo-bleaching and photo-toxicity, and improving signal-to-noise ratio (SNR). Conclusions We present here a new method that allows to standardize and improve mounting and imaging of embryos. The 3D-printed stamp creates a 2D coordinate system of μ-wells in an agarose cast thus standardizing specimen mounting and allowing high-content imaging of up to 44 live or mounted zebrafish embryos simultaneously in a semi-automated, well-plate like manner on inverted confocal microscopes. In summary, image data quality and acquisition efficiency (amount of data per time) are significantly improved. The latter might also be crucial when using the services of a microscopy facility.
Collapse
Affiliation(s)
- David Simon Kleinhans
- Department of Developmental Biology of Vertebrates, Institute for Cell biology and Neuroscience, Goethe University, Max-von-Laue-Str. 13, 60438, Frankfurt am Main, Germany
| | - Virginie Lecaudey
- Department of Developmental Biology of Vertebrates, Institute for Cell biology and Neuroscience, Goethe University, Max-von-Laue-Str. 13, 60438, Frankfurt am Main, Germany.
| |
Collapse
|
15
|
Rgma-Induced Neo1 Proteolysis Promotes Neural Tube Morphogenesis. J Neurosci 2019; 39:7465-7484. [PMID: 31399534 DOI: 10.1523/jneurosci.3262-18.2019] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2018] [Revised: 07/01/2019] [Accepted: 07/31/2019] [Indexed: 01/02/2023] Open
Abstract
Neuroepithelial cell (NEC) elongation is one of several key cell behaviors that mediate the tissue-level morphogenetic movements that shape the neural tube (NT), the precursor of the brain and spinal cord. However, the upstream signals that promote NEC elongation have been difficult to tease apart from those regulating apico-basal polarity and hingepoint formation, due to their confounding interdependence. The Repulsive Guidance Molecule a (Rgma)/Neogenin 1 (Neo1) signaling pathway plays a conserved role in NT formation (neurulation) and is reported to regulate both NEC elongation and apico-basal polarity, through signal transduction events that have not been identified. We examine here the role of Rgma/Neo1 signaling in zebrafish (sex unknown), an organism that does not use hingepoints to shape its hindbrain, thereby enabling a direct assessment of the role of this pathway in NEC elongation. We confirm that Rgma/Neo1 signaling is required for microtubule-mediated NEC elongation, and demonstrate via cell transplantation that Neo1 functions cell autonomously to promote elongation. However, in contrast to previous findings, our data do not support a role for this pathway in establishing apical junctional complexes. Last, we provide evidence that Rgma promotes Neo1 glycosylation and intramembrane proteolysis, resulting in the production of a transient, nuclear intracellular fragment (NeoICD). Partial rescue of Neo1a and Rgma knockdown embryos by overexpressing neoICD suggests that this proteolytic cleavage is essential for neurulation. Based on these observations, we propose that RGMA-induced NEO1 proteolysis orchestrates NT morphogenesis by promoting NEC elongation independently of the establishment of apical junctional complexes.SIGNIFICANCE STATEMENT The neural tube, the CNS precursor, is shaped during neurulation. Neural tube defects occur frequently, yet underlying genetic risk factors are poorly understood. Neuroepithelial cell (NEC) elongation is essential for proper completion of neurulation. Thus, connecting NEC elongation with the molecular pathways that control this process is expected to reveal novel neural tube defect risk factors and increase our understanding of NT development. Effectors of cell elongation include microtubules and microtubule-associated proteins; however, upstream regulators remain controversial due to the confounding interdependence of cell elongation and establishment of apico-basal polarity. Here, we reveal that Rgma-Neo1 signaling controls NEC elongation independently of the establishment of apical junctional complexes and identify Rgma-induced Neo1 proteolytic cleavage as a key upstream signaling event.
Collapse
|
16
|
Abstract
Mouse genetic approaches when combined with live imaging tools are revolutionizing our current understanding of mammalian developmental biology. The availability and improvement of a wide variety of genetically encoded fluorescent proteins have provided indispensable tools to visualize cells and subcellular features in living organisms. It is now possible to generate genetically modified mouse lines expressing several spectrally distinct fluorescent proteins in a tissue-specific or -inducible manner. Such reporter-expressing lines make it possible to image dynamic cellular behaviors in the context of living embryos undergoing normal or aberrant development. As with all viviparous mammals, mouse embryos develop within the uterus, and so live imaging experiments require culture conditions that closely mimic the in vivo environment. Over the past decades, significant advances have been made in developing conditions for culturing both pre- and postimplantation-stage mouse embryos. In this chapter, we discuss routine methods for ex utero culture of preimplantation- and postimplantation-stage mouse embryos. In particular, we describe protocols for collecting mouse embryos of various stages, setting up culture conditions for their ex utero culture and imaging, and using laser scanning confocal microscopy to visualize live processes in mouse embryos expressing fluorescent reporters.
Collapse
Affiliation(s)
- Sonja Nowotschin
- Developmental Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Vidur Garg
- Developmental Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Anna Piliszek
- Department of Experimental Embryology, Institute of Genetics and Animal Breeding, Polish Academy of Sciences, Jastrzębiec, Poland
| | - Anna-Katerina Hadjantonakis
- Developmental Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| |
Collapse
|
17
|
Prakash S, de Boer BA, Hagoort J, Gunst QD, Ruijter JM, van den Hoff MJB. Considerations for Measurement of Embryonic Organ Growth. Anat Rec (Hoboken) 2018; 302:49-57. [PMID: 30289204 PMCID: PMC7028151 DOI: 10.1002/ar.23908] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2017] [Revised: 11/08/2017] [Accepted: 11/27/2017] [Indexed: 11/11/2022]
Abstract
Organogenesis is a complex coordinated process of cell proliferation, growth, migration, and apoptosis. Differential growth rates, particularly during cardiogenesis, play a role in establishing morphology. Studies using stereological and cell sorting methods derive averages of morphogenetic parameters for an organ. To understand tissue composition and differential growth, the researcher must determine a number of morphogenetic parameters in the developing organ. Such measurements require sectioning to enable identification of organ borders, tissue components and cell types, three-dimensional (3D)-reconstruction of sections to visualize morphology and a 3D-measurement scheme to build local morphogenetic information. Although thick the section confocal microscopy partially solves these issues, information loss at the section surface hampers the reconstruction of 3D morphology. Episcopic imaging provides the correct morphology but lacks histological procedures to identify multiple cell types. The 3D-measurement scheme is based on systematic sampling, with overlapping sample volumes, of the entire organ in the aligned image stack. For each sample volume, morphogenetic variables are calculated and results projected back to the cube (boxel) at the sample volume center. Boxel size determines spatial resolution of the final quantitative 3D-reconstruction whereas size of the sample volume determines the precision of the morphogenetic information. The methods described here can be used to measure tissue volume, proliferation and cell size, to determine contribution and distribution of cell types in a tissue and to display this information in a quantitative 3D-reconstruction. Anat Rec, 302:49-57, 2019. © 2018 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Stuti Prakash
- Department of Medical Biology, Amsterdam University Medical Centers, Academic Medical Center, Amsterdam, The Netherlands
| | - Bouke A de Boer
- Department of Medical Biology, Amsterdam University Medical Centers, Academic Medical Center, Amsterdam, The Netherlands
| | - Jaco Hagoort
- Department of Medical Biology, Amsterdam University Medical Centers, Academic Medical Center, Amsterdam, The Netherlands
| | - Quinn D Gunst
- Department of Medical Biology, Amsterdam University Medical Centers, Academic Medical Center, Amsterdam, The Netherlands
| | - Jan M Ruijter
- Department of Medical Biology, Amsterdam University Medical Centers, Academic Medical Center, Amsterdam, The Netherlands
| | - Maurice J B van den Hoff
- Department of Medical Biology, Amsterdam University Medical Centers, Academic Medical Center, Amsterdam, The Netherlands
| |
Collapse
|
18
|
Shindo A, Audrey A, Takagishi M, Takahashi M, Wallingford JB, Kinoshita M. Septin-dependent remodeling of cortical microtubule drives cell reshaping during epithelial wound healing. J Cell Sci 2018; 131:jcs212647. [PMID: 29777035 PMCID: PMC6031381 DOI: 10.1242/jcs.212647] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Accepted: 05/10/2018] [Indexed: 12/31/2022] Open
Abstract
Wounds in embryos heal rapidly through contraction of the wound edges. Despite well-recognized significance of the actomyosin purse string for wound closure, roles for other cytoskeletal components are largely unknown. Here, we report that the septin cytoskeleton cooperates with actomyosin and microtubules to coordinate circumferential contraction of the wound margin and concentric elongation of wound-proximal cells in Xenopus laevis embryos. Microtubules reoriented radially, forming bundles along lateral cell cortices in elongating wound-proximal cells. Depletion of septin 7 (Sept7) slowed wound closure by attenuating the wound edge contraction and cell elongation. ROCK/Rho-kinase inhibitor-mediated suppression of actomyosin contractility enhanced the Sept7 phenotype, whereas the Sept7 depletion did not affect the accumulation of actomyosin at the wound edge. The cortical microtubule bundles were reduced in wound-proximal cells in Sept7 knockdown (Sept7-KD) embryos, but forced bundling of microtubules mediated by the microtubule-stabilizing protein Map7 did not rescue the Sept7-KD phenotype. Nocodazole-mediated microtubule depolymerization enhanced the Sept7-KD phenotype, suggesting that Sept7 is required for microtubule reorganization during cell elongation. Our findings indicate that septins are required for the rapid wound closure by facilitating cortical microtubule reorganization and the concentric elongation of surrounding cells.
Collapse
Affiliation(s)
- Asako Shindo
- Division of Biological Sciences, Department of Molecular Biology, Nagoya University Graduate School of Science, Nagoya 464-8602, Japan
- Department of Molecular Biosciences, University of Texas at Austin, Austin 78712, USA
| | - Anastasia Audrey
- Division of Biological Sciences, Department of Molecular Biology, Nagoya University Graduate School of Science, Nagoya 464-8602, Japan
| | - Maki Takagishi
- Department of Tumor Pathology, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Masahide Takahashi
- Department of Tumor Pathology, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - John B Wallingford
- Department of Molecular Biosciences, University of Texas at Austin, Austin 78712, USA
| | - Makoto Kinoshita
- Division of Biological Sciences, Department of Molecular Biology, Nagoya University Graduate School of Science, Nagoya 464-8602, Japan
| |
Collapse
|
19
|
Torres Crigna A, Daniele C, Gamez C, Medina Balbuena S, Pastene DO, Nardozi D, Brenna C, Yard B, Gretz N, Bieback K. Stem/Stromal Cells for Treatment of Kidney Injuries With Focus on Preclinical Models. Front Med (Lausanne) 2018; 5:179. [PMID: 29963554 PMCID: PMC6013716 DOI: 10.3389/fmed.2018.00179] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Accepted: 05/24/2018] [Indexed: 12/18/2022] Open
Abstract
Within the last years, the use of stem cells (embryonic, induced pluripotent stem cells, or hematopoietic stem cells), Progenitor cells (e.g., endothelial progenitor cells), and most intensely mesenchymal stromal cells (MSC) has emerged as a promising cell-based therapy for several diseases including nephropathy. For patients with end-stage renal disease (ESRD), dialysis or finally organ transplantation are the only therapeutic modalities available. Since ESRD is associated with a high healthcare expenditure, MSC therapy represents an innovative approach. In a variety of preclinical and clinical studies, MSC have shown to exert renoprotective properties, mediated mainly by paracrine effects, immunomodulation, regulation of inflammation, secretion of several trophic factors, and possibly differentiation to renal precursors. However, studies are highly diverse; thus, knowledge is still limited regarding the exact mode of action, source of MSC in comparison to other stem cell types, administration route and dose, tracking of cells and documentation of therapeutic efficacy by new imaging techniques and tissue visualization. The aim of this review is to provide a summary of published studies of stem cell therapy in acute and chronic kidney injury, diabetic nephropathy, polycystic kidney disease, and kidney transplantation. Preclinical studies with allogeneic or xenogeneic cell therapy were first addressed, followed by a summary of clinical trials carried out with autologous or allogeneic hMSC. Studies were analyzed with respect to source of cell type, mechanism of action etc.
Collapse
Affiliation(s)
- Adriana Torres Crigna
- Medical Faculty Mannheim, Institute of Transfusion Medicine and Immunology, University of Heidelberg, German Red Cross Blood Service Baden-Württemberg-Hessen, Mannheim, Germany
| | - Cristina Daniele
- Medical Faculty Mannheim, Medical Research Centre, University of Heidelberg, Mannheim, Germany
| | - Carolina Gamez
- Department for Experimental Orthopaedics and Trauma Surgery, Medical Faculty Mannheim, Orthopaedic and Trauma Surgery Centre (OUZ), Heidelberg University, Mannheim, Germany
| | - Sara Medina Balbuena
- Department of Medicine (Nephrology/Endrocrinology/Rheumathology), University Medical Centre Mannheim, University of Heidelberg, Mannheim, Germany
| | - Diego O. Pastene
- Department of Medicine (Nephrology/Endrocrinology/Rheumathology), University Medical Centre Mannheim, University of Heidelberg, Mannheim, Germany
| | - Daniela Nardozi
- Medical Faculty Mannheim, Medical Research Centre, University of Heidelberg, Mannheim, Germany
| | - Cinzia Brenna
- Medical Faculty Mannheim, Medical Research Centre, University of Heidelberg, Mannheim, Germany
| | - Benito Yard
- Department of Medicine (Nephrology/Endrocrinology/Rheumathology), University Medical Centre Mannheim, University of Heidelberg, Mannheim, Germany
| | - Norbert Gretz
- Medical Faculty Mannheim, Medical Research Centre, University of Heidelberg, Mannheim, Germany
| | - Karen Bieback
- Medical Faculty Mannheim, Institute of Transfusion Medicine and Immunology, University of Heidelberg, German Red Cross Blood Service Baden-Württemberg-Hessen, Mannheim, Germany
| |
Collapse
|
20
|
Åkerfelt M, Toriseva M, Nees M. Quantitative Phenotypic Image Analysis of Three-Dimensional Organotypic Cultures. Methods Mol Biol 2018. [PMID: 28634961 DOI: 10.1007/978-1-4939-7021-6_31] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Glandular epithelial cells differentiate into three-dimensional (3D) multicellular or acinar structures, particularly when embedded in laminin-rich extracellular matrix (ECM). The spectrum of different multicellular morphologies formed in 3D is a reliable indicator for the differentiation potential of normal, non-transformed cells compared to different stages of malignant progression. Motile cancer cells may actively invade the matrix, utilizing epithelial, mesenchymal, or mixed modes of motility. Dynamic phenotypic changes involved in 3D tumor cell invasion are also very sensitive to small-molecule inhibitors that, e.g., target the actin cytoskeleton. Our strategy is to recapitulate the formation and the histology of complex solid cancer tissues in vitro, based on cell culture technologies that promote the intrinsic differentiation potential of normal and transformed epithelial cells, and also including stromal fibroblasts and other key components of the tumor microenvironment. We have developed a streamlined stand-alone software solution that supports the detailed quantitative phenotypic analysis of organotypic 3D cultures. This approach utilizes the power of automated image analysis as a phenotypic readout in cell-based assays. AMIDA (Automated Morphometric Image Data Analysis) allows quantitative measurements of a large number of multicellular structures, which can form a multitude of different organoid shapes, sizes, and textures according to their capacity to engage in epithelial differentiation programs or not. At the far end of this spectrum of tumor-relevant differentiation properties, there are highly invasive tumor cells or multicellular structures that may rapidly invade the surrounding ECM, but fail to form higher-order epithelial tissue structures. Furthermore, this system allows us to monitor dynamic changes that can result from the extraordinary plasticity of tumor cells, e.g., epithelial-to-mesenchymal transition in live cell settings. Furthermore, AMIDA supports an automated workflow, and can be combined with quality control and statistical tools for data interpretation and visualization. Our approach supports the growing needs for user-friendly, straightforward solutions that facilitate cell-based organotypic 3D assays in basic research, drug discovery, and target validation.
Collapse
Affiliation(s)
- Malin Åkerfelt
- High-Content Screening Laboratory (HCSLab), Institute of Biomedicine, University of Turku, Itäinen Pitkäkatu 4B, 20520, Turku, Finland
| | - Mervi Toriseva
- High-Content Screening Laboratory (HCSLab), Institute of Biomedicine, University of Turku, Itäinen Pitkäkatu 4B, 20520, Turku, Finland
| | - Matthias Nees
- High-Content Screening Laboratory (HCSLab), Institute of Biomedicine, University of Turku, Itäinen Pitkäkatu 4B, 20520, Turku, Finland.
| |
Collapse
|
21
|
Wellmann J. Animating embryos: the in toto representation of life. BRITISH JOURNAL FOR THE HISTORY OF SCIENCE 2017; 50:521-535. [PMID: 28923124 DOI: 10.1017/s0007087417000656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
With the recent advent of systems biology, developmental biology is taking a new turn. Attempts to create a 'digital embryo' are prominent among systems approaches. At the heart of these systems-based endeavours, variously described as 'in vivo imaging', 'live imaging' or 'in toto representation', are visualization techniques that allow researchers to image whole, live embryos at cellular resolution over time. Ultimately, the aim of the visualizations is to build a computer model of embryogenesis. This article examines the role of such visualization techniques in the building of a computational model, focusing, in particular, on the cinematographic character of these representations. It asks how the animated representation of development may change the biological understanding of embryogenesis. By situating the animations of the digital embryo within the iconography of developmental biology, it brings to light the inextricably entwined, yet shifting, borders between the animated, the living and the computational.
Collapse
Affiliation(s)
- Janina Wellmann
- *Institute for Advanced Study on Media Cultures of Computer Simulation,Leuphana University Lüneburg,Wallstr.3,21335 Lüneburg,Germany.
| |
Collapse
|
22
|
Posfai E, Petropoulos S, de Barros FRO, Schell JP, Jurisica I, Sandberg R, Lanner F, Rossant J. Position- and Hippo signaling-dependent plasticity during lineage segregation in the early mouse embryo. eLife 2017; 6:22906. [PMID: 28226240 PMCID: PMC5370188 DOI: 10.7554/elife.22906] [Citation(s) in RCA: 116] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Accepted: 02/13/2017] [Indexed: 12/16/2022] Open
Abstract
The segregation of the trophectoderm (TE) from the inner cell mass (ICM) in the mouse blastocyst is determined by position-dependent Hippo signaling. However, the window of responsiveness to Hippo signaling, the exact timing of lineage commitment and the overall relationship between cell commitment and global gene expression changes are still unclear. Single-cell RNA sequencing during lineage segregation revealed that the TE transcriptional profile stabilizes earlier than the ICM and prior to blastocyst formation. Using quantitative Cdx2-eGFP expression as a readout of Hippo signaling activity, we assessed the experimental potential of individual blastomeres based on their level of Cdx2-eGFP expression and correlated potential with gene expression dynamics. We find that TE specification and commitment coincide and occur at the time of transcriptional stabilization, whereas ICM cells still retain the ability to regenerate TE up to the early blastocyst stage. Plasticity of both lineages is coincident with their window of sensitivity to Hippo signaling. DOI:http://dx.doi.org/10.7554/eLife.22906.001 In female mammals, conception is a complex process that involves several stages. First, an egg is released from the ovary and travels along a tube called the oviduct, where sperm from a male may fertilize it. If the egg is fertilized, the newly formed embryo moves into the womb, where it will then implant into the walls. In mice, it takes around four days for the embryo to implant and during this time, the cells in the embryo divide several times and start to specialize to form distinct cell types called lineages. The first two lineages to form are known as the inner cell mass and the trophectoderm. The inner cell mass forms a ball of cells within the embryo and contains the precursors of all cells that build the animal’s body. The trophectoderm forms a layer that surrounds the inner cell mass and will become part of the placenta (the organ that supplies the embryo with nutrients while it is in the womb). The embryo can organize these lineages without any instructions from the mother. However, it is still not clear when the cells start to differ from each other, and when they ‘commit’ to stay in these lineages. Cells in the inner cell mass and trophectoderm have different gene expression profiles, meaning that many genes display different levels of activity in these two lineages. Posfai et al. use a technique called single-cell RNA sequencing to analyse gene activity as the inner cell mass and trophectoderm form in mouse embryos. By measuring changes in gene activity, it is possible to track their development and show which genes change expression levels when each lineage specifies and commits. The experiments reveal that the inner cell mass and trophectoderm lineages develop at different times. As the inner cell mass forms, cells adopt the inner cell mass ‘identity’ before they commit to remaining in this lineage, revealing a window of time where different signals could still change the fate of the cells. However, when the early trophectoderm cells show the first signs of specialization, they also commit to their new identity at the same time. These findings suggest that the different timings at which these cell lineages form might provide embryos with the means to organize their own cells. An important future challenge is to understand exactly how the cells commit to their fate. DOI:http://dx.doi.org/10.7554/eLife.22906.002
Collapse
Affiliation(s)
- Eszter Posfai
- Program in Developmental and Stem Cell Biology, Hospital for Sick Children, Toronto, Canada
| | - Sophie Petropoulos
- Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden.,Ludwig Institute for Cancer Research, Karolinska Institutet, Stockholm, Sweden.,Division of Obstetrics and Gynecology, Karolinska Universitetssjukhuset, Stockholm, Sweden
| | | | - John Paul Schell
- Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden.,Division of Obstetrics and Gynecology, Karolinska Universitetssjukhuset, Stockholm, Sweden
| | - Igor Jurisica
- Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, Canada.,Departments of Medical Biophysics and Computer Science, University of Toronto, Toronto, Canada.,Institute of Neuroimmunology, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Rickard Sandberg
- Ludwig Institute for Cancer Research, Karolinska Institutet, Stockholm, Sweden.,Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
| | - Fredrik Lanner
- Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden.,Division of Obstetrics and Gynecology, Karolinska Universitetssjukhuset, Stockholm, Sweden
| | - Janet Rossant
- Program in Developmental and Stem Cell Biology, Hospital for Sick Children, Toronto, Canada.,Department of Molecular Genetics, University of Toronto, Toronto, Canada
| |
Collapse
|
23
|
All-in-one 3D printed microscopy chamber for multidimensional imaging, the UniverSlide. Sci Rep 2017; 7:42378. [PMID: 28186188 PMCID: PMC5301227 DOI: 10.1038/srep42378] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2016] [Accepted: 12/29/2016] [Indexed: 12/11/2022] Open
Abstract
While live 3D high resolution microscopy techniques are developing rapidly, their use for biological applications is partially hampered by practical difficulties such as the lack of a versatile sample chamber. Here, we propose the design of a multi-usage observation chamber adapted for live 3D bio-imaging. We show the usefulness and practicality of this chamber, which we named the UniverSlide, for live imaging of two case examples, namely multicellular systems encapsulated in sub-millimeter hydrogel shells and zebrafish larvae. We also demonstrate its versatility and compatibility with all microscopy devices by using upright or inverted microscope configurations after loading the UniverSlide with fixed or living samples. Further, the device is applicable for medium/high throughput screening and automatized multi-position image acquisition, providing a constraint-free but stable and parallelized immobilization of the samples. The frame of the UniverSlide is fabricated using a stereolithography 3D printer, has the size of a microscopy slide, is autoclavable and sealed with a removable lid, which makes it suitable for use in a controlled culture environment. We describe in details how to build this chamber and we provide all the files necessary to print the different pieces in the lab.
Collapse
|
24
|
Abstract
Skeletal muscle performs an essential function in human physiology with defects in genes encoding a variety of cellular components resulting in various types of inherited muscle disorders. Muscular dystrophies (MDs) are a severe and heterogeneous type of human muscle disease, manifested by progressive muscle wasting and degeneration. The disease pathogenesis and therapeutic options for MDs have been investigated for decades using rodent models, and considerable knowledge has been accumulated on the cause and pathogenetic mechanisms of this group of human disorders. However, due to some differences between disease severity and progression, what is learned in mammalian models does not always transfer to humans, prompting the desire for additional and alternative models. More recently, zebrafish have emerged as a novel and robust animal model for the study of human muscle disease. Zebrafish MD models possess a number of distinct advantages for modeling human muscle disorders, including the availability and ease of generating mutations in homologous disease-causing genes, the ability to image living muscle tissue in an intact animal, and the suitability of zebrafish larvae for large-scale chemical screens. In this chapter, we review the current understanding of molecular and cellular mechanisms involved in MDs, the process of myogenesis in zebrafish, and the structural and functional characteristics of zebrafish larval muscles. We further discuss the insights gained from the key zebrafish MD models that have been so far generated, and we summarize the attempts that have been made to screen for small molecules inhibitors of the dystrophic phenotypes using these models. Overall, these studies demonstrate that zebrafish is a useful in vivo system for modeling aspects of human skeletal muscle disorders. Studies using these models have contributed both to the understanding of the pathogenesis of muscle wasting disorders and demonstrated their utility as highly relevant models to implement therapeutic screening regimens.
Collapse
Affiliation(s)
- M Li
- Monash University, Clayton, VIC, Australia
| | - K J Hromowyk
- The Ohio State University, Columbus, OH, United States
| | - S L Amacher
- The Ohio State University, Columbus, OH, United States
| | - P D Currie
- Monash University, Clayton, VIC, Australia
| |
Collapse
|
25
|
Xiong F, Megason SG. Abstracting the principles of development using imaging and modeling. Integr Biol (Camb) 2016; 7:633-42. [PMID: 25946995 DOI: 10.1039/c5ib00025d] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Here we look at modern developmental biology with a focus on the relationship between different approaches of investigation. We argue that direct imaging is a powerful approach not only for obtaining descriptive information but also for model generation and testing that lead to mechanistic insights. Modeling, on the other hand, conceptualizes imaging data and provides guidance to perturbations. The inquiry progresses most efficiently when a trinity of approaches—quantitative imaging (measurement), modeling (theory) and perturbation (test)—are pursued in concert, but not when one approach is dominant. Using recent studies of the zebrafish system, we show how this combination has effectively advanced classic topics in developmental biology compared to a perturbation-centric approach. Finally, we show that interdisciplinary expertise and perhaps specialization are necessary for carrying out a systematic approach, and discuss the technical hurdles.
Collapse
Affiliation(s)
- Fengzhu Xiong
- Department of Systems Biology, Harvard Medical School, Boston, MA, USA.
| | | |
Collapse
|
26
|
Jayachandran P, Olmo VN, Sanchez SP, McFarland RJ, Vital E, Werner JM, Hong E, Sanchez-Alberola N, Molodstov A, Brewster RM. Microtubule-associated protein 1b is required for shaping the neural tube. Neural Dev 2016; 11:1. [PMID: 26782621 PMCID: PMC4717579 DOI: 10.1186/s13064-015-0056-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2015] [Accepted: 12/29/2015] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Shaping of the neural tube, the precursor of the brain and spinal cord, involves narrowing and elongation of the neural tissue, concomitantly with other morphogenetic changes that contribue to this process. In zebrafish, medial displacement of neural cells (neural convergence or NC), which drives the infolding and narrowing of the neural ectoderm, is mediated by polarized migration and cell elongation towards the dorsal midline. Failure to undergo proper NC results in severe neural tube defects, yet the molecular underpinnings of this process remain poorly understood. RESULTS We investigated here the role of the microtubule (MT) cytoskeleton in mediating NC in zebrafish embryos using the MT destabilizing and hyperstabilizing drugs nocodazole and paclitaxel respectively. We found that MTs undergo major changes in organization and stability during neurulation and are required for the timely completion of NC by promoting cell elongation and polarity. We next examined the role of Microtubule-associated protein 1B (Map1b), previously shown to promote MT dynamicity in axons. map1b is expressed earlier than previously reported, in the developing neural tube and underlying mesoderm. Loss of Map1b function using morpholinos (MOs) or δMap1b (encoding a truncated Map1b protein product) resulted in delayed NC and duplication of the neural tube, a defect associated with impaired NC. We observed a loss of stable MTs in these embryos that is likely to contribute to the NC defect. Lastly, we found that Map1b mediates cell elongation in a cell autonomous manner and polarized protrusive activity, two cell behaviors that underlie NC and are MT-dependent. CONCLUSIONS Together, these data highlight the importance of MTs in the early morphogenetic movements that shape the neural tube and reveal a novel role for the MT regulator Map1b in mediating cell elongation and polarized cell movement in neural progenitor cells.
Collapse
Affiliation(s)
- Pradeepa Jayachandran
- Department of Biological Sciences, University of Maryland Baltimore County, Baltimore, MD, USA.
| | - Valerie N Olmo
- Department of Biological Sciences, University of Maryland Baltimore County, Baltimore, MD, USA.
| | - Stephanie P Sanchez
- Department of Biological Sciences, University of Maryland Baltimore County, Baltimore, MD, USA.
| | - Rebecca J McFarland
- Department of Biological Sciences, University of Maryland Baltimore County, Baltimore, MD, USA.
| | - Eudorah Vital
- Department of Biological Sciences, University of Maryland Baltimore County, Baltimore, MD, USA.
| | - Jonathan M Werner
- Department of Biological Sciences, University of Maryland Baltimore County, Baltimore, MD, USA.
| | - Elim Hong
- Department of Biological Sciences, University of Maryland Baltimore County, Baltimore, MD, USA. .,Institut de Biologie Paris Seine-Laboratoire Neuroscience Paris Seine INSERM UMRS 1130, CNRS UMR 8246, UPMC UM 118 Université Pierre et Marie Curie, Paris, France.
| | - Neus Sanchez-Alberola
- Department of Biological Sciences, University of Maryland Baltimore County, Baltimore, MD, USA.
| | - Aleksey Molodstov
- Department of Biological Sciences, University of Maryland Baltimore County, Baltimore, MD, USA.
| | - Rachel M Brewster
- Department of Biological Sciences, University of Maryland Baltimore County, Baltimore, MD, USA.
| |
Collapse
|
27
|
Abstract
Zebrafish cancer models have greatly advanced our understanding of malignancy in humans. This is made possible due to the unique advantages of the zebrafish model including ex vivo development and large clutch sizes, which enable large-scale genetic and chemical screens. Transparency of the embryo and the creation of adult zebrafish devoid of pigmentation (casper) have permitted unprecedented ability to dynamically visualize cancer progression in live animals. When coupled with fluorescent reporters and transgenic approaches that drive oncogenesis, it is now possible to label entire or subpopulations of cancer cells and follow cancer growth in near real-time. Here, we will highlight aspects of in vivo imaging using the zebrafish and how it has enhanced our understanding of the fundamental aspects of tumor initiation, self-renewal, neovascularization, tumor cell heterogeneity, invasion and metastasis. Importantly, we will highlight the contribution of cancer imaging in zebrafish for drug discovery.
Collapse
|
28
|
Abstract
Lineage tracing is a widely used method for understanding cellular dynamics in multicellular organisms during processes such as development, adult tissue maintenance, injury repair and tumorigenesis. Advances in tracing or tracking methods, from light microscopy-based live cell tracking to fluorescent label-tracing with two-photon microscopy, together with emerging tissue clearing strategies and intravital imaging approaches have enabled scientists to decipher adult stem and progenitor cell properties in various tissues and in a wide variety of biological processes. Although technical advances have enabled time-controlled genetic labeling and simultaneous live imaging, a number of obstacles still need to be overcome. In this review, we aim to provide an in-depth description of the traditional use of lineage tracing as well as current strategies and upcoming new methods of labeling and imaging.
Collapse
Affiliation(s)
| | | | - Bon-Kyoung Koo
- Department of Genetics and Wellcome Trust - Medical Research Council Stem Cell Institute, University of Cambridge, Cambridge, CB2 1QR, United Kingdom
| |
Collapse
|
29
|
Tyrosine glycosylation of Rho by Yersinia toxin impairs blastomere cell behaviour in zebrafish embryos. Nat Commun 2015; 6:7807. [PMID: 26190758 PMCID: PMC4518317 DOI: 10.1038/ncomms8807] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2014] [Accepted: 06/15/2015] [Indexed: 12/20/2022] Open
Abstract
Yersinia species cause zoonotic infections, including enterocolitis and plague. Here we studied Yersinia ruckeri antifeeding prophage 18 (Afp18), the toxin component of the phage tail-derived protein translocation system Afp, which causes enteric redmouth disease in salmonid fish species. Here we show that microinjection of the glycosyltransferase domain Afp18G into zebrafish embryos blocks cytokinesis, actin-dependent motility and cell blebbing, eventually abrogating gastrulation. In zebrafish ZF4 cells, Afp18G depolymerizes actin stress fibres by mono-O-GlcNAcylation of RhoA at tyrosine-34; thereby Afp18G inhibits RhoA activation by guanine nucleotide exchange factors, and blocks RhoA, but not Rac and Cdc42 downstream signalling. The crystal structure of tyrosine-GlcNAcylated RhoA reveals an open conformation of the effector loop distinct from recently described structures of GDP- or GTP-bound RhoA. Unravelling of the molecular mechanism of the toxin component Afp18 as glycosyltransferase opens new perspectives in studies of phage tail-derived protein translocation systems, which are preserved from archaea to human pathogenic prokaryotes. Yersinia ruckeri is the source of redmouth disease in fish. Here the authors analysed the Yersinia toxin Afp18 and show that it acts to inhibit RhoA activation by glycosylating a distinct tyrosine residue inducing a signalling incompetent structural conformation.
Collapse
|
30
|
Li Y, Trivedi V, Truong TV, Koos DS, Lansford R, Chuong CM, Warburton D, Moats RA, Fraser SE. Dynamic imaging of the growth plate cartilage reveals multiple contributors to skeletal morphogenesis. Nat Commun 2015; 6:6798. [PMID: 25865282 PMCID: PMC4403347 DOI: 10.1038/ncomms7798] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2014] [Accepted: 02/27/2015] [Indexed: 11/09/2022] Open
Abstract
The diverse morphology of vertebrate skeletal system is genetically controlled, yet the means by which cells shape the skeleton remains to be fully illuminated. Here we perform quantitative analyses of cell behaviours in the growth plate cartilage, the template for long bone formation, to gain insights into this process. Using a robust avian embryonic organ culture, we employ time-lapse two-photon laser scanning microscopy to observe proliferative cells' behaviours during cartilage growth, resulting in cellular trajectories with a spreading displacement mainly along the tissue elongation axis. We build a novel software toolkit of quantitative methods to segregate the contributions of various cellular processes to the cellular trajectories. We find that convergent-extension, mitotic cell division, and daughter cell rearrangement do not contribute significantly to the observed growth process; instead, extracellular matrix deposition and cell volume enlargement are the key contributors to embryonic cartilage elongation.
Collapse
Affiliation(s)
- Yuwei Li
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, California 91125, USA.,Department of Molecular and Computational Biology, University of Southern California, Los Angeles, California, USA.,Developmental Biology and Regenerative Medicine Program, Saban Research Institute, Children's Hospital Los Angeles, Los Angeles, California 90027, USA
| | - Vikas Trivedi
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, California 91125, USA.,Department of Molecular and Computational Biology, University of Southern California, Los Angeles, California, USA
| | - Thai V Truong
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, California 91125, USA.,Department of Molecular and Computational Biology, University of Southern California, Los Angeles, California, USA
| | - David S Koos
- Department of Radiology, The Saban Research Institute, Children's Hospital Los Angeles, Los Angeles, California 90027, USA
| | - Rusty Lansford
- Department of Radiology, The Saban Research Institute, Children's Hospital Los Angeles, Los Angeles, California 90027, USA
| | - Cheng-Ming Chuong
- Department of Pathology, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - David Warburton
- Developmental Biology and Regenerative Medicine Program, Saban Research Institute, Children's Hospital Los Angeles, Los Angeles, California 90027, USA
| | - Rex A Moats
- Department of Radiology, The Saban Research Institute, Children's Hospital Los Angeles, Los Angeles, California 90027, USA
| | - Scott E Fraser
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, California 91125, USA.,Department of Molecular and Computational Biology, University of Southern California, Los Angeles, California, USA.,Developmental Biology and Regenerative Medicine Program, Saban Research Institute, Children's Hospital Los Angeles, Los Angeles, California 90027, USA.,Department of Biomedical Engineering, Viterbi School of Engineering, University of Southern California, Los Angeles, California, USA
| |
Collapse
|
31
|
Xiong F, Ma W, Hiscock TW, Mosaliganti KR, Tentner AR, Brakke KA, Rannou N, Gelas A, Souhait L, Swinburne IA, Obholzer ND, Megason SG. Interplay of cell shape and division orientation promotes robust morphogenesis of developing epithelia. Cell 2015; 159:415-27. [PMID: 25303534 DOI: 10.1016/j.cell.2014.09.007] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2014] [Revised: 05/29/2014] [Accepted: 09/03/2014] [Indexed: 11/17/2022]
Abstract
Epithelial cells acquire functionally important shapes (e.g., squamous, cuboidal, columnar) during development. Here, we combine theory, quantitative imaging, and perturbations to analyze how tissue geometry, cell divisions, and mechanics interact to shape the presumptive enveloping layer (pre-EVL) on the zebrafish embryonic surface. We find that, under geometrical constraints, pre-EVL flattening is regulated by surface cell number changes following differentially oriented cell divisions. The division pattern is, in turn, determined by the cell shape distribution, which forms under geometrical constraints by cell-cell mechanical coupling. An integrated mathematical model of this shape-division feedback loop recapitulates empirical observations. Surprisingly, the model predicts that cell shape is robust to changes of tissue surface area, cell volume, and cell number, which we confirm in vivo. Further simulations and perturbations suggest the parameter linking cell shape and division orientation contributes to epithelial diversity. Together, our work identifies an evolvable design logic that enables robust cell-level regulation of tissue-level development.
Collapse
Affiliation(s)
- Fengzhu Xiong
- Department of Systems Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Wenzhe Ma
- Department of Systems Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Tom W Hiscock
- Department of Systems Biology, Harvard Medical School, Boston, MA 02115, USA
| | | | - Andrea R Tentner
- Department of Systems Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Kenneth A Brakke
- Mathematics Department, Susquehanna University, Selinsgrove, PA 17870, USA
| | - Nicolas Rannou
- Department of Systems Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Arnaud Gelas
- Department of Systems Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Lydie Souhait
- Department of Systems Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Ian A Swinburne
- Department of Systems Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Nikolaus D Obholzer
- Department of Systems Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Sean G Megason
- Department of Systems Biology, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
32
|
Carlson M, Reeves W, Veeman M. Stochasticity and stereotypy in the Ciona notochord. Dev Biol 2014; 397:248-56. [PMID: 25459659 DOI: 10.1016/j.ydbio.2014.11.016] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2014] [Revised: 10/17/2014] [Accepted: 11/13/2014] [Indexed: 12/21/2022]
Abstract
Fate mapping with single cell resolution has typically been confined to embryos with completely stereotyped development. The lineages giving rise to the 40 cells of the Ciona notochord are invariant, but the intercalation of those cells into a single-file column is not. Here we use genetic labeling methods to fate map the Ciona notochord with both high resolution and large sample sizes. We find that the ordering of notochord cells into a single column is not random, but instead shows a distinctive signature characteristic of mediolaterally-biased intercalation. We find that patterns of cell intercalation in the notochord are somewhat stochastic but far more stereotyped than previously believed. Cell behaviors vary by lineage, with the secondary notochord lineage being much more constrained than the primary lineage. Within the primary lineage, patterns of intercalation reflect the geometry of the intercalating tissue. We identify the latest point at which notochord morphogenesis is largely stereotyped, which is shortly before the onset of mediolateral intercalation and immediately after the final cell divisions in the primary lineage. These divisions are consistently oriented along the AP axis. Our results indicate that the interplay between stereotyped and stochastic cell behaviors in morphogenesis can only be assessed by fate mapping experiments that have both cellular resolution and large sample sizes.
Collapse
Affiliation(s)
- Maia Carlson
- Division of Biology, Kansas State University, Manhattan, KS 66506, United States
| | - Wendy Reeves
- Division of Biology, Kansas State University, Manhattan, KS 66506, United States
| | - Michael Veeman
- Division of Biology, Kansas State University, Manhattan, KS 66506, United States.
| |
Collapse
|
33
|
Auer TO, Duroure K, Concordet JP, Del Bene F. CRISPR/Cas9-mediated conversion of eGFP- into Gal4-transgenic lines in zebrafish. Nat Protoc 2014; 9:2823-40. [DOI: 10.1038/nprot.2014.187] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
34
|
ptk7 mutant zebrafish models of congenital and idiopathic scoliosis implicate dysregulated Wnt signalling in disease. Nat Commun 2014; 5:4777. [PMID: 25182715 PMCID: PMC4155517 DOI: 10.1038/ncomms5777] [Citation(s) in RCA: 106] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2013] [Accepted: 07/23/2014] [Indexed: 01/09/2023] Open
Abstract
Scoliosis is a complex genetic disorder of the musculoskeletal system, characterized by three-dimensional rotation of the spine. Curvatures caused by malformed vertebrae (congenital scoliosis (CS)) are apparent at birth. Spinal curvatures with no underlying vertebral abnormality (idiopathic scoliosis (IS)) most commonly manifest during adolescence. The genetic and biological mechanisms responsible for IS remain poorly understood due largely to limited experimental models. Here we describe zygotic ptk7 (Zptk7) mutant zebrafish, deficient in a critical regulator of Wnt signalling, as the first genetically defined developmental model of IS. We identify a novel sequence variant within a single IS patient that disrupts PTK7 function, consistent with a role for dysregulated Wnt activity in disease pathogenesis. Furthermore, we demonstrate that embryonic loss-of-gene function in maternal-zygotic ptk7 mutants (MZptk7) leads to vertebral anomalies associated with CS. Our data suggest novel molecular origins of, and genetic links between, congenital and idiopathic forms of disease. Scoliosis is a complex genetic disorder characterized by spinal curvature. Here, the authors present experimental zebrafish models of idiopathic and congenital scoliosis and suggest a role for dysregulated Wnt activity in scoliosis aetiology.
Collapse
|
35
|
Anani S, Bhat S, Honma-Yamanaka N, Krawchuk D, Yamanaka Y. Initiation of Hippo signaling is linked to polarity rather than to cell position in the pre-implantation mouse embryo. Development 2014; 141:2813-24. [PMID: 24948601 DOI: 10.1242/dev.107276] [Citation(s) in RCA: 148] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
In the mouse embryo, asymmetric divisions during the 8-16 cell division generate two cell types, polar and apolar cells, that are allocated to outer and inner positions, respectively. This outer/inner configuration is the first sign of the formation of the first two cell lineages: trophectoderm (TE) and inner cell mass (ICM). Outer polar cells become TE and give rise to the placenta, whereas inner apolar cells become ICM and give rise to the embryo proper and yolk sac. Here, we analyze the frequency of asymmetric divisions during the 8-16 cell division and assess the relationships between cell polarity, cell and nuclear position, and Hippo signaling activation, the pathway that initiates lineage-specific gene expression in 16-cell embryos. Although the frequency of asymmetric divisions varied in each embryo, we found that more than six blastomeres divided asymmetrically in most embryos. Interestingly, many apolar cells in 16-cell embryos were located at outer positions, whereas only one or two apolar cells were located at inner positions. Live imaging analysis showed that outer apolar cells were eventually internalized by surrounding polar cells. Using isolated 8-cell blastomeres, we carefully analyzed the internalization process of apolar cells and found indications of higher cortical tension in apolar cells than in polar cells. Last, we found that apolar cells activate Hippo signaling prior to taking inner positions. Our results suggest that polar and apolar cells have intrinsic differences that establish outer/inner configuration and differentially regulate Hippo signaling to activate lineage-specific gene expression programs.
Collapse
Affiliation(s)
- Shihadeh Anani
- Goodman Cancer Research Centre, McGill University, 1160 Pine Avenue West, Montreal, QC H3A1A3, Canada Department of Human Genetics, McGill University, 1160 Pine Avenue West, Montreal, QC H3A1A3, Canada
| | - Shivani Bhat
- Goodman Cancer Research Centre, McGill University, 1160 Pine Avenue West, Montreal, QC H3A1A3, Canada
| | - Nobuko Honma-Yamanaka
- Goodman Cancer Research Centre, McGill University, 1160 Pine Avenue West, Montreal, QC H3A1A3, Canada
| | - Dayana Krawchuk
- Goodman Cancer Research Centre, McGill University, 1160 Pine Avenue West, Montreal, QC H3A1A3, Canada
| | - Yojiro Yamanaka
- Goodman Cancer Research Centre, McGill University, 1160 Pine Avenue West, Montreal, QC H3A1A3, Canada Department of Human Genetics, McGill University, 1160 Pine Avenue West, Montreal, QC H3A1A3, Canada
| |
Collapse
|
36
|
Härmä V, Schukov HP, Happonen A, Ahonen I, Virtanen J, Siitari H, Åkerfelt M, Lötjönen J, Nees M. Quantification of dynamic morphological drug responses in 3D organotypic cell cultures by automated image analysis. PLoS One 2014; 9:e96426. [PMID: 24810913 PMCID: PMC4014501 DOI: 10.1371/journal.pone.0096426] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2013] [Accepted: 04/07/2014] [Indexed: 02/03/2023] Open
Abstract
Glandular epithelial cells differentiate into complex multicellular or acinar structures, when embedded in three-dimensional (3D) extracellular matrix. The spectrum of different multicellular morphologies formed in 3D is a sensitive indicator for the differentiation potential of normal, non-transformed cells compared to different stages of malignant progression. In addition, single cells or cell aggregates may actively invade the matrix, utilizing epithelial, mesenchymal or mixed modes of motility. Dynamic phenotypic changes involved in 3D tumor cell invasion are sensitive to specific small-molecule inhibitors that target the actin cytoskeleton. We have used a panel of inhibitors to demonstrate the power of automated image analysis as a phenotypic or morphometric readout in cell-based assays. We introduce a streamlined stand-alone software solution that supports large-scale high-content screens, based on complex and organotypic cultures. AMIDA (Automated Morphometric Image Data Analysis) allows quantitative measurements of large numbers of images and structures, with a multitude of different spheroid shapes, sizes, and textures. AMIDA supports an automated workflow, and can be combined with quality control and statistical tools for data interpretation and visualization. We have used a representative panel of 12 prostate and breast cancer lines that display a broad spectrum of different spheroid morphologies and modes of invasion, challenged by a library of 19 direct or indirect modulators of the actin cytoskeleton which induce systematic changes in spheroid morphology and differentiation versus invasion. These results were independently validated by 2D proliferation, apoptosis and cell motility assays. We identified three drugs that primarily attenuated the invasion and formation of invasive processes in 3D, without affecting proliferation or apoptosis. Two of these compounds block Rac signalling, one affects cellular cAMP/cGMP accumulation. Our approach supports the growing needs for user-friendly, straightforward solutions that facilitate large-scale, cell-based 3D assays in basic research, drug discovery, and target validation.
Collapse
Affiliation(s)
- Ville Härmä
- Medical Biotechnology Knowledge Centre, VTT Technical Research Centre of Finland, Turku, Finland
- Turku Centre for Biotechnology, University of Turku, Turku, Finland
| | - Hannu-Pekka Schukov
- Medical Biotechnology Knowledge Centre, VTT Technical Research Centre of Finland, Turku, Finland
- Turku Centre for Biotechnology, University of Turku, Turku, Finland
| | - Antti Happonen
- Department of Signal Processing, Tampere University of Technology, Tampere, Finland
| | - Ilmari Ahonen
- Department of Information Technology, University of Turku, Turku, Finland
| | - Johannes Virtanen
- Medical Biotechnology Knowledge Centre, VTT Technical Research Centre of Finland, Turku, Finland
- Turku Centre for Biotechnology, University of Turku, Turku, Finland
| | - Harri Siitari
- Medical Biotechnology Knowledge Centre, VTT Technical Research Centre of Finland, Turku, Finland
| | - Malin Åkerfelt
- Medical Biotechnology Knowledge Centre, VTT Technical Research Centre of Finland, Turku, Finland
- Turku Centre for Biotechnology, University of Turku, Turku, Finland
| | - Jyrki Lötjönen
- Knowledge Intensive Services, VTT Technical Research Centre of Finland, Tampere, Finland
| | - Matthias Nees
- Medical Biotechnology Knowledge Centre, VTT Technical Research Centre of Finland, Turku, Finland
- Turku Centre for Biotechnology, University of Turku, Turku, Finland
- * E-mail:
| |
Collapse
|
37
|
Jemielita M, Taormina MJ, DeLaurier A, Kimmel CB, Parthasarathy R. Comparing phototoxicity during the development of a zebrafish craniofacial bone using confocal and light sheet fluorescence microscopy techniques. JOURNAL OF BIOPHOTONICS 2013; 6:920-8. [PMID: 23242824 PMCID: PMC3843985 DOI: 10.1002/jbio.201200144] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/26/2012] [Revised: 10/31/2012] [Accepted: 11/15/2012] [Indexed: 05/25/2023]
Abstract
The combination of genetically encoded fluorescent proteins and three-dimensional imaging enables cell-type-specific studies of embryogenesis. Light sheet microscopy, in which fluorescence excitation is provided by a plane of laser light, is an appealing approach to live imaging due to its high speed and efficient use of photons. While the advantages of rapid imaging are apparent from recent work, the importance of low light levels to studies of development is not well established. We examine the zebrafish opercle, a craniofacial bone that exhibits pronounced shape changes at early developmental stages, using both spinning disk confocal and light sheet microscopies of fluorescent osteoblast cells. We find normal and aberrant opercle morphologies for specimens imaged with short time intervals using light sheet and spinning disk confocal microscopies, respectively, under equivalent exposure conditions over developmentally-relevant time scales. Quantification of shapes reveals that the differently imaged specimens travel along distinct trajectories in morphological space.
Collapse
Affiliation(s)
- Matthew Jemielita
- Department of Physics, 1274 University of Oregon, Eugene, OR, 97403, USA
| | | | - April DeLaurier
- Institute of Neuroscience, 1254 University of Oregon, Eugene, OR, 97403, USA
| | - Charles B. Kimmel
- Institute of Neuroscience, 1254 University of Oregon, Eugene, OR, 97403, USA
| | - Raghuveer Parthasarathy
- Department of Physics, 1274 University of Oregon, Eugene, OR, 97403, USA
- Materials Science Institute, 1252 University of Oregon, Eugene, OR, 97403, USA
- Institute of Molecular Biology, 1229 University of Oregon, Eugene, OR, 97403, USA
| |
Collapse
|
38
|
Rizzi B, Peyrieras N. Towards 3D in silico modeling of the sea urchin embryonic development. J Chem Biol 2013; 7:17-28. [PMID: 24386014 PMCID: PMC3877407 DOI: 10.1007/s12154-013-0101-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2013] [Accepted: 07/22/2013] [Indexed: 11/29/2022] Open
Abstract
Embryogenesis is a dynamic process with an intrinsic variability whose understanding requires the integration of molecular, genetic, and cellular dynamics. Biological circuits function over time at the level of single cells and require a precise analysis of the topology, temporality, and probability of events. Integrative developmental biology is currently looking for the appropriate strategies to capture the intrinsic properties of biological systems. The "-omic" approaches require disruption of the function of the biological circuit; they provide static information, with low temporal resolution and usually with population averaging that masks fast or variable features at the cellular scale and in a single individual. This data should be correlated with cell behavior as cells are the integrators of biological activity. Cellular dynamics are captured by the in vivo microscopy observation of live organisms. This can be used to reconstruct the 3D + time cell lineage tree to serve as the basis for modeling the organism's multiscale dynamics. We discuss here the progress that has been made in this direction, starting with the reconstruction over time of three-dimensional digital embryos from in toto time-lapse imaging. Digital specimens provide the means for a quantitative description of the development of model organisms that can be stored, shared, and compared. They open the way to in silico experimentation and to a more theoretical approach to biological processes. We show, with some unpublished results, how the proposed methodology can be applied to sea urchin species that have been model organisms in the field of classical embryology and modern developmental biology for over a century.
Collapse
Affiliation(s)
- Barbara Rizzi
- CNRS-MDAM, UPR 3294 and BioEmergences-IBiSA, Institut de Neurobiologie Alfred Fessard, CNRS, Gif-sur-Yvette, France
- Institut des Systèmes Complexes, 57-59 rue Lhomond, Paris, France
| | - Nadine Peyrieras
- CNRS-MDAM, UPR 3294 and BioEmergences-IBiSA, Institut de Neurobiologie Alfred Fessard, CNRS, Gif-sur-Yvette, France
- Institut des Systèmes Complexes, 57-59 rue Lhomond, Paris, France
| |
Collapse
|
39
|
Snyder CS, Harrington AR, Kaushal S, Mose E, Lowy AM, Hoffman RM, Bouvet M. A dual-color genetically engineered mouse model for multispectral imaging of the pancreatic microenvironment. Pancreas 2013; 42:952-8. [PMID: 23648841 PMCID: PMC3713119 DOI: 10.1097/mpa.0b013e31828643df] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
OBJECTIVES To develop a mouse model for multispectral fluorescence imaging of the pancreas and pancreatic microenvironment. METHODS Cre/loxP technology was used to develop this model. We crossed mT/mG indicator mice, engineered to constitutively express a conditional tdTomato transgene that converts to green fluorescent protein (GFP) expression after exposure to Cre recombinase, with Pdx1-Cre transgenic mice. To characterize this model for studies of pancreas biology, we performed bright light and fluorescence imaging of body cavities and intact organs and confocal microscopy of pancreata from offspring of Pdx1-Cre and mT/mG crosses. RESULTS Pdx1-Cre-mT/mG mice demonstrated bright GFP expression within the pancreas and duodenum and intense tdTomato expression in all other organs. Green fluorescent protein expression was mosaic in Pdx1-Cre-mT/mG pancreata, with most showing extensive conversion from tdTomato to GFP expression within the epithelial-derived elements of the pancreatic parenchyma. Because both GFP and tdTomato are membrane targeted, individual cell borders were clearly outlined in confocal images of mT/mG pancreata. CONCLUSIONS This mouse model enables multispectral fluorescence imaging of individual cells and cell processes at the microscopic level of the pancreatic microenvironment; it should prove valuable for a variety of fluorescence imaging studies, ranging from pancreatic development to pancreatic cancer biology.
Collapse
Affiliation(s)
- Cynthia S Snyder
- Department of Surgery, UCSD Moores Cancer Center, La Jolla, CA 92093-0987, USA
| | | | | | | | | | | | | |
Collapse
|
40
|
El Omari K, Hoosdally SJ, Tuladhar K, Karia D, Hall-Ponselé E, Platonova O, Vyas P, Patient R, Porcher C, Mancini EJ. Structural basis for LMO2-driven recruitment of the SCL:E47bHLH heterodimer to hematopoietic-specific transcriptional targets. Cell Rep 2013; 4:135-47. [PMID: 23831025 PMCID: PMC3714592 DOI: 10.1016/j.celrep.2013.06.008] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2013] [Revised: 04/23/2013] [Accepted: 06/06/2013] [Indexed: 01/25/2023] Open
Abstract
Cell fate is governed by combinatorial actions of transcriptional regulators assembling into multiprotein complexes. However, the molecular details of how these complexes form are poorly understood. One such complex, which contains the basic-helix-loop-helix heterodimer SCL:E47 and bridging proteins LMO2:LDB1, critically regulates hematopoiesis and induces T cell leukemia. Here, we report the crystal structure of (SCL:E47)bHLH:LMO2:LDB1LID bound to DNA, providing a molecular account of the network of interactions assembling this complex. This reveals an unexpected role for LMO2. Upon binding to SCL, LMO2 induces new hydrogen bonds in SCL:E47, thereby strengthening heterodimer formation. This imposes a rotation movement onto E47 that weakens the heterodimer:DNA interaction, shifting the main DNA-binding activity onto additional protein partners. Along with biochemical analyses, this illustrates, at an atomic level, how hematopoietic-specific SCL sequesters ubiquitous E47 and associated cofactors and supports SCL's reported DNA-binding-independent functions. Importantly, this work will drive the design of small molecules inhibiting leukemogenic processes.
Collapse
Affiliation(s)
- Kamel El Omari
- Division of Structural Biology, The Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK
| | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Xiong F, Tentner AR, Huang P, Gelas A, Mosaliganti KR, Souhait L, Rannou N, Swinburne IA, Obholzer ND, Cowgill PD, Schier AF, Megason SG. Specified neural progenitors sort to form sharp domains after noisy Shh signaling. Cell 2013; 153:550-61. [PMID: 23622240 DOI: 10.1016/j.cell.2013.03.023] [Citation(s) in RCA: 126] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2012] [Revised: 01/22/2013] [Accepted: 03/13/2013] [Indexed: 01/09/2023]
Abstract
Sharply delineated domains of cell types arise in developing tissues under instruction of inductive signal (morphogen) gradients, which specify distinct cell fates at different signal levels. The translation of a morphogen gradient into discrete spatial domains relies on precise signal responses at stable cell positions. However, cells in developing tissues undergoing morphogenesis and proliferation often experience complex movements, which may affect their morphogen exposure, specification, and positioning. How is a clear pattern achieved with cells moving around? Using in toto imaging of the zebrafish neural tube, we analyzed specification patterns and movement trajectories of neural progenitors. We found that specified progenitors of different fates are spatially mixed following heterogeneous Sonic Hedgehog signaling responses. Cell sorting then rearranges them into sharply bordered domains. Ectopically induced motor neuron progenitors also robustly sort to correct locations. Our results reveal that cell sorting acts to correct imprecision of spatial patterning by noisy inductive signals.
Collapse
Affiliation(s)
- Fengzhu Xiong
- Department of Systems Biology, Harvard Medical School, Boston, MA 02115, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Hayes M, Naito M, Daulat A, Angers S, Ciruna B. Ptk7 promotes non-canonical Wnt/PCP-mediated morphogenesis and inhibits Wnt/β-catenin-dependent cell fate decisions during vertebrate development. Development 2013; 140:1807-18. [PMID: 23533179 DOI: 10.1242/dev.090183] [Citation(s) in RCA: 85] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Using zebrafish, we have characterised the function of Protein tyrosine kinase 7 (Ptk7), a transmembrane pseudokinase implicated in Wnt signal transduction during embryonic development and in cancer. Ptk7 is a known regulator of mammalian neural tube closure and Xenopus convergent extension movement. However, conflicting reports have indicated both positive and negative roles for Ptk7 in canonical Wnt/β-catenin signalling. To clarify the function of Ptk7 in vertebrate embryonic patterning and morphogenesis, we generated maternal-zygotic (MZ) ptk7 mutant zebrafish using a zinc-finger nuclease (ZFN) gene targeting approach. Early loss of zebrafish Ptk7 leads to defects in axial convergence and extension, neural tube morphogenesis and loss of planar cell polarity (PCP). Furthermore, during late gastrula and segmentation stages, we observe significant upregulation of β-catenin target gene expression and demonstrate a clear role for Ptk7 in attenuating canonical Wnt/β-catenin activity in vivo. MZptk7 mutants display expanded differentiation of paraxial mesoderm within the tailbud, suggesting an important role for Ptk7 in regulating canonical Wnt-dependent fate specification within posterior stem cell pools post-gastrulation. Furthermore, we demonstrate that a plasma membrane-tethered Ptk7 extracellular fragment is sufficient to rescue both PCP morphogenesis and Wnt/β-catenin patterning defects in MZptk7 mutant embryos. Our results indicate that the extracellular domain of Ptk7 acts as an important regulator of both non-canonical Wnt/PCP and canonical Wnt/β-catenin signalling in multiple vertebrate developmental contexts, with important implications for the upregulated PTK7 expression observed in human cancers.
Collapse
Affiliation(s)
- Madeline Hayes
- Program in Developmental & Stem Cell Biology, The Hospital for Sick Children, Toronto, ON, M5G 1X8, Canada
| | | | | | | | | |
Collapse
|
43
|
Sasai Y. Cytosystems dynamics in self-organization of tissue architecture. Nature 2013; 493:318-26. [DOI: 10.1038/nature11859] [Citation(s) in RCA: 315] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2012] [Accepted: 11/08/2012] [Indexed: 02/08/2023]
|
44
|
Froschauer A, Khatun MM, Sprott D, Franz A, Rieger C, Pfennig F, Gutzeit HO. oct4-EGFP reporter gene expression marks the stem cells in embryonic development and in adult gonads of transgenic medaka. Mol Reprod Dev 2012; 80:48-58. [PMID: 23139203 DOI: 10.1002/mrd.22135] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2012] [Accepted: 10/30/2012] [Indexed: 11/12/2022]
Abstract
Maintenance of pluripotency in stem cells is tightly regulated among vertebrates. One of the key genes in this process is oct4, also referred to as pou5f1 in mammals and pou2 in teleosts. Pou5f1 evolved by duplication of pou2 early in the tetrapod lineage, but only monotremes and marsupials retained both genes. Either pou2 or pou5f1 was lost from the genomes of the other tetrapods that have been analyzed to date. Consequently, these two homologous genes are often designated oct4 in functional studies. In most vertebrates oct4 is expressed in pluripotent cells of the early embryo until the blastula stage, and later persist in germline stem cells until adulthood. The isolation and analysis of stem cells from embryo or adult individuals is hampered by the need for reliable markers that can identify and define the cell populations. Here, we report the faithful expression of EGFP under the control of endogenous pou2/oct4 promoters in transgenic medaka (Oryzias latipes). In vivo imaging in oct4-EGFP transgenic medaka reveals the temporal and spatial expression of pou2 in embryos and adults alike. We describe the temporal and spatial patterns of endogenous pou2 and oct4-EGFP expression in medaka with respect to germline and adult stem cells, and discuss applications of oct4-EGFP transgenic medaka in reproductive and stem cell biology.
Collapse
|
45
|
Xia S, Zhu Y, Xu X, Xia W. Computational techniques in zebrafish image processing and analysis. J Neurosci Methods 2012; 213:6-13. [PMID: 23219894 DOI: 10.1016/j.jneumeth.2012.11.009] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2012] [Revised: 11/10/2012] [Accepted: 11/19/2012] [Indexed: 12/11/2022]
Abstract
The zebrafish (Danio rerio) has been widely used as a vertebrate animal model in neurobiological. The zebrafish has several unique advantages that make it well suited for live microscopic imaging, including its fast development, large transparent embryos that develop outside the mother, and the availability of a large selection of mutant strains. As the genome of zebrafish has been fully sequenced it is comparatively easier to carry out large scale forward genetic screening in zebrafish to investigate relevant human diseases, from neurological disorders like epilepsy, Alzheimer's disease, and Parkinson's disease to other conditions, such as polycystic kidney disease and cancer. All of these factors contribute to an increasing number of microscopic images of zebrafish that require advanced image processing methods to objectively, quantitatively, and quickly analyze the image dataset. In this review, we discuss the development of image analysis and quantification techniques as applied to zebrafish images, with the emphasis on phenotype evaluation, neuronal structure quantification, vascular structure reconstruction, and behavioral monitoring. Zebrafish image analysis is continually developing, and new types of images generated from a wide variety of biological experiments provide the dataset and foundation for the future development of image processing algorithms.
Collapse
Affiliation(s)
- Shunren Xia
- Key laboratory of Biomedical Engineering, of Ministry of Education Zhejiang University, Hangzhou, China.
| | | | | | | |
Collapse
|
46
|
Mosaliganti KR, Noche RR, Xiong F, Swinburne IA, Megason SG. ACME: automated cell morphology extractor for comprehensive reconstruction of cell membranes. PLoS Comput Biol 2012; 8:e1002780. [PMID: 23236265 PMCID: PMC3516542 DOI: 10.1371/journal.pcbi.1002780] [Citation(s) in RCA: 78] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2012] [Accepted: 09/13/2012] [Indexed: 01/21/2023] Open
Abstract
The quantification of cell shape, cell migration, and cell rearrangements is important for addressing classical questions in developmental biology such as patterning and tissue morphogenesis. Time-lapse microscopic imaging of transgenic embryos expressing fluorescent reporters is the method of choice for tracking morphogenetic changes and establishing cell lineages and fate maps in vivo. However, the manual steps involved in curating thousands of putative cell segmentations have been a major bottleneck in the application of these technologies especially for cell membranes. Segmentation of cell membranes while more difficult than nuclear segmentation is necessary for quantifying the relations between changes in cell morphology and morphogenesis. We present a novel and fully automated method to first reconstruct membrane signals and then segment out cells from 3D membrane images even in dense tissues. The approach has three stages: 1) detection of local membrane planes, 2) voting to fill structural gaps, and 3) region segmentation. We demonstrate the superior performance of the algorithms quantitatively on time-lapse confocal and two-photon images of zebrafish neuroectoderm and paraxial mesoderm by comparing its results with those derived from human inspection. We also compared with synthetic microscopic images generated by simulating the process of imaging with fluorescent reporters under varying conditions of noise. Both the over-segmentation and under-segmentation percentages of our method are around 5%. The volume overlap of individual cells, compared to expert manual segmentation, is consistently over 84%. By using our software (ACME) to study somite formation, we were able to segment touching cells with high accuracy and reliably quantify changes in morphogenetic parameters such as cell shape and size, and the arrangement of epithelial and mesenchymal cells. Our software has been developed and tested on Windows, Mac, and Linux platforms and is available publicly under an open source BSD license (https://github.com/krm15/ACME).
Collapse
Affiliation(s)
| | | | | | | | - Sean G. Megason
- Department of Systems Biology, Harvard Medical School, Boston, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
47
|
Höckendorf B, Thumberger T, Wittbrodt J. Quantitative Analysis of Embryogenesis: A Perspective for Light Sheet Microscopy. Dev Cell 2012; 23:1111-20. [DOI: 10.1016/j.devcel.2012.10.008] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2012] [Revised: 10/04/2012] [Accepted: 10/04/2012] [Indexed: 01/06/2023]
|
48
|
Ma R, Distel M, Deán-Ben XL, Ntziachristos V, Razansky D. Non-invasive whole-body imaging of adult zebrafish with optoacoustic tomography. Phys Med Biol 2012; 57:7227-37. [PMID: 23075767 DOI: 10.1088/0031-9155/57/22/7227] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Zebrafish has emerged as an excellent vertebrate model organism for studies of evolution, development and disease. Due to its external development and optical transparency in embryonic stages, zebrafish offers a major advantage over other vertebrate model organisms by being amenable for microscopic studies of biological processes within their natural environment directly in the living organism. However, commonly used zebrafish strains lose their transparency within their first two weeks of development and thus are no longer accessible for optical imaging approaches at juvenile or adult stages. In this study we successfully apply optoacoustic imaging for non-invasive three-dimensional imaging of adult zebrafish. Since optoacoustics does not necessarily require labeling, but can instead rely on the intrinsic tissue contrast, this imaging method has the potential to become a versatile tool for developmental studies from juvenile to adult stages in the intact zebrafish.
Collapse
Affiliation(s)
- Rui Ma
- Technical University of Munich, Ingolstädter Landstraße 1, Neuherberg, Germany
| | | | | | | | | |
Collapse
|
49
|
Ohn J, Tsai HJ, Liebling M. Joint dynamic imaging of morphogenesis and function in the developing heart. Organogenesis 2012; 5:248-55. [PMID: 20539745 DOI: 10.4161/org.5.4.10568] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2009] [Revised: 11/07/2009] [Accepted: 11/09/2009] [Indexed: 01/28/2023] Open
Abstract
In the developing heart, time-lapse imaging is particularly challenging. Changes in heart morphology due to tissue growth or long-term reorganization are difficult to follow because they are much subtler than the rapid shape changes induced by the heartbeat. Therefore, imaging heart development usually requires slowing or stopping the heart. This, however, leads to information loss about the unperturbed heart shape and the dynamics of heart function. To overcome this limitation, we have developed a non-invasive heart imaging technique to jointly document heart function (at fixed stages of development) as well as its morphogenesis (at any fixed phase in the heartbeat) that does not require stopping or slowing the heart. We review the challenges for imaging heart development and our methodology, which is based on computationally combining and analyzing multiple high-speed image sequences acquired throughout the course of development. We present results obtained in the developing zebrafish heart. Image analysis of the acquired data yielded blood flow velocity maps and made it possible to follow the relative movement of individual cells over several hours.
Collapse
Affiliation(s)
- Jungho Ohn
- Electrical & Computer Engineering; University of California; Santa Barbara, CA USA
| | | | | |
Collapse
|
50
|
Nowotschin S, Hadjantonakis AK. Photomodulatable fluorescent proteins for imaging cell dynamics and cell fate. Organogenesis 2012; 5:217-26. [PMID: 20539741 DOI: 10.4161/org.5.4.10939] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2009] [Accepted: 11/09/2009] [Indexed: 12/25/2022] Open
Abstract
An organism arises from the coordinate generation of different cell types and the stereotypical organization of these cells into tissues and organs. Even so, the dynamic behaviors, as well as the ultimate fates, of cells driving the morphogenesis of an organism, or even an individual organ, remain largely unknown. Continued innovations in optical imaging modalities, along with the discovery and evolution of improved genetically-encoded fluorescent protein reporters in combination with model organism, stem cell and tissue engineering paradigms are providing the means to investigate these unresolved questions. The emergence of fluorescent proteins whose spectral properties can be photomodulated is one of the most significant new developments in the field of cell biology where they are primarily used for studying protein dynamics in cells. Likewise, the use of photomodulatable fluorescent proteins holds great promise for use in developmental biology. Photomodulatable fluorescent proteins also represent attractive and emergent tools for studying cell dynamics in complex populations by facilitating the labeling and tracking of individual or defined groups of cells. Here, we review the currently available photomodulatable fluorescent proteins and their application in model organisms. We also discuss prospects for their use in mice, and by extension in embryonic stem cell and tissue engineering paradigms.
Collapse
Affiliation(s)
- Sonja Nowotschin
- Developmental Biology Program; Sloan-Kettering Institute; New York, NY USA
| | | |
Collapse
|