101
|
Minchin JEN, Williams VC, Hinits Y, Low S, Tandon P, Fan CM, Rawls JF, Hughes SM. Oesophageal and sternohyal muscle fibres are novel Pax3-dependent migratory somite derivatives essential for ingestion. Development 2013; 140:2972-84. [PMID: 23760954 DOI: 10.1242/dev.090050] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Striated muscles that enable mouth opening and swallowing during feeding are essential for efficient energy acquisition, and are likely to have played a fundamental role in the success of early jawed vertebrates. The developmental origins and genetic requirements of these muscles are uncertain. Here, we determine by indelible lineage tracing in mouse that fibres of sternohyoid muscle (SHM), which is essential for mouth opening during feeding, and oesophageal striated muscle (OSM), which is crucial for voluntary swallowing, arise from Pax3-expressing somite cells. In vivo Kaede lineage tracing in zebrafish reveals the migratory route of cells from the anteriormost somites to OSM and SHM destinations. Expression of pax3b, a zebrafish duplicate of Pax3, is restricted to the hypaxial region of anterior somites that generate migratory muscle precursors (MMPs), suggesting that Pax3b plays a role in generating OSM and SHM. Indeed, loss of pax3b function led to defective MMP migration and OSM formation, disorganised SHM differentiation, and inefficient ingestion and swallowing of microspheres. Together, our data demonstrate Pax3-expressing somite cells as a source of OSM and SHM fibres, and highlight a conserved role of Pax3 genes in the genesis of these feeding muscles of vertebrates.
Collapse
Affiliation(s)
- James E N Minchin
- Randall Division of Cell and Molecular Biophysics, New Hunt's House, Guy's Campus, King's College London, London SE1 1UL, UK
| | | | | | | | | | | | | | | |
Collapse
|
102
|
Lee RTH, Knapik EW, Thiery JP, Carney TJ. An exclusively mesodermal origin of fin mesenchyme demonstrates that zebrafish trunk neural crest does not generate ectomesenchyme. Development 2013; 140:2923-32. [PMID: 23739134 DOI: 10.1242/dev.093534] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The neural crest is a multipotent stem cell population that arises from the dorsal aspect of the neural tube and generates both non-ectomesenchymal (melanocytes, peripheral neurons and glia) and ectomesenchymal (skeletogenic, odontogenic, cartilaginous and connective tissue) derivatives. In amniotes, only cranial neural crest generates both classes, with trunk neural crest restricted to non-ectomesenchyme. By contrast, it has been suggested that anamniotes might generate derivatives of both classes at all axial levels, with trunk neural crest generating fin osteoblasts, scale mineral-forming cells and connective tissue cells; however, this has not been fully tested. The cause and evolutionary significance of this cranial/trunk dichotomy, and its absence in anamniotes, are debated. Recent experiments have disputed the contribution of fish trunk neural crest to fin osteoblasts and scale mineral-forming cells. This prompted us to test the contribution of anamniote trunk neural crest to fin connective tissue cells. Using genetics-based lineage tracing in zebrafish, we find that these fin mesenchyme cells derive entirely from the mesoderm and that neural crest makes no contribution. Furthermore, contrary to previous suggestions, larval fin mesenchyme cells do not generate the skeletogenic cells of the adult fin, but persist to form fibroblasts associated with adult fin rays. Our data demonstrate that zebrafish trunk neural crest does not generate ectomesenchymal derivatives and challenge long-held ideas about trunk neural crest fate. These findings have important implications for the ontogeny and evolution of the neural crest.
Collapse
Affiliation(s)
- Raymond Teck Ho Lee
- Institute of Molecular and Cell Biology-IMCB, A*STAR-Agency for Science, Technology and Research, 61 Biopolis Drive, Singapore 138673, Singapore
| | | | | | | |
Collapse
|
103
|
Asakawa K, Abe G, Kawakami K. Cellular dissection of the spinal cord motor column by BAC transgenesis and gene trapping in zebrafish. Front Neural Circuits 2013; 7:100. [PMID: 23754985 PMCID: PMC3664770 DOI: 10.3389/fncir.2013.00100] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2013] [Accepted: 05/04/2013] [Indexed: 11/13/2022] Open
Abstract
Bacterial artificial chromosome (BAC) transgenesis and gene/enhancer trapping are effective approaches for identification of genetically defined neuronal populations in the central nervous system (CNS). Here, we applied these techniques to zebrafish (Danio rerio) in order to obtain insights into the cellular architecture of the axial motor column in vertebrates. First, by using the BAC for the Mnx class homeodomain protein gene mnr2b/mnx2b, we established the mnGFF7 transgenic line expressing the Gal4FF transcriptional activator in a large part of the motor column. Single cell labeling of Gal4FF-expressing cells in the mnGFF7 line enabled a detailed investigation of the morphological characteristics of individual spinal motoneurons, as well as the overall organization of the motor column in a spinal segment. Secondly, from a large-scale gene trap screen, we identified transgenic lines that marked discrete subpopulations of spinal motoneurons with Gal4FF. Molecular characterization of these lines led to the identification of the ADAMTS3 gene, which encodes an evolutionarily conserved ADAMTS family of peptidases and is dynamically expressed in the ventral spinal cord. The transgenic fish established here, along with the identified gene, should facilitate an understanding of the cellular and molecular architecture of the spinal cord motor column and its connection to muscles in vertebrates.
Collapse
Affiliation(s)
- Kazuhide Asakawa
- Department of Developmental Genetics, Division of Molecular and Developmental Biology, National Institute of Genetics Mishima, Shizuoka, Japan ; Department of Genetics, Graduate University for Advanced Studies (SOKENDAI) Mishima, Shizuoka, Japan
| | | | | |
Collapse
|
104
|
Peter M, Bathellier B, Fontinha B, Pliota P, Haubensak W, Rumpel S. Transgenic mouse models enabling photolabeling of individual neurons in vivo. PLoS One 2013; 8:e62132. [PMID: 23626779 PMCID: PMC3633923 DOI: 10.1371/journal.pone.0062132] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2012] [Accepted: 03/19/2013] [Indexed: 01/26/2023] Open
Abstract
One of the biggest tasks in neuroscience is to explain activity patterns of individual neurons during behavior by their cellular characteristics and their connectivity within the neuronal network. To greatly facilitate linking in vivo experiments with a more detailed molecular or physiological analysis in vitro, we have generated and characterized genetically modified mice expressing photoactivatable GFP (PA-GFP) that allow conditional photolabeling of individual neurons. Repeated photolabeling at the soma reveals basic morphological features due to diffusion of activated PA-GFP into the dendrites. Neurons photolabeled in vivo can be re-identified in acute brain slices and targeted for electrophysiological recordings. We demonstrate the advantages of PA-GFP expressing mice by the correlation of in vivo firing rates of individual neurons with their expression levels of the immediate early gene c-fos. Generally, the mouse models described in this study enable the combination of various analytical approaches to characterize living cells, also beyond the neurosciences.
Collapse
Affiliation(s)
- Manuel Peter
- Research Institute of Molecular Pathology (IMP), Vienna, Austria
| | - Brice Bathellier
- Research Institute of Molecular Pathology (IMP), Vienna, Austria
| | - Bruno Fontinha
- Research Institute of Molecular Pathology (IMP), Vienna, Austria
| | - Pinelopi Pliota
- Research Institute of Molecular Pathology (IMP), Vienna, Austria
| | - Wulf Haubensak
- Research Institute of Molecular Pathology (IMP), Vienna, Austria
| | - Simon Rumpel
- Research Institute of Molecular Pathology (IMP), Vienna, Austria
- * E-mail:
| |
Collapse
|
105
|
Levesque MP, Krauss J, Koehler C, Boden C, Harris MP. New tools for the identification of developmentally regulated enhancer regions in embryonic and adult zebrafish. Zebrafish 2013; 10:21-9. [PMID: 23461416 DOI: 10.1089/zeb.2012.0775] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
We have conducted a screen to identify developmentally regulated enhancers that drive tissue-specific Gal4 expression in zebrafish. We obtained 63 stable transgenic lines with expression patterns in embryonic or adult zebrafish. The use of a newly identified minimal promoter from the medaka edar locus resulted in a relatively unbiased set of expression patterns representing many tissue types derived from all germ layers. Subsequent detailed characterization of selected lines showed strong and reproducible Gal4-driven GFP expression in diverse tissues, including neurons from the central and peripheral nervous systems, pigment cells, erythrocytes, and peridermal cells. By screening adults for GFP expression, we also isolated lines expressed in tissues of the adult zebrafish, including scales, fin rays, and joints. The new and efficient minimal promoter and large number of transactivating driver-lines we identified will provide the zebrafish community with a useful resource for further enhancer trap screening, as well as precise investigation of tissue-specific processes in vivo.
Collapse
Affiliation(s)
- Mitchell P Levesque
- Department of Genetics, Max-Planck-Institut für Entwicklungsbiologie, Tübingen, Germany .
| | | | | | | | | |
Collapse
|
106
|
Agasti SS, Kohler RH, Liong M, Peterson VM, Lee H, Weissleder R. Dual imaging and photoactivated nanoprobe for controlled cell tracking. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2013; 9:222-7. [PMID: 22996932 PMCID: PMC3552149 DOI: 10.1002/smll.201201007] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/09/2012] [Revised: 06/22/2012] [Indexed: 05/24/2023]
Abstract
A photoactivated nanoprobe for cell labeling and tracking is demonstrated. The nanoprobe enables all targeted cells to be imaged (at 680 nm) as well as specific cells to be photoactivated using 405 nm light. Photoactivated cells can then be tracked (at 525 nm) spatiotemporally in a separate channel over prolonged periods.
Collapse
Affiliation(s)
- Sarit S Agasti
- Center for Systems Biology, Massachusetts General Hospital/Harvard, Medical School, Boston, MA 02114, USA
| | | | | | | | | | | |
Collapse
|
107
|
Embryonic fate map of first pharyngeal arch structures in the sox10: kaede zebrafish transgenic model. J Craniofac Surg 2013; 23:1333-7. [PMID: 22948622 DOI: 10.1097/scs.0b013e318260f20b] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Cranial neural crest cells follow stereotypic patterns of migration to form craniofacial structures. The zebrafish is a powerful vertebrate genetic model where transgenics with reporter proteins under the transcriptional regulation of lineage-specific promoters can be generated. Numerous studies demonstrate that the zebrafish ethmoid plate is embryologically analogous to the mammalian palate. A fate map correlating embryonic cranial neural crest to defined jaw structures would provide a useful context for the morphogenetic analysis of craniofacial development. To that end, the sox10:kaede transgenic was generated, where sox10 provides lineage restriction to the neural crest. Specific regions of neural crest were labeled at the 10-somite stage by photoconversion of the kaede reporter protein. Lineage analysis was carried out during pharyngeal development in wild-type animals, after miR140 injection, and after estradiol treatment. At the 10-somite stage, cranial neural crest cells anterior of the eye contributed to the median ethmoid plate, whereas cells medial to the eye formed the lateral ethmoid plate and trabeculae and a posterior population formed the mandible. miR-140 overexpression and estradiol inhibition of Hedgehog signaling resulted in cleft development, with failed migration of the anterior cell population to form the median ethmoid plate. The sox10:kaede transgenic line provides a useful tool for neural crest lineage analysis. These studies illustrate the advantages of the zebrafish model for application in morphogenetic studies of vertebrate craniofacial development.
Collapse
|
108
|
Itou J, Oishi I, Kawakami H, Glass TJ, Richter J, Johnson A, Lund TC, Kawakami Y. Migration of cardiomyocytes is essential for heart regeneration in zebrafish. Development 2012; 139:4133-42. [PMID: 23034636 DOI: 10.1242/dev.079756] [Citation(s) in RCA: 108] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Adult zebrafish possess a significant ability to regenerate injured heart tissue through proliferation of pre-existing cardiomyocytes, which contrasts with the inability of mammals to do so after the immediate postnatal period. Zebrafish therefore provide a model system in which to study how an injured heart can be repaired. However, it remains unknown what important processes cardiomyocytes are involved in other than partial de-differentiation and proliferation. Here we show that migration of cardiomyocytes to the injury site is essential for heart regeneration. Ventricular amputation induced expression of cxcl12a and cxcr4b, genes encoding a chemokine ligand and its receptor. We found that cxcl12a was expressed in the epicardial tissue and that Cxcr4 was expressed in cardiomyocytes. We show that pharmacological blocking of Cxcr4 function as well as genetic loss of cxcr4b function causes failure to regenerate the heart after ventricular resection. Cardiomyocyte proliferation was not affected but a large portion of proliferating cardiomyocytes remained localized outside the injury site. A photoconvertible fluorescent reporter-based cardiomyocyte-tracing assay demonstrates that cardiomyocytes migrated into the injury site in control hearts but that migration was inhibited in the Cxcr4-blocked hearts. By contrast, the epicardial cells and vascular endothelial cells were not affected by blocking Cxcr4 function. Our data show that the migration of cardiomyocytes into the injury site is regulated independently of proliferation, and that coordination of both processes is necessary for heart regeneration.
Collapse
Affiliation(s)
- Junji Itou
- Department of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, MN 55455, USA
| | | | | | | | | | | | | | | |
Collapse
|
109
|
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
|
110
|
Lombardo VA, Sporbert A, Abdelilah-Seyfried S. Cell tracking using photoconvertible proteins during zebrafish development. J Vis Exp 2012:4350. [PMID: 23052298 DOI: 10.3791/4350] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Embryogenesis is a dynamic process that is best studied by using techniques that allow the documentation of developmental changes in vivo. The use of genetically-encoded fluorescent proteins has proven a valuable strategy for elucidating dynamic morphogenetic processes as they occur in the intact organism. During the past decade, the development of photoactivatable and photoconvertible fluorescent proteins has opened the possibility to investigate the fate of discrete subpopulations of tagged proteins. Unlike photoactivatable proteins, photoconvertible fluorescent proteins (PCFPs) are readily tracked and imaged in their native emission state prior to photoconversion, making it easier to identify and select regions by optical inspection. PCFPs, such as Kaede, KikGR, Dendra and EosFP, can be shifted from green to red upon exposure to UV or blue light due to a His-Tyr-Gly tripeptide sequence which forms a green chromophore that can be photoconverted to a red one by a light-catalyzed β-elimination and subsequent extension of a π-conjugated system. PCFPs and their monomeric variants are useful tools for tracking cells and studying protein dynamics, respectively. During recent years, PCFPs have been expressed in different animal model, such as zebrafish, chicken and mouse for cell fate tracking. Here we report a protocol for cell-specific photoconversion of PCFPs in the living zebrafish embryo and further tracking of photoconverted proteins at later developmental stages. This methodology allows studying, in a tissue-specific manner, cell biological events underlying morphogenesis in the zebrafish animal model.
Collapse
|
111
|
Xu L, Feng Z, Sinha D, Ducos B, Ebenstein Y, Tadmor AD, Gauron C, Le Saux T, Lin S, Weiss S, Vriz S, Jullien L, Bensimon D. Spatiotemporal manipulation of retinoic acid activity in zebrafish hindbrain development via photo-isomerization. Development 2012; 139:3355-62. [DOI: 10.1242/dev.077776] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
All-trans retinoic acid (RA) is a key player in many developmental pathways. Most methods used to study its effects in development involve continuous all-trans RA activation by incubation in a solution of all-trans RA or by implanting all-trans RA-soaked beads at desired locations in the embryo. Here we show that the UV-driven photo-isomerization of 13-cis RA to the trans-isomer (and vice versa) can be used to non-invasively and quantitatively control the concentration of all-trans RA in a developing embryo in time and space. This facilitates the global or local perturbation of developmental pathways with a pulse of all-trans RA of known concentration or its inactivation by UV illumination. In zebrafish embryos in which endogenous synthesis of all-trans RA is impaired, incubation for as little as 5 minutes in 1 nM all-trans RA (a pulse) or 5 nM 13-cis RA followed by 1-minute UV illumination is sufficient to rescue the development of the hindbrain if performed no later than bud stage. However, if subsequent to this all-trans RA pulse the embryo is illuminated (no later than bud stage) for 1 minute with UV light (to isomerize, i.e. deactivate, all-trans RA), the rescue of hindbrain development is impaired. This suggests that all-trans RA is sequestered in embryos that have been transiently exposed to it. Using 13-cis RA isomerization with UV light, we further show that local illumination at bud stage of the head region (but not the tail) is sufficient to rescue hindbrain formation in embryos whose all-trans RA synthetic pathway has been impaired.
Collapse
Affiliation(s)
- Lijun Xu
- Laboratoire de Physique Statistique, Ecole Normale Supérieure, UPMC Université Paris 06, Université Paris Diderot, CNRS UMR8550, 24 rue Lhomond, 75005 Paris, France
- Institut de Biologie de l’Ecole Normale Supérieure (IBENS), 46 rue d’Ulm, 75005 Paris, France
- Ecole Normale Supérieure, Department of Chemistry, UMR CNRS-ENS-UPMC 8640, 24 rue Lhomond, 75005 Paris, France
| | - Zhiping Feng
- Department of Molecular, Cellular and Integrative Physiology, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Deepak Sinha
- Laboratoire de Physique Statistique, Ecole Normale Supérieure, UPMC Université Paris 06, Université Paris Diderot, CNRS UMR8550, 24 rue Lhomond, 75005 Paris, France
- Institut de Biologie de l’Ecole Normale Supérieure (IBENS), 46 rue d’Ulm, 75005 Paris, France
| | - Bertrand Ducos
- Laboratoire de Physique Statistique, Ecole Normale Supérieure, UPMC Université Paris 06, Université Paris Diderot, CNRS UMR8550, 24 rue Lhomond, 75005 Paris, France
- Institut de Biologie de l’Ecole Normale Supérieure (IBENS), 46 rue d’Ulm, 75005 Paris, France
| | - Yuval Ebenstein
- Department of Chemistry and Biochemistry, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Arbel D. Tadmor
- TRON–Translational Oncology, University Medical Center Mainz, Mainz, Germany
| | - Carole Gauron
- Collège de France, Center for Interdisciplinary Research in Biology (CIRB), CNRS, UMR 7241, INSERM, U1050, France
| | - Thomas Le Saux
- Ecole Normale Supérieure, Department of Chemistry, UMR CNRS-ENS-UPMC 8640, 24 rue Lhomond, 75005 Paris, France
| | - Shuo Lin
- Department of Molecular, Cell and Developmental Biology, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Shimon Weiss
- Department of Chemistry and Biochemistry, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Sophie Vriz
- Collège de France, Center for Interdisciplinary Research in Biology (CIRB), CNRS, UMR 7241, INSERM, U1050, France
- Université Paris Diderot, Sorbonne Paris Cité, 75005 Paris, France
| | - Ludovic Jullien
- Ecole Normale Supérieure, Department of Chemistry, UMR CNRS-ENS-UPMC 8640, 24 rue Lhomond, 75005 Paris, France
| | - David Bensimon
- Laboratoire de Physique Statistique, Ecole Normale Supérieure, UPMC Université Paris 06, Université Paris Diderot, CNRS UMR8550, 24 rue Lhomond, 75005 Paris, France
- Institut de Biologie de l’Ecole Normale Supérieure (IBENS), 46 rue d’Ulm, 75005 Paris, France
- Department of Chemistry and Biochemistry, University of California Los Angeles, Los Angeles, CA 90095, USA
| |
Collapse
|
112
|
Kruse-Bend R, Rosenthal J, Quist TS, Veien ES, Fuhrmann S, Dorsky RI, Chien CB. Extraocular ectoderm triggers dorsal retinal fate during optic vesicle evagination in zebrafish. Dev Biol 2012; 371:57-65. [PMID: 22921921 DOI: 10.1016/j.ydbio.2012.08.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2012] [Revised: 07/10/2012] [Accepted: 08/09/2012] [Indexed: 01/30/2023]
Abstract
Dorsal retinal fate is established early in eye development, via expression of spatially restricted dorsal-specific transcription factors in the optic vesicle; yet the events leading to initiation of dorsal fate are not clear. We hypothesized that induction of dorsal fate would require an extraocular signal arising from a neighboring tissue to pattern the prospective dorsal retina, however no such signal has been identified. We used the zebrafish embryo to determine the source, timing, and identity of the dorsal retina-inducing signal. Extensive cell movements occur during zebrafish optic vesicle morphogenesis, however the location of prospective dorsal cells within the early optic vesicle and their spatial relationship to early dorsal markers is currently unknown. Our mRNA expression and fate mapping analyses demonstrate that the dorsolateral optic vesicle is the earliest region to express dorsal specific markers, and cells from this domain contribute to the dorsal retinal pole at 24 hpf. We show that three bmp genes marking dorsal retina at 25 hpf are also expressed extraocularly before retinal patterning begins. We identified gdf6a as a dorsal initiation signal acting from the extraocular non-neural ectoderm during optic vesicle evagination. We find that bmp2b is involved in dorsal retina initiation, acting upstream of gdf6a. Together, this work has identified the nature and source of extraocular signals required to pattern the dorsal retina.
Collapse
Affiliation(s)
- Renee Kruse-Bend
- Department of Neurobiology and Anatomy, 20 North 1900 East, Room 401 MREB, University of Utah, Salt Lake City, UT 84132, USA
| | | | | | | | | | | | | |
Collapse
|
113
|
Breau MA, Wilson D, Wilkinson DG, Xu Q. Chemokine and Fgf signalling act as opposing guidance cues in formation of the lateral line primordium. Development 2012; 139:2246-53. [PMID: 22619392 DOI: 10.1242/dev.080275] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The directional migration of many cell populations occurs as a coherent group. An amenable model is provided by the posterior lateral line in zebrafish, which is formed by a cohesive primordium that migrates from head to tail and deposits future neuromasts at intervals. We found that prior to the onset of migration, the compact state of the primordium is not fully established, as isolated cells with lateral line identity are present caudal to the main primordium. These isolated cells are retained in position such that they fuse with the migrating primordium as it advances, and later contribute to the leading zone and terminal neuromasts. We found that the isolated lateral line cells are positioned by two antagonistic cues: Fgf signalling attracts them towards the primordium, which counteracts Sdf1α/Cxcr4b-mediated caudal attraction. These findings reveal a novel chemotactic role for Fgf signalling in which it enables the coalescence of the lateral line primordium from an initial fuzzy pattern into a compact group of migrating cells.
Collapse
Affiliation(s)
- Marie A Breau
- Division of Developmental Neurobiology, MRC National Institute for Medical Research, London NW7 1AA, UK
| | | | | | | |
Collapse
|
114
|
Li WH, Zheng G. Photoactivatable fluorophores and techniques for biological imaging applications. Photochem Photobiol Sci 2012; 11:460-71. [PMID: 22252510 PMCID: PMC3677749 DOI: 10.1039/c2pp05342j] [Citation(s) in RCA: 100] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2011] [Accepted: 11/28/2011] [Indexed: 11/21/2022]
Abstract
Photoactivatable fluorophores (PAFs) are powerful imaging probes for tracking molecular and cellular dynamics with high spatiotemporal resolution in biological systems. Recent developments in biological microscopy have raised new demands for engineering new PAFs with improved properties, such as high two photon excitation efficiency, reversibility, cellular delivery and targeting. Here we review the history and some of the recent developments in this area, emphasizing our efforts in developing a new class of caged coumarins and related imaging methods for studying dynamic cell-cell communication through gap junction channels, and in extending the application of these caged coumarins to new areas including spatiotemporal control of microRNA activity in vivo.
Collapse
Affiliation(s)
- Wen-hong Li
- Departments of Cell Biology and of Biochemistry, University of Texas Southwestern Medical Center at Dallas, Dallas, Texas 75390, USA.
| | | |
Collapse
|
115
|
Nakayama S, Ikenaga T, Kawakami K, Ono F, Hatta K. Transgenic line with gal4 insertion useful to study morphogenesis of craniofacial perichondrium, vascular endothelium-associated cells, floor plate, and dorsal midline radial glia during zebrafish development. Dev Growth Differ 2012; 54:202-15. [PMID: 22348745 DOI: 10.1111/j.1440-169x.2011.01322.x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Zebrafish is a good model for studying vertebrate development because of the availability of powerful genetic tools. We are interested in the study of the craniofacial skeletal structure of the zebrafish. For this purpose, we performed a gene trap screen and identified a Gal4 gene trap line, SAGFF(LF)134A. We then analyzed the expression pattern of SAGFF(LF)134A;Tg(UAS:GFP) and found that green fluorescent protein (GFP) was expressed not only in craniofacial skeletal elements but also in the vascular system, as well as in the nervous system. In craniofacial skeletal elements, strong GFP expression was detected not only in chondrocytes but also in the perichondrium. In the vascular system, GFP was expressed in endothelium-associated cells. In the spinal cord, strong GFP expression was found in the floor plate, and later in the dorsal radial glia located on the midline. Taking advantage of this transgenic line, which drives Gal4 expression in specific tissues, we crossed SAGFF(LF)134A with several UAS reporter lines. In particular, time-lapse imaging of photoconverted floor-plate cells of SAGFF(LF)134A;Tg(UAS:KikGR) revealed that the floor-plate cells changed their shape within 36 h from cuboidal/trapezoidal to wine glass shaped. Moreover, we identified a novel mode of association between axons and glia. The putative paths for the commissural axons, including pax8-positive CoBL interneurons, were identified as small openings in the basal endfoot of each floor plate. Our results indicate that the transgenic line would be useful for studying the morphogenesis of less-well-characterized tissues of interest, including the perichondrium, dorsal midline radial glia, late-stage floor plate, and vascular endothelium-associated cells.
Collapse
Affiliation(s)
- Sohei Nakayama
- Graduate School of Life Science, University of Hyogo, 3-2-1 Koto, Kamigori, Akou-gun, Hyogo 678-1297, Japan
| | | | | | | | | |
Collapse
|
116
|
Bestman JE, Lee-Osbourne J, Cline HT. In vivo time-lapse imaging of cell proliferation and differentiation in the optic tectum of Xenopus laevis tadpoles. J Comp Neurol 2012; 520:401-33. [PMID: 22113462 PMCID: PMC3366109 DOI: 10.1002/cne.22795] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
We analyzed the function of neural progenitors in the developing central nervous system of Xenopus laevis tadpoles by using in vivo time-lapse confocal microscopy to collect images through the tectum at intervals of 2-24 hours over 3 days. Neural progenitor cells were labeled with fluorescent protein reporters based on expression of endogenous Sox2 transcription factor. With this construct, we identified Sox2-expressing cells as radial glia and as a component of the progenitor pool of cells in the developing tectum that gives rise to neurons and other radial glia. Lineage analysis of individual radial glia and their progeny demonstrated that less than 10% of radial glia undergo symmetric divisions resulting in two radial glia, whereas the majority of radial glia divide asymmetrically to generate neurons and radial glia. Time-lapse imaging revealed the direct differentiation of radial glia into neurons. Although radial glia may guide axons as they navigate to the superficial tectum, we find no evidence that radial glia function as a scaffold for neuronal migration at early stages of tectal development. Over 3 days, the number of labeled cells increased 20%, as the fraction of radial glia dropped and the proportion of neuronal progeny increased to approximately 60% of the labeled cells. Tadpoles provided with short-term visual enhancement generated significantly more neurons, with a corresponding decrease in cell proliferation. Together these results demonstrate that radial glial cells are neural progenitors in the developing optic tectum and reveal that visual experience increases the proportion of neurons generated in an intact animal.
Collapse
Affiliation(s)
- Jennifer E. Bestman
- The Dorris Neuroscience Center, Department of Cell Biology, The Scripps Research Institute, La Jolla, CA 92037
| | | | - Hollis T. Cline
- The Dorris Neuroscience Center, Department of Cell Biology, The Scripps Research Institute, La Jolla, CA 92037
| |
Collapse
|
117
|
Paskaradevan S, Scott IC. The Aplnr GPCR regulates myocardial progenitor development via a novel cell-non-autonomous, Gα(i/o) protein-independent pathway. Biol Open 2012; 1:275-85. [PMID: 23213418 PMCID: PMC3507289 DOI: 10.1242/bio.2012380] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
Myocardial progenitor development involves the migration of cells to the anterior lateral plate mesoderm (ALPM) where they are exposed to the necessary signals for heart development to proceed. Whether the arrival of cells to this location is sufficient, or whether earlier signaling events are required, for progenitor development is poorly understood. Here we demonstrate that in the absence of Aplnr signaling, cells fail to migrate to the heart-forming region of the ALPM. Our work uncovers a previously uncharacterized cell-non-autonomous function for Aplnr signaling in cardiac development. Furthermore, we show that both the single known Aplnr ligand, Apelin, and the canonical Gαi/o proteins that signal downstream of Aplnr are dispensable for Aplnr function in the context of myocardial progenitor development. This novel Aplnr signal can be substituted for by activation of Gata5/Smarcd3 in myocardial progenitors, suggesting a novel mechanism for Aplnr signaling in the establishment of a niche required for the proper migration/development of myocardial progenitor cells.
Collapse
Affiliation(s)
- Sivani Paskaradevan
- Program in Developmental and Stem Cell Biology, The Hospital for Sick Children, 555 University Avenue , Toronto, ON M5G 1X8 , Canada ; Department of Molecular Genetics, University of Toronto , Toronto, ON M5S 1A8 , Canada
| | | |
Collapse
|
118
|
Chahal DS, Chahal HS, Bayles AR, Rudié EM, Helms BA. Synthetic development of cell-permeable polymer colloids decorated with nanocrystal imaging probes optimized for cell tracking. Chem Sci 2012. [DOI: 10.1039/c2sc20206a] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
|
119
|
Xenopoulos P, Nowotschin S, Hadjantonakis AK. Live imaging fluorescent proteins in early mouse embryos. Methods Enzymol 2012; 506:361-89. [PMID: 22341233 DOI: 10.1016/b978-0-12-391856-7.00042-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Mouse embryonic development comprises highly dynamic and coordinated events that drive key cell lineage specification and morphogenetic events. These processes involve cellular behaviors including proliferation, migration, apoptosis, and differentiation, each of which is regulated both spatially and temporally. Live imaging of developing embryos provides an essential tool to investigate these coordinated processes in three-dimensional space over time. For this purpose, the development and application of genetically encoded fluorescent protein (FP) reporters has accelerated over the past decade allowing for the high-resolution visualization of developmental progression. Ongoing efforts are aimed at generating improved reporters, where spectrally distinct as well as novel FPs whose optical properties can be photomodulated, are exploited for live imaging of mouse embryos. Moreover, subcellular tags in combination with using FPs allow for the visualization of multiple subcellular characteristics, such as cell position and cell morphology, in living embryos. Here, we review recent advances in the application of FPs for live imaging in the early mouse embryo, as well as some of the methods used for ex utero embryo development that facilitate on-stage time-lapse specimen visualization.
Collapse
|
120
|
Optical highlighter molecules in neurobiology. Curr Opin Neurobiol 2011; 22:111-20. [PMID: 22129781 DOI: 10.1016/j.conb.2011.11.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2011] [Revised: 11/05/2011] [Accepted: 11/07/2011] [Indexed: 12/18/2022]
Abstract
The development of advanced optical methods has played a key role in propelling progress in neurobiology. Genetically-encoded fluorescent molecules found in nature have enabled labeling of individual neurons to study their physiology and anatomy. Here we discuss the recent use of both native and synthetic optical highlighter proteins to address key problems in neurobiology, including questions relevant to synaptic function, neuroanatomy, and the organization of neural circuits.
Collapse
|
121
|
Abstract
Reconstructing the lineage of cells is central to understanding development and is now also an important issue in stem cell research. Technological advances in genetically engineered permanent cell labeling, together with a multiplicity of fluorescent markers and sophisticated imaging, open new possibilities for prospective and retrospective clonal analysis.
Collapse
Affiliation(s)
- Margaret E Buckingham
- Molecular Genetics of Development Unit, CNRS URA 2578, Department of Developmental Biology, Institut Pasteur, Paris, France.
| | | |
Collapse
|
122
|
Abstract
The ability to differentially label single cells has important implications in developmental biology. For instance, determining how hematopoietic, lymphatic, and blood vessel lineages arise in developing embryos requires fate mapping and lineage tracing of undifferentiated precursor cells. Recently, photoactivatable proteins which include: Eos1, 2, PAmCherry3, Kaede4-7, pKindling8, and KikGR9, 10 have received wide interest as cell tracing probes. The fluorescence spectrum of these photosensitive proteins can be easily converted with UV excitation, allowing a population of cells to be distinguished from adjacent ones. However, the photoefficiency of the activated protein may limit long-term cell tracking11. As an alternative to photoactivatable proteins, caged fluorescein-dextran has been widely used in embryo model systems7, 12-14. Traditionally, to uncage fluorescein-dextran, UV excitation from a fluorescence lamp house or a single photon UV laser has been used; however, such sources limit the spatial resolution of photoactivation. Here we report a protocol to fate map, lineage trace, and detect single labeled cells. Single cells in embryos injected with caged fluorescein-dextran are photoactivated with near-infrared laser pulses produced from a titanium sapphire femtosecond laser. This laser is customary in all two-photon confocal microscopes such as the LSM 510 META NLO microscope used in this paper. Since biological tissue is transparent to near-infrared irradiation15, the laser pulses can be focused deep within the embryo without uncaging cells above or below the selected focal plane. Therefore, non-linear two-photon absorption is induced only at the geometric focus to uncage fluorescein-dextran in a single cell. To detect the cell containing uncaged fluorescein-dextran, we describe a simple immunohistochemistry protocol16 to rapidly visualize the activated cell. The activation and detection protocol presented in this paper is versatile and can be applied to any model system. Note: The reagents used in this protocol can be found in the table appended at the end of the article.
Collapse
Affiliation(s)
- Vikram Kohli
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, USA
| | | | | |
Collapse
|
123
|
Abstract
Despite recognizing the devastating consequences of metastasis, we are not yet able to effectively treat cancer that has spread to vital organs. The inherent complexity of genomic alterations in late-stage cancers, coupled with numerous heterotypic interactions that occur between tumour and stromal cells, represent fundamental challenges in our quest to understand and control metastatic disease. The incorporation of genomic and other systems level approaches, as well as technological breakthroughs in imaging and animal modelling, have galvanized the effort to overcome gaps in our understanding of metastasis. Future research carries with it the potential to translate the wealth of new knowledge and conceptual advances into effective targeted therapies.
Collapse
Affiliation(s)
- Nilay Sethi
- Department of Molecular Biology, Washington Road, LTL 255, Princeton University, Princeton, New Jersey 08544, USA
| | | |
Collapse
|
124
|
Blastomere injection of cleavage-stage zebrafish embryos and imaging of labeled cells. Cold Spring Harb Protoc 2011; 2011:958-66. [PMID: 21807850 DOI: 10.1101/pdb.prot5654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
INTRODUCTIONZebrafish embryos are useful for studying morphogenesis. Their foremost advantage is their optical transparency. Because the embryos are small, they can be imaged as whole-mount preparations. Crucially, this preserves both the mechanical and the molecular integrity of the embryo. This protocol describes the preparation of live zebrafish embryos for the collection of movies using four-dimensional time-lapse confocal microscopy. Methods are presented for the labeling of cells, for the orientation and immobilization of embryos, and for establishing a robust reproducible imaging environment with which to acquire high-resolution dynamic movies of cell behaviors during morphogenesis. These methods concentrate on the visualization of notochord morphogenesis in the zebrafish embryo. However, they also apply to the analysis of many other developing tissues. Factors that can influence the success of time-lapse confocal microscopy are discussed, such as the choice of cell label, the effects of genetic manipulation, and the consequences of environmental variation during imaging—all of which may influence the rate of development.
Collapse
|
125
|
Griswold SL, Sajja KC, Jang CW, Behringer RR. Generation and characterization of iUBC-KikGR photoconvertible transgenic mice for live time-lapse imaging during development. Genesis 2011; 49:591-8. [PMID: 21309067 PMCID: PMC3409694 DOI: 10.1002/dvg.20718] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2010] [Revised: 01/06/2011] [Accepted: 01/11/2011] [Indexed: 11/12/2022]
Abstract
A transgenic mouse line named iUBC-KikGR was generated, which expresses the photoconvertible fluorescent protein Kikume Green-Red (KikGR) under the control of the human Ubiquitin C promoter. KikGR is natively a green fluorophore, which can be converted into a red fluorophore upon exposure to UV light. KikGR is expressed broadly throughout transgenic embryos from the two-cell stage onward and in the adult. Specificity of photoconversion can range from the entire embryo to a region of an organ, to a few individual cells, depending on the needs of the experimenter. Cell movements, tissue reorganization, and migration can then be observed in real time by culturing the tissue of interest as an explant on the microscope stage. The iUBC-KikGR transgenic line represents a singular genetic reagent, which can be used for fate mapping, lineage tracing, and live visualization of cell behaviors and tissue movements in multiple organs at multiple time points.
Collapse
Affiliation(s)
- Shannon L Griswold
- Program in Developmental Biology, Baylor College of Medicine, Houston, Texas, USA
| | | | | | | |
Collapse
|
126
|
Rieger S, Wang F, Sagasti A. Time-lapse imaging of neural development: zebrafish lead the way into the fourth dimension. Genesis 2011; 49:534-45. [PMID: 21305690 PMCID: PMC3132295 DOI: 10.1002/dvg.20729] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2010] [Revised: 01/24/2011] [Accepted: 01/25/2011] [Indexed: 01/01/2023]
Abstract
Time-lapse imaging is often the only way to appreciate fully the many dynamic cell movements critical to neural development. Zebrafish possess many advantages that make them the best vertebrate model organism for live imaging of dynamic development events. This review will discuss technical considerations of time-lapse imaging experiments in zebrafish, describe selected examples of imaging studies in zebrafish that revealed new features or principles of neural development, and consider the promise and challenges of future time-lapse studies of neural development in zebrafish embryos and adults.
Collapse
Affiliation(s)
- Sandra Rieger
- Department of Molecular, Cell, and Developmental Biology, University of California, Los Angeles, California, USA
| | | | | |
Collapse
|
127
|
In vivo imaging of hematopoietic stem cell development in the zebrafish. Front Med 2011; 5:239-47. [DOI: 10.1007/s11684-011-0123-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2010] [Accepted: 02/14/2011] [Indexed: 02/01/2023]
|
128
|
Mittag A, Tárnok A. Perspectives in Cytometry. ADVANCED OPTICAL FLOW CYTOMETRY 2011:1-23. [DOI: 10.1002/9783527634286.ch1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
129
|
Akitake CM, Macurak M, Halpern ME, Goll MG. Transgenerational analysis of transcriptional silencing in zebrafish. Dev Biol 2011; 352:191-201. [PMID: 21223961 PMCID: PMC3065955 DOI: 10.1016/j.ydbio.2011.01.002] [Citation(s) in RCA: 118] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2010] [Revised: 12/31/2010] [Accepted: 01/04/2011] [Indexed: 12/11/2022]
Abstract
The yeast Gal4/UAS transcriptional activation system is a powerful tool for regulating gene expression in Drosophila and has been increasing in popularity for developmental studies in zebrafish. It is also useful for studying the basis of de novo transcriptional silencing. Fluorescent reporter genes under the control of multiple tandem copies of the upstream activator sequence (UAS) often show evidence of variegated expression and DNA methylation in transgenic zebrafish embryos. To characterize this systematically, we monitored the progression of transcriptional silencing of UAS-regulated transgenes that differ in their integration sites and in the repetitive nature of the UAS. Transgenic larvae were examined in three generations for tissue-specific expression of a green fluorescent protein (GFP) reporter and DNA methylation at the UAS. Single insertions containing four distinct upstream activator sequences were far less susceptible to methylation than insertions containing fourteen copies of the same UAS. In addition, transgenes that integrated in or adjacent to transposon sequence exhibited silencing regardless of the number of UAS sites included in the transgene. Placement of promoter-driven Gal4 upstream of UAS-regulated responder genes in a single bicistronic construct also appeared to accelerate silencing and methylation. The results demonstrate the utility of the zebrafish for efficient tracking of gene silencing mechanisms across several generations, as well as provide useful guidelines for optimal Gal4-regulated gene expression in organisms subject to DNA methylation.
Collapse
Affiliation(s)
- Courtney M. Akitake
- Carnegie Institution for Science, Department of Embryology, Baltimore, MD 21218, USA
| | - Michelle Macurak
- Carnegie Institution for Science, Department of Embryology, Baltimore, MD 21218, USA
| | - Marnie E. Halpern
- Carnegie Institution for Science, Department of Embryology, Baltimore, MD 21218, USA
| | - Mary G. Goll
- Carnegie Institution for Science, Department of Embryology, Baltimore, MD 21218, USA
| |
Collapse
|
130
|
Moly PK, Hatta K. Early glycinergic axon contact with the Mauthner neuron during zebrafish development. Neurosci Res 2011; 70:251-9. [PMID: 21397641 DOI: 10.1016/j.neures.2011.03.003] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2011] [Revised: 03/04/2011] [Accepted: 03/04/2011] [Indexed: 10/18/2022]
Abstract
Glycinergic neurons are the major inhibitory neurons in the vertebrate central nervous system. In teleosts, they play important roles in the escape response by regulating the activity of the Mauthner (M-) cells. Here we studied the contact between glycinergic axons and the M-cells in early zebrafish embryos by double immunostaining with an anti-glycine antibody and the 3A10 antibody that labels M-cells. We also studied a transgenic line, Tg(GlyT2:GFP), in which GFP is expressed under the control of the promoter for the glycine transporter-2 gene. The initial contacts by ascending glycinergic axons on the M-soma were observed within 27h post-fertilization (hpf) on the lateral part of the ventral surface of the M-soma. Stochastic labeling of glycinergic neurons was then performed by injecting a GlyT2:GFP construct into early cleaving eggs. We identified the origin of the earliest glycinergic axons that contact the M-soma as commissural neurons, located in the anterior spinal cord, whose axons ascend along the lateral longitudinal fascicles with a short descending branch. We also found, in the fourth rhombomere, late-developed glycinergic commissural neurons whose axons contact anterior or posterior edge of both M-somas. This study provides the first example of the initial development of an inhibitory network on an identifiable neuron in vertebrates.
Collapse
Affiliation(s)
- Pricila Khan Moly
- Graduate School of Life Science, University of Hyogo, Hyogo 678-1297, Japan
| | | |
Collapse
|
131
|
Abstract
Due to the powerful combination of genetic and embryological techniques, the teleost fish Danio rerio has emerged in the last decade as an important model organism for the study of embryonic development. It is relatively easy to inject material such as mRNA or synthetic oligonucleotides to reduce or increase the expression of a gene product. Changes in gene expression can be analyzed at the level of mRNA, by whole-mount in situ hybridization, or at the level of protein, by immunofluorescence. It is also possible to quantitatively analyze protein levels by Western and immunoprecipitation. Cell behavior can be analyzed in detail by cell transplantation and by fate mapping. Because a large number of mutations have been identified in recent years, these methods can be applied in a variety of contexts to provide a deep understanding of gene function that is often more difficult to achieve in other vertebrate model systems.
Collapse
Affiliation(s)
- Yuhua Sun
- Department of Cellular Biology, The University of Georgia, Athens, GA, USA.
| | | | | |
Collapse
|
132
|
Abstract
Compared with other vertebrate animal models, zebrafish (Danio rerio) has its superior advantages for studying stem cell migration. Zebrafish have similar tissues and organs as mammals, where tissue-specific stem cells reside in. Zebrafish eggs are externally fertilized and remain transparent until most of the organs are fully developed. This allows imaging stem cells in vivo very easily. Recently, a zebrafish double pigmentation mutant, casper, became a new popular imaging model in the zebrafish field due to its completely transparent bodies in adulthood. It has been used as an excellent model to study adult hematopoietic stem cell (HSC) in the transplantation setting. The unparalleled imaging power of zebrafish provides great opportunities of tracing stem cells in vivo in the developmental and regenerative context. In this chapter, we use HSC as an example and combine the powerful imaging techniques in zebrafish, to provide protocols for in vivo imaging fluorescence-labeled stem cell migration, stem cell fate tracing in zebrafish embryos, HSC transplantation, and in vivo imaging in both zebrafish embryos and adults. These techniques can also be applied to other types of stem cells in zebrafish embryos and adults.
Collapse
|
133
|
Bibliowicz J, Tittle RK, Gross JM. Toward a better understanding of human eye disease insights from the zebrafish, Danio rerio. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2011; 100:287-330. [PMID: 21377629 PMCID: PMC3159961 DOI: 10.1016/b978-0-12-384878-9.00007-8] [Citation(s) in RCA: 80] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Visual impairment and blindness is widespread across the human population, and the development of therapies for ocular pathologies is of high priority. The zebrafish represents a valuable model organism for studying human ocular disease; it is utilized in eye research to understand underlying developmental processes, to identify potential causative genes for human disorders, and to develop therapies. Zebrafish eyes are similar in morphology, physiology, gene expression, and function to human eyes. Furthermore, zebrafish are highly amenable to laboratory research. This review outlines the use of zebrafish as a model for human ocular diseases such as colobomas, glaucoma, cataracts, photoreceptor degeneration, as well as dystrophies of the cornea and retinal pigmented epithelium.
Collapse
Affiliation(s)
- Jonathan Bibliowicz
- University of Texas at Austin, Section of Molecular Cell and Developmental Biology, Austin, TX 78712 USA
- University of Texas at Austin, Institute for Cellular & Molecular Biology, Austin, TX 78712 USA
| | - Rachel K. Tittle
- University of Texas at Austin, Section of Molecular Cell and Developmental Biology, Austin, TX 78712 USA
| | - Jeffrey M. Gross
- University of Texas at Austin, Section of Molecular Cell and Developmental Biology, Austin, TX 78712 USA
- University of Texas at Austin, Institute for Cellular & Molecular Biology, Austin, TX 78712 USA
- University of Texas at Austin, Institute for Neuroscience, Austin, TX 78712 USA
| |
Collapse
|
134
|
Russek-Blum N, Nabel-Rosen H, Levkowitz G. Two-photon-based photoactivation in live zebrafish embryos. J Vis Exp 2010:1902. [PMID: 21206475 DOI: 10.3791/1902] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
Photoactivation of target compounds in a living organism has proven a valuable approach to investigate various biological processes such as embryonic development, cellular signaling and adult physiology. In this respect, the use of multi-photon microscopy enables quantitative photoactivation of a given light responsive agent in deep tissues at a single cell resolution. As zebrafish embryos are optically transparent, their development can be monitored in vivo. These traits make the zebrafish a perfect model organism for controlling the activity of a variety of chemical agents and proteins by focused light. Here we describe the use of two-photon microscopy to induce the activation of chemically caged fluorescein, which in turn allows us to follow cell's destiny in live zebrafish embryos. We use embryos expressing a live genetic landmark (GFP) to locate and precisely target any cells of interest. This procedure can be similarly used for precise light induced activation of proteins, hormones, small molecules and other caged compounds.
Collapse
|
135
|
|
136
|
|
137
|
Brown SC, Bolte S, Gaudin M, Pereira C, Marion J, Soler MN, Satiat-Jeunemaitre B. Exploring plant endomembrane dynamics using the photoconvertible protein Kaede. THE PLANT JOURNAL : FOR CELL AND MOLECULAR BIOLOGY 2010; 63:696-711. [PMID: 20545892 DOI: 10.1111/j.1365-313x.2010.04272.x] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
Photoactivatable and photoconvertible fluorescent proteins capable of pronounced light-induced spectral changes are a powerful addition to the fluorescent protein toolbox of the cell biologist. They permit specific tracking of one subcellular structure (organelle or cell subdomain) within a differentially labelled population. They also enable pulse-chase analysis of protein traffic. The Kaede gene codes for a tetrameric protein found in the stony coral Trachyphyllia geoffroyi, which emits green fluorescence that irreversibly shifts to red following radiation with UV or violet light. We report here the use of Kaede to explore the plant secretory pathway. Kaede versions of the Golgi marker sialyl-transferase (ST-Kaede) and of the vacuolar pathway marker cardosin A (cardA-Kaede) were engineered. Several optical devices enabling photoconversion and observation of Kaede using these two constructs were assessed to optimize Kaede-based imaging protocols. Photoconverted ST-Kaede red-labelled organelles can be followed within neighbouring populations of non-converted green Golgi stacks, by their gradual development of orange/yellow coloration from de novo synthesis of Golgi proteins (green). Results highlight some aspects on the dynamics of the plant Golgi. For plant bio-imaging, the photoconvertible Kaede offers a powerful tool to track the dynamic behaviour of designated subpopulations of Golgi within living cells, while visualizing the de novo formation of proteins and structures, such as a Golgi stack.
Collapse
Affiliation(s)
- Spencer C Brown
- Laboratoire Dynamique de la Compartimentation Cellulaire, CNRS, Institut des Sciences du Végétal, Centre de recherche de Gif (FRC3115), 91198, Gif-sur-Yvette Cedex, France
| | | | | | | | | | | | | |
Collapse
|
138
|
Chen CH, Sun YH, Pei DS, Zhu ZY. Comparative expression of zebrafish lats1 and lats2 and their implication in gastrulation movements. Dev Dyn 2010; 238:2850-9. [PMID: 19842174 DOI: 10.1002/dvdy.22105] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Large tumor suppressor (Lats) is a Ser/Thr kinase, and it presents an important function in tumor suppression. lats was originally identified in Drosophila and recently in mammals. In mammals, it contains two homologues, lats1 and lats2. In the present study, lats1 and lats2 were characterized from zebrafish (Danio rerio), which is the first report of lats in a nonmammalian vertebrate. The primary structure, genomic organization, and phylogenesis of lats from different species were studied, and the results suggest that lats1 is the direct descendant of invertebrate lats, whereas lats2 is formed by genome duplication. In zebrafish, both lats genes are maternally expressed, while they show distinctly different expression profiles during gastrulation. lats1 is almost ubiquitously expressed through development, and lats2 is more prominently expressed in the non-neural ectoderm region of zebrafish gastrula. Most intriguingly, as revealed by cell tracing and gene expression analysis, morpholino-mediated knockdown of either lats1 or lats2 led to obvious defects of cell migration in gastrulation, indicating the functional significance of lats in gastrulation movements.
Collapse
Affiliation(s)
- Chun-Hong Chen
- Department of Genetics and Center for Developmental Biology, College of Life Sciences, Wuhan University, Wuhan, China
| | | | | | | |
Collapse
|
139
|
Esain V, Postlethwait JH, Charnay P, Ghislain J. FGF-receptor signalling controls neural cell diversity in the zebrafish hindbrain by regulating olig2 and sox9. Development 2010; 137:33-42. [PMID: 20023158 DOI: 10.1242/dev.038026] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
The mechanisms underlying the generation of neural cell diversity are the subject of intense investigation, which has highlighted the involvement of different signalling molecules including Shh, BMP and Wnt. By contrast, relatively little is known about FGF in this process. In this report we identify an FGF-receptor-dependent pathway in zebrafish hindbrain neural progenitors that give rise to somatic motoneurons, oligodendrocyte progenitors and differentiating astroglia. Using a combination of chemical and genetic approaches to conditionally inactivate FGF-receptor signalling, we investigate the role of this pathway. We show that FGF-receptor signalling is not essential for the survival or maintenance of hindbrain neural progenitors but controls their fate by coordinately regulating key transcription factors. First, by cooperating with Shh, FGF-receptor signalling controls the expression of olig2, a patterning gene essential for the specification of somatic motoneurons and oligodendrocytes. Second, FGF-receptor signalling controls the development of both oligodendrocyte progenitors and astroglia through the regulation of sox9, a gliogenic transcription factor the function of which we show to be conserved in the zebrafish hindbrain. Overall, for the first time in vivo, our results reveal a mechanism of FGF in the control of neural cell diversity.
Collapse
Affiliation(s)
- Virginie Esain
- INSERM, U784, Laboratoire de Génétique Moléculaire du Développement, 46 rue d'Ulm, 75230 Paris Cedex 05, France
| | | | | | | |
Collapse
|
140
|
Phan TG, Bullen A. Practical intravital two‐photon microscopy for immunological research: faster, brighter, deeper. Immunol Cell Biol 2010; 88:438-44. [DOI: 10.1038/icb.2009.116] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Tri Giang Phan
- Immunology Programme, Garvan Institute of Medical Research, University of New South Wales Darlinghurst New South Wales Australia
- St Vincent's Clinical School, University of New South Wales Darlinghurst New South Wales Australia
| | - Andrew Bullen
- Department of Pathology, University of California San Francisco San Francisco CA USA
- Department of Biological Imaging Development Centre, University of California San Francisco San Francisco CA USA
| |
Collapse
|
141
|
Abstract
For more than a decade, the zebrafish has proven to be an excellent model organism to investigate the mechanisms of neurogenesis during development. The often cited advantages, namely external development, genetic, and optical accessibility, have permitted direct examination and experimental manipulations of neurogenesis during development. Recent studies have begun to investigate adult neurogenesis, taking advantage of its widespread occurrence in the mature zebrafish brain to investigate the mechanisms underlying neural stem cell maintenance and recruitment. Here we provide a comprehensive overview of the tools and techniques available to study neurogenesis in zebrafish both during development and in adulthood. As useful resources, we provide tables of available molecular markers, transgenic, and mutant lines. We further provide optimized protocols for studying neurogenesis in the adult zebrafish brain, including in situ hybridization, immunohistochemistry, in vivo lipofection and electroporation methods to deliver expression constructs, administration of bromodeoxyuridine (BrdU), and finally slice cultures. These currently available tools have put zebrafish on par with other model organisms used to investigate neurogenesis.
Collapse
Affiliation(s)
- Prisca Chapouton
- Institute of Developmental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | | |
Collapse
|
142
|
Abstract
The zebrafish is one of the leading models for the analysis of the vertebrate visual system. A wide assortment of molecular, genetic, and cell biological approaches is available to study zebrafish visual system development and function. As new techniques become available, genetic analysis and imaging continue to be the strengths of the zebrafish model. In particular, recent developments in the use of transposons and zinc finger nucleases to produce new generations of mutant strains enhance both forward and reverse genetic analysis. Similarly, the imaging of developmental and physiological processes benefits from a wide assortment of fluorescent proteins and the ways to express them in the embryo. The zebrafish is also highly attractive for high-throughput screening of small molecules, a promising strategy to search for compounds with therapeutic potential. Here we discuss experimental approaches used in the zebrafish model to study morphogenetic transformations, cell fate decisions, and the differentiation of fine morphological features that ultimately lead to the formation of the functional vertebrate visual system.
Collapse
Affiliation(s)
- Andrei Avanesov
- Division of Craniofacial and Molecular Genetics, Tufts University, Boston, Massachusetts, USA
| | | |
Collapse
|
143
|
Placinta M, Shen MC, Achermann M, Karlstrom RO. A laser pointer driven microheater for precise local heating and conditional gene regulation in vivo. Microheater driven gene regulation in zebrafish. BMC DEVELOPMENTAL BIOLOGY 2009; 9:73. [PMID: 20042114 PMCID: PMC2810295 DOI: 10.1186/1471-213x-9-73] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/25/2009] [Accepted: 12/30/2009] [Indexed: 01/02/2023]
Abstract
Background Tissue heating has been employed to study a variety of biological processes, including the study of genes that control embryonic development. Conditional regulation of gene expression is a particularly powerful approach for understanding gene function. One popular method for mis-expressing a gene of interest employs heat-inducible heat shock protein (hsp) promoters. Global heat shock of hsp-promoter-containing transgenic animals induces gene expression throughout all tissues, but does not allow for spatial control. Local heating allows for spatial control of hsp-promoter-driven transgenes, but methods for local heating are cumbersome and variably effective. Results We describe a simple, highly controllable, and versatile apparatus for heating biological tissue and other materials on the micron-scale. This microheater employs micron-scale fiber optics and uses an inexpensive laser-pointer as a power source. Optical fibers can be pulled on a standard electrode puller to produce tips of varying sizes that can then be used to reliably heat 20-100 μm targets. We demonstrate precise spatiotemporal control of hsp70l:GFP transgene expression in a variety of tissue types in zebrafish embryos and larvae. We also show how this system can be employed as part of a new method for lineage tracing that would greatly facilitate the study of organogenesis and tissue regulation at any time in the life cycle. Conclusion This versatile and simple local heater has broad utility for the study of gene function and for lineage tracing. This system could be used to control hsp-driven gene expression in any organism simply by bringing the fiber optic tip in contact with the tissue of interest. Beyond these uses for the study of gene function, this device has wide-ranging utility in materials science and could easily be adapted for therapeutic purposes in humans.
Collapse
Affiliation(s)
- Mike Placinta
- Department of Biology, University of Massachusetts, Amherst, MA 01003, USA.
| | | | | | | |
Collapse
|
144
|
Deguchi T, Itoh M, Urawa H, Matsumoto T, Nakayama S, Kawasaki T, Kitano T, Oda S, Mitani H, Takahashi T, Todo T, Sato J, Okada K, Hatta K, Yuba S, Kamei Y. Infrared laser-mediated local gene induction in medaka, zebrafish and Arabidopsis thaliana. Dev Growth Differ 2009; 51:769-75. [DOI: 10.1111/j.1440-169x.2009.01135.x] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
|
145
|
Russek-Blum N, Nabel-Rosen H, Levkowitz G. High resolution fate map of the zebrafish diencephalon. Dev Dyn 2009; 238:1827-35. [PMID: 19504459 DOI: 10.1002/dvdy.21987] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
The diencephalon acts as an interactive site between the sensory, central, and endocrine systems and is one of the most elaborate structures in the vertebrate brain. To better understand the embryonic development and morphogenesis of the diencephalon, we developed an improved photoactivation (uncaging)-based lineage tracing strategy. To determine the exact position of a given diencephalic progenitor domain, we used a transgenic line driving green fluorescent protein (GFP) in cells expressing the proneural protein, Neurogenin1 (Neurog1), which was used as a visible neural plate landmark. This approach facilitated precise labeling of defined groups of cells in the prospective diencephalon of the zebrafish neural plate. In this manner, we labeled multiple overlapping areas of the diencephalon, thereby ensuring both accuracy and reproducibility of our lineage tracing regardless of the dynamic changes of the developing neural plate. We present a fate map of the zebrafish diencephalon at a higher spatial resolution than previously described.
Collapse
Affiliation(s)
- Niva Russek-Blum
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | | | | |
Collapse
|
146
|
Nowotschin S, Hadjantonakis AK. Use of KikGR a photoconvertible green-to-red fluorescent protein for cell labeling and lineage analysis in ES cells and mouse embryos. BMC DEVELOPMENTAL BIOLOGY 2009; 9:49. [PMID: 19740427 PMCID: PMC2872819 DOI: 10.1186/1471-213x-9-49] [Citation(s) in RCA: 101] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/06/2009] [Accepted: 09/09/2009] [Indexed: 11/10/2022]
Abstract
BACKGROUND The use of genetically-encoded fluorescent proteins has revolutionized the fields of cell and developmental biology and in doing so redefined our understanding of the dynamic morphogenetic processes that shape the embryo. With the advent of more accessible and sophisticated imaging technologies as well as an abundance of fluorescent proteins with different spectral characteristics, the dynamic processes taking place in situ in living cells and tissues can now be probed. Photomodulatable fluorescent proteins are one of the emerging classes of genetically-encoded fluorescent proteins. RESULTS We have compared PA-GFP, PS-CFP2, Kaede and KikGR four readily available and commonly used photomodulatable fluorescent proteins for use in ES cells and mice. Our results suggest that the green-to-red photoconvertible fluorescent protein, Kikume Green-Red (KikGR), is most suitable for cell labeling and lineage studies in ES cells and mice because it is developmentally neutral, bright and undergoes rapid and complete photoconversion. We have generated transgenic ES cell lines and strains of mice exhibiting robust widespread expression of KikGR. By efficient photoconversion of KikGR we labeled subpopulations of ES cells in culture, and groups of cells within ex utero cultured mouse embryos. Red fluorescent photoconverted cells and their progeny could be followed for extended periods of time. CONCLUSION Transgenic ES cells and mice exhibiting widespread readily detectable expression of KikGR are indistinguishable from their wild type counterparts and are amenable to efficient photoconversion. They represent novel tools for non-invasive selective labeling specific cell populations and live imaging cell dynamics and cell fate. Genetically-encoded photomodulatable proteins such as KikGR represent emergent attractive alternatives to commonly used vital dyes, tissue grafts and genetic methods for investigating dynamic behaviors of individual cells, collective cell dynamics and fate mapping applications.
Collapse
Affiliation(s)
- Sonja Nowotschin
- Developmental Biology Program, Sloan-Kettering Institute, New York, NY 10065, USA
| | | |
Collapse
|
147
|
de Pater E, Clijsters L, Marques SR, Lin YF, Garavito-Aguilar ZV, Yelon D, Bakkers J. Distinct phases of cardiomyocyte differentiation regulate growth of the zebrafish heart. Development 2009; 136:1633-41. [PMID: 19395641 DOI: 10.1242/dev.030924] [Citation(s) in RCA: 206] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Amongst animal species, there is enormous variation in the size and complexity of the heart, ranging from the simple one-chambered heart of Ciona intestinalis to the complex four-chambered heart of lunged animals. To address possible mechanisms for the evolutionary adaptation of heart size, we studied how growth of the simple two-chambered heart in zebrafish is regulated. Our data show that the embryonic zebrafish heart tube grows by a substantial increase in cardiomyocyte number. Augmented cardiomyocyte differentiation, as opposed to proliferation, is responsible for the observed growth. By using transgenic assays to monitor developmental timing, we visualized for the first time the dynamics of cardiomyocyte differentiation in a vertebrate embryo. Our data identify two previously unrecognized phases of cardiomyocyte differentiation separated in time, space and regulation. During the initial phase, a continuous wave of cardiomyocyte differentiation begins in the ventricle, ends in the atrium, and requires Islet1 for its completion. In the later phase, new cardiomyocytes are added to the arterial pole, and this process requires Fgf signaling. Thus, two separate processes of cardiomyocyte differentiation independently regulate growth of the zebrafish heart. Together, our data support a model in which modified regulation of these distinct phases of cardiomyocyte differentiation has been responsible for the changes in heart size and morphology among vertebrate species.
Collapse
Affiliation(s)
- Emma de Pater
- Hubrecht Institute and University Medical Centre Utrecht, 3584 CT, Utrecht, The Netherlands
| | | | | | | | | | | | | |
Collapse
|
148
|
Nowotschin S, Eakin GS, Hadjantonakis AK. Live-imaging fluorescent proteins in mouse embryos: multi-dimensional, multi-spectral perspectives. Trends Biotechnol 2009; 27:266-76. [PMID: 19339068 PMCID: PMC2878313 DOI: 10.1016/j.tibtech.2009.02.006] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2008] [Revised: 01/29/2009] [Accepted: 02/13/2009] [Indexed: 12/18/2022]
Abstract
Microscopy has always been an obligate tool in the field of developmental biology, a goal of which is to elucidate the essential cellular and molecular interactions that coordinate the specification of different cell types and the establishment of body plans. The 2008 Nobel Prize in chemistry was awarded 'for the discovery and development of the green fluorescent protein, GFP' in recognition that the discovery of genetically encoded fluorescent proteins (FPs) has spearheaded a revolution in applications for imaging of live cells. With the development of more-sophisticated imaging technology and availability of FPs with different spectral characteristics, dynamic processes can now be live-imaged at high resolution in situ in embryos. Here, we review some recent advances in this rapidly evolving field as applied to live-imaging capabilities in the mouse, the most genetically tractable mammalian model organism for embryologists.
Collapse
Affiliation(s)
- Sonja Nowotschin
- Developmental Biology Program, Sloan-Kettering Institute, New York, NY10065, USA
| | | | | |
Collapse
|
149
|
LaMora A, Voigt MM. Cranial sensory ganglia neurons require intrinsic N-cadherin function for guidance of afferent fibers to their final targets. Neuroscience 2009; 159:1175-84. [PMID: 19356698 PMCID: PMC2667798 DOI: 10.1016/j.neuroscience.2009.01.049] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2009] [Revised: 01/21/2009] [Accepted: 01/24/2009] [Indexed: 11/19/2022]
Abstract
Cell adhesion molecules, such as N-cadherin (cdh2), are essential for normal neuronal development, and as such have been implicated in an array of processes including neuronal differentiation and migration, and axon growth and fasciculation. cdh2 is expressed in neurons of the peripheral nervous system during development, but its role in these cells during this time is poorly understood. Using the transgenic zebrafish line, tg(p2xr3.2:eGFP(sl1)), we have examined the involvement of cdh2 in the formation of sensory circuits by the peripheral nervous system. The tg(p2xr3.2:eGFP(sl1)) fish allows visualization of neurons comprising the trigeminal, facial, glossopharyngeal and vagal ganglia and their axons throughout development. Reduction of cdh2 in this line was achieved by either crosses to the cdh2-mutant strain, glass onion (glo) or injection of a cdh2 morpholino (MO) into single-cell embryos. Here we show that cdh2 function is required to alter the directional vectors of growing axons upon reaching intermediate targets. The central axons enter the hindbrain appropriately but fail to turn caudally towards their final targets. Similarly, the peripheral axons extend ventrally, but fail to turn and project along a rostral/caudal axis. Furthermore, by expressing dominant negative cdh2 constructs selectively within cranial sensory ganglia (CSG) neurons, we found that cdh2 function is necessary within the axons to elicit these stereotypic turns, thus demonstrating that cdh2 acts cell autonomously. Together, our in vivo data reveal a novel role for cdh2 in the establishment of circuits by peripheral sensory neurons.
Collapse
Affiliation(s)
- Angela LaMora
- Department of Pharmacological and Physiological Science, Saint Louis University School of Medicine, 1402 S. Grand Blvd., St. Louis, MO 63104
| | - Mark M. Voigt
- Department of Pharmacological and Physiological Science, Saint Louis University School of Medicine, 1402 S. Grand Blvd., St. Louis, MO 63104
| |
Collapse
|
150
|
Sun Z, Gitler AD. Discovery and characterization of three novel synuclein genes in zebrafish. Dev Dyn 2009; 237:2490-5. [PMID: 18521955 DOI: 10.1002/dvdy.21569] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Abstract
The neuronal protein alpha-synuclein has been linked to the pathogenesis of synucleinopathies, a collection of neurodegenerative disorders, including Parkinson's disease. alpha-Synuclein belongs to a family of synuclein genes that includes beta- and gamma-synuclein. However, despite being associated with several fatal human neurodegenerative diseases, little is known about the normal function of synucleins. Here we report the cloning and characterization of three synucleins from zebrafish, sncga, sncgb, and sncb. The sequences of these zebrafish synucleins are very similar to those of the human proteins. We used whole-mount in situ hybridization to analyze their spatial and temporal expression patterns during development. sncgb was expressed exclusively in the notochord, while sncga and sncb were expressed strongly in the nervous system. Our identification of synuclein genes in zebrafish and the characterization of their expression patterns will facilitate future experiments aimed at assessing their functions in normal physiology as well as their role in pathophysiology.
Collapse
Affiliation(s)
- Zhihui Sun
- Department of Cell and Developmental Biology, The University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA
| | | |
Collapse
|