Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
bioRxiv ; 2023 Jul 01.
Article in English | MEDLINE | ID: mdl-37214942

ABSTRACT

During Hedgehog (Hh) signal transduction in development and disease, the atypical G protein-coupled receptor (GPCR) SMOOTHENED (SMO) communicates with GLI transcription factors by binding the protein kinase A catalytic subunit (PKA-C) and physically blocking its enzymatic activity. Here we show that GPCR kinase 2 (GRK2) orchestrates this process during endogenous Hh pathway activation in the primary cilium. Upon SMO activation, GRK2 rapidly relocalizes from the ciliary base to the shaft, triggering SMO phosphorylation and PKA-C interaction. Reconstitution studies reveal that GRK2 phosphorylation enables active SMO to bind PKA-C directly. Lastly, the SMO-GRK2-PKA pathway underlies Hh signal transduction in a range of cellular and in vivo models. Thus, GRK2 phosphorylation of ciliary SMO, and the ensuing PKA-C binding and inactivation, are critical initiating events for the intracellular steps in Hh signaling. More broadly, our study suggests an expanded role for GRKs in enabling direct GPCR interactions with diverse intracellular effectors.

2.
Annu Rev Vis Sci ; 9: 221-243, 2023 09 15.
Article in English | MEDLINE | ID: mdl-37040791

ABSTRACT

Proper eye structure is essential for visual function: Multiple essential eye tissues must take shape and assemble into a precise three-dimensional configuration. Accordingly, alterations to eye structure can lead to pathological conditions of visual impairment. Changes in eye shape can also be adaptive over evolutionary time. Eye structure is first established during development with the formation of the optic cup, which contains the neural retina, retinal pigment epithelium, and lens. This crucial yet deceptively simple hemispherical structure lays the foundation for all later elaborations of the eye. Building on descriptions of the embryonic eye that started with hand drawings and micrographs, the field is beginning to identify mechanisms driving dynamic changes in three-dimensional cell and tissue shape. A combination of molecular genetics, imaging, and pharmacological approaches is defining connections among transcription factors, signaling pathways, and the intracellular machinery governing the emergence of this crucial structure.


Subject(s)
Vertebrates , Vision, Low , Animals , Retina , Retinal Pigment Epithelium , Morphogenesis
3.
Development ; 150(8)2023 04 15.
Article in English | MEDLINE | ID: mdl-36975217

ABSTRACT

Transgenesis is an essential technique for any genetic model. Tol2-based transgenesis paired with Gateway-compatible vector collections has transformed zebrafish transgenesis with an accessible modular system. Here, we establish several next-generation transgenesis tools for zebrafish and other species to expand and enhance transgenic applications. To facilitate gene regulatory element testing, we generated Gateway middle entry vectors harboring the small mouse beta-globin minimal promoter coupled to several fluorophores, CreERT2 and Gal4. To extend the color spectrum for transgenic applications, we established middle entry vectors encoding the bright, blue-fluorescent protein mCerulean and mApple as an alternative red fluorophore. We present a series of p2A peptide-based 3' vectors with different fluorophores and subcellular localizations to co-label cells expressing proteins of interest. Finally, we established Tol2 destination vectors carrying the zebrafish exorh promoter driving different fluorophores as a pineal gland-specific transgenesis marker that is active before hatching and through adulthood. exorh-based reporters and transgenesis markers also drive specific pineal gland expression in the eye-less cavefish (Astyanax). Together, our vectors provide versatile reagents for transgenesis applications in zebrafish, cavefish and other models.


Subject(s)
Gene Transfer Techniques , Zebrafish , Animals , Mice , Zebrafish/genetics , Zebrafish/metabolism , Animals, Genetically Modified , Plasmids/genetics , Promoter Regions, Genetic/genetics , DNA Transposable Elements/genetics
4.
PLoS One ; 17(3): e0265327, 2022.
Article in English | MEDLINE | ID: mdl-35286359

ABSTRACT

Cilia are essential for the development and function of many different tissues. Although cilia machinery is crucial in the eye for photoreceptor development and function, a role for cilia in early eye development and morphogenesis is still somewhat unclear: many zebrafish cilia mutants retain cilia at early stages due to maternal deposition of cilia components. An eye phenotype has been described in the mouse Arl13 mutant, however, zebrafish arl13b is maternally deposited, and an early role for cilia proteins has not been tested in zebrafish eye development. Here we use the zebrafish dzip1 mutant, which exhibits a loss of cilia throughout stages of early eye development, to examine eye development and morphogenesis. We find that in dzip1 mutants, initial formation of the optic cup proceeds normally, however, the optic fissure subsequently fails to close and embryos develop the structural eye malformation ocular coloboma. Further, neural crest cells, which are implicated in optic fissure closure, do not populate the optic fissure correctly, suggesting that their inappropriate localization may be the underlying cause of coloboma. Overall, our results indicate a role for dzip1 in proper neural crest localization in the optic fissure and optic fissure closure.


Subject(s)
Carrier Proteins/metabolism , Coloboma , Optic Disk , Adaptor Proteins, Signal Transducing/metabolism , Animals , Coloboma/genetics , Eye/metabolism , Mesoderm/metabolism , Mice , Optic Disk/metabolism , Zebrafish/genetics , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism
5.
G3 (Bethesda) ; 12(3)2022 03 04.
Article in English | MEDLINE | ID: mdl-35079792

ABSTRACT

Morphogenesis, the formation of three-dimensional organ structures, requires precise coupling of genetic regulation and complex cell behaviors. The genetic networks governing many morphogenetic systems, including that of the embryonic eye, are poorly understood. In zebrafish, several forward genetic screens have sought to identify factors regulating eye development. These screens often look for eye defects at stages after the optic cup is formed and when retinal neurogenesis is under way. This approach can make it difficult to identify mutants specific for morphogenesis, as opposed to neurogenesis. To this end, we carried out a forward genetic, small-scale haploid mutagenesis screen in zebrafish (Danio rerio) to identify factors that govern optic cup morphogenesis. We screened ∼100 genomes and isolated shutdown corner (sco), a mutant that exhibits multiple tissue defects and harbors a ∼10-Mb deletion that encompasses 89 annotated genes. Using a combination of live imaging and antibody staining, we found cell proliferation, cell death, and tissue patterning defects in the sco optic cup. We also observed other phenotypes, including paralysis, neuromuscular defects, and ocular vasculature defects. To date, the largest deletion mutants reported in zebrafish are engineered using CRISPR-Cas9 and are less than 300 kb. Because of the number of genes within the deletion interval, shutdown corner [Df(Chr05:sco)z207] could be a useful resource to the zebrafish community, as it may be helpful for gene mapping, understanding genetic interactions, or studying many genes lost in the mutant.


Subject(s)
Zebrafish Proteins , Zebrafish , Animals , Haploidy , Morphogenesis/genetics , Mutagenesis/genetics , Mutation , Neurogenesis/genetics , Retina , Zebrafish/genetics , Zebrafish Proteins/genetics
6.
Dev Dyn ; 251(4): 625-644, 2022 04.
Article in English | MEDLINE | ID: mdl-34535934

ABSTRACT

BACKGROUND: Pax2 is required for optic fissure development in many organisms, including humans and zebrafish. Zebrafish loss-of-function mutations in pax2a display coloboma, yet the etiology of the morphogenetic defects is unclear. Further, pax2 is duplicated in zebrafish, and a role for pax2b in optic fissure development has not been examined. RESULTS: Using a combination of imaging and molecular genetics, we interrogated a potential role for pax2b and examined how loss of pax2 affects optic fissure development. Although optic fissure formation appears normal in pax2 mutants, an endothelial-specific subset of periocular mesenchyme (POM) fails to initially localize within the optic fissure, yet both neural crest and endothelial-derived POM ectopically accumulate at later stages in pax2a and pax2a; pax2b mutants. Apoptosis is not up-regulated within the optic fissure in pax2 mutants, yet cell death is increased in tissues outside of the optic fissure, and when apoptosis is inhibited, coloboma is partially rescued. In contrast to pax2a, loss of pax2b does not appear to affect optic fissure morphogenesis. CONCLUSIONS: Our results suggest that pax2a, but not pax2b, supports cell survival outside of the optic fissure and POM abundance within it to facilitate optic fissure closure.


Subject(s)
Zebrafish Proteins , Zebrafish , Animals , Cell Survival/genetics , Eye , Mesoderm/metabolism , Morphogenesis/genetics , PAX2 Transcription Factor/genetics , PAX2 Transcription Factor/metabolism , Zebrafish/genetics , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism
7.
J Vis Exp ; (171)2021 05 26.
Article in English | MEDLINE | ID: mdl-34125104

ABSTRACT

Visual system function requires the establishment of precise tissue and organ structures. In the vertebrate eye, structural defects are a common cause of visual impairment, yet mechanisms of eye morphogenesis are still poorly understood. The basic organization of the embryonic eye is conserved throughout vertebrates, thus live imaging of zebrafish embryos has become a powerful approach to directly observe eye development at real time under normal and pathological conditions. Dynamic cell processes including movements, morphologies, interactions, division, and death can be visualized in the embryo. We have developed methods for uniform labeling of subcellular structures and timelapse confocal microscopy of early eye development in zebrafish. This protocol outlines the method of generating capped mRNA for injection into the 1-cell zebrafish embryo, mounting embryos at optic vesicle stage (~12 hours post fertilization, hpf), and performing multi-dimensional timelapse imaging of optic cup morphogenesis on a laser scanning confocal microscope, such that multiple datasets are acquired sequentially in the same imaging session. Such an approach yields data that can be used for a variety of purposes, including cell tracking, volume measurements, three-dimensional (3D) rendering, and visualization. Our approaches allow us to pinpoint the cellular and molecular mechanisms driving optic cup development, in both wild type and genetic mutant conditions. These methods can be employed directly by other groups or adapted to visualize many additional aspects of zebrafish eye development.


Subject(s)
Eye , Zebrafish , Animals , Morphogenesis , Organogenesis , Zebrafish Proteins
8.
Dev Biol ; 476: 128-136, 2021 08.
Article in English | MEDLINE | ID: mdl-33811855

ABSTRACT

The basic structure of the eye, which is crucial for visual function, is established during the embryonic process of optic cup morphogenesis. Molecular pathways of specification and patterning are integrated with spatially distinct cell and tissue shape changes to generate the eye, with discrete domains and structural features: retina and retinal pigment epithelium enwrap the lens, and the optic fissure occupies the ventral surface of the eye and optic stalk. Interest in the underlying cell biology of eye morphogenesis has led to a growing body of work, combining molecular genetics and imaging to quantify cellular processes such as adhesion and actomyosin activity. These studies reveal that intrinsic machinery and spatiotemporally specific extrinsic inputs collaborate to control dynamics of cell movements and morphologies. Here we consider recent advances in our understanding of eye morphogenesis, with a focus on the mechanics of eye formation throughout vertebrate systems, including insights and potential opportunities using organoids, which may provide a tractable system to test hypotheses from embryonic models.


Subject(s)
Eye/embryology , Optic Disk/embryology , Actomyosin/metabolism , Animals , Cell Movement , Eye/metabolism , Eye/pathology , Humans , Lens, Crystalline/embryology , Morphogenesis/genetics , Morphogenesis/physiology , Optic Disk/metabolism , Organogenesis/genetics , Organogenesis/physiology , Retina/embryology , Retinal Pigment Epithelium/cytology , Signal Transduction , Vertebrates/physiology
9.
Elife ; 92020 05 15.
Article in English | MEDLINE | ID: mdl-32412410

ABSTRACT

Efficient precision genome engineering requires high frequency and specificity of integration at the genomic target site. Here, we describe a set of resources to streamline reporter gene knock-ins in zebrafish and demonstrate the broader utility of the method in mammalian cells. Our approach uses short homology of 24-48 bp to drive targeted integration of DNA reporter cassettes by homology-mediated end joining (HMEJ) at high frequency at a double strand break in the targeted gene. Our vector series, pGTag (plasmids for Gene Tagging), contains reporters flanked by a universal CRISPR sgRNA sequence which enables in vivo liberation of the homology arms. We observed high rates of germline transmission (22-100%) for targeted knock-ins at eight zebrafish loci and efficient integration at safe harbor loci in porcine and human cells. Our system provides a straightforward and cost-effective approach for high efficiency gene targeting applications in CRISPR and TALEN compatible systems.


Subject(s)
CRISPR-Associated Proteins/genetics , CRISPR-Cas Systems , Clustered Regularly Interspaced Short Palindromic Repeats , Gene Knock-In Techniques , Genes, Reporter , Green Fluorescent Proteins/genetics , Transcription Activator-Like Effector Nucleases/genetics , Zebrafish/genetics , Animals , Animals, Genetically Modified , CRISPR-Associated Proteins/metabolism , Fibroblasts/metabolism , Gene Expression Regulation , Green Fluorescent Proteins/metabolism , Humans , K562 Cells , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism , RNA, Guide, Kinetoplastida/genetics , RNA, Guide, Kinetoplastida/metabolism , Recombinational DNA Repair , Sequence Homology, Nucleic Acid , Sus scrofa , Transcription Activator-Like Effector Nucleases/metabolism
10.
Development ; 147(4)2020 02 21.
Article in English | MEDLINE | ID: mdl-31988185

ABSTRACT

Organogenesis requires precise interactions between a developing tissue and its environment. In vertebrates, the developing eye is surrounded by a complex extracellular matrix as well as multiple mesenchymal cell populations. Disruptions to either the matrix or periocular mesenchyme can cause defects in early eye development, yet in many cases the underlying mechanism is unknown. Here, using multidimensional imaging and computational analyses in zebrafish, we establish that cell movements in the developing optic cup require neural crest. Ultrastructural analysis reveals that basement membrane formation around the developing eye is also dependent on neural crest, but only specifically around the retinal pigment epithelium. Neural crest cells produce the extracellular matrix protein nidogen: impairing nidogen function disrupts eye development, and, strikingly, expression of nidogen in the absence of neural crest partially restores optic cup morphogenesis. These results demonstrate that eye formation is regulated in part by extrinsic control of extracellular matrix assembly.This article has an associated 'The people behind the papers' interview.


Subject(s)
Basement Membrane/embryology , Eye/embryology , Neural Crest/embryology , Alleles , Animals , CRISPR-Cas Systems , Calcium-Binding Proteins/physiology , Cell Movement , Electrophoresis, Capillary , Extracellular Matrix/physiology , Extracellular Matrix Proteins/physiology , Forkhead Transcription Factors/physiology , Gene Expression Regulation, Developmental , Genotype , Mesoderm/embryology , Microscopy, Electron, Transmission , Morphogenesis , Mutation , Neural Crest/cytology , Organogenesis , Retina/embryology , Retinal Pigment Epithelium/embryology , Signal Transduction , Transcription Factor AP-2/physiology , Zebrafish , Zebrafish Proteins/physiology
11.
Dev Biol ; 458(1): 1-11, 2020 02 01.
Article in English | MEDLINE | ID: mdl-31589834

ABSTRACT

Epithelial morphogenesis, a fundamental aspect of development, generates 3-dimensional tissue structures crucial for organ function. Underlying morphogenetic mechanisms are, in many cases, poorly understood, but mutations that perturb organ development can affect epithelial cell shape and orientation - difficult features to quantify in three dimensions. The basic structure of the eye is established via epithelial morphogenesis: in the embryonic optic cup, the retinal progenitor epithelium enwraps the lens. We previously found that loss of the extracellular matrix protein laminin-alpha1 (lama1) led to mislocalization of apical polarity markers and apparent misorientation of retinal progenitors. We sought to visualize and quantify this phenotype, and determine whether loss of the apical polarity determinant pard3 might rescue the phenotype. To this end, we developed LongAxis, a MATLAB-based program optimized for the retinal progenitor neuroepithelium. LongAxis facilitates 3-dimensional cell segmentation, visualization, and quantification of cell orientation and morphology. Using LongAxis, we find that retinal progenitors in the lama1-/- optic cup are misoriented and slightly less elongated. In the lama1;MZpard3 double mutant, cells are still misoriented, but larger. Therefore, loss of pard3 does not rescue loss of lama1, and in fact uncovers a novel cell size phenotype. LongAxis enables population-level visualization and quantification of retinal progenitor cell orientation and morphology. These results underscore the importance of visualizing and quantifying cell orientation and shape in three dimensions within the retina.


Subject(s)
Cell Shape , Epithelial Cells , Image Processing, Computer-Assisted , Retina , Software , Zebrafish/embryology , Animals , Animals, Genetically Modified , Carrier Proteins/genetics , Carrier Proteins/metabolism , Epithelial Cells/cytology , Epithelial Cells/metabolism , Laminin/genetics , Laminin/metabolism , Retina/cytology , Retina/embryology , Zebrafish/genetics , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism
12.
Mol Biol Cell ; 30(16): 1890-1899, 2019 07 22.
Article in English | MEDLINE | ID: mdl-30785842

ABSTRACT

Epithelial wound healing requires a complex orchestration of cellular rearrangements and movements to restore tissue architecture and function after injury. While it is well known that mechanical forces can affect tissue morphogenesis and patterning, how the biophysical cues generated after injury influence cellular behaviors during tissue repair is not well understood. Using time-lapse confocal imaging of epithelial tissues in living zebrafish larvae, we provide evidence that localized increases in cellular crowding during wound closure promote the extrusion of nonapoptotic cells via mechanically regulated stretch-activated ion channels (SACs). Directed cell migration toward the injury site promoted rapid changes in cell number and generated shifts in tension at cellular interfaces over long spatial distances. Perturbation of SAC activity resulted in failed extrusion and increased proliferation in crowded areas of the tissue. Together, we conclude that localized cell number plays a key role in dictating cellular behaviors that facilitate wound closure and tissue repair.


Subject(s)
Epithelium/pathology , Wound Healing , Actins/metabolism , Animals , Cell Count , Cell Proliferation , Ion Channels/metabolism , Myosins/metabolism , Zebrafish
13.
Development ; 145(22)2018 11 19.
Article in English | MEDLINE | ID: mdl-30333214

ABSTRACT

Establishment of precise three-dimensional tissue structure is vital for organ function. In the visual system, optic fissure and stalk morphogenesis is a crucial yet poorly understood process, disruptions of which can lead to coloboma, a birth defect causing visual impairment. Here, we use four-dimensional imaging, cell tracking, and molecular genetics in zebrafish to define the cell movements underlying normal optic fissure and stalk formation. We determine how these events are disrupted in a coloboma model in which the Hedgehog (Hh) receptor ptch2 is lost, resulting in overactive Hh signaling. In the ptch2 mutant, cells exhibit defective motile behaviors and morphology. Cells that should contribute to the fissure do not arrive at their correct position, and instead contribute to an ectopically large optic stalk. Our results suggest that overactive Hh signaling, through overexpression of downstream transcriptional targets, impairs cell motility underlying optic fissure and stalk formation, via non-cell-autonomous and cell-autonomous mechanisms. More broadly, our cell motility and morphology analyses provide a new framework for studying other coloboma-causing mutations that disrupt optic fissure or stalk formation.


Subject(s)
Cell Movement , Eye/cytology , Eye/growth & development , Hedgehog Proteins/metabolism , Morphogenesis , Signal Transduction , Zebrafish/growth & development , Zebrafish/metabolism , Animals , Eye/anatomy & histology , Models, Biological , Mutation/genetics , Transcription, Genetic , Zebrafish Proteins/metabolism
14.
Dev Biol ; 416(2): 324-37, 2016 08 15.
Article in English | MEDLINE | ID: mdl-27339294

ABSTRACT

The vertebrate eye forms via a complex set of morphogenetic events. The optic vesicle evaginates and undergoes transformative shape changes to form the optic cup, in which neural retina and retinal pigmented epithelium enwrap the lens. It has long been known that a complex, glycoprotein-rich extracellular matrix layer surrounds the developing optic cup throughout the process, yet the functions of the matrix and its specific molecular components have remained unclear. Previous work established a role for laminin extracellular matrix in particular steps of eye development, including optic vesicle evagination, lens differentiation, and retinal ganglion cell polarization, yet it is unknown what role laminin might play in the early process of optic cup formation subsequent to the initial step of optic vesicle evagination. Here, we use the zebrafish lama1 mutant (lama1(UW1)) to determine the function of laminin during optic cup morphogenesis. Using live imaging, we find, surprisingly, that loss of laminin leads to divergent effects on focal adhesion assembly in a spatiotemporally-specific manner, and that laminin is required for multiple steps of optic cup morphogenesis, including optic stalk constriction, invagination, and formation of a spherical lens. Laminin is not required for single cell behaviors and changes in cell shape. Rather, in lama1(UW1) mutants, loss of epithelial polarity and altered adhesion lead to defective tissue architecture and formation of a disorganized retina. These results demonstrate that the laminin extracellular matrix plays multiple critical roles regulating adhesion and polarity to establish and maintain tissue structure during optic cup morphogenesis.


Subject(s)
Eye Proteins/physiology , Laminin/physiology , Lens, Crystalline/embryology , Retina/embryology , Zebrafish Proteins/physiology , Zebrafish/embryology , Animals , Cell Movement , Cell Polarity , Extracellular Matrix/physiology , Eye Proteins/genetics , Focal Adhesions , Laminin/deficiency , Laminin/genetics , Luminescent Proteins/analysis , Luminescent Proteins/genetics , Microscopy, Confocal , Organogenesis , Retina/cytology , Retinal Ganglion Cells/cytology , Retinal Pigment Epithelium/cytology , Retinal Pigment Epithelium/embryology , Time-Lapse Imaging , Zebrafish Proteins/deficiency , Zebrafish Proteins/genetics
15.
Dev Dyn ; 244(6): 785-96, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25694140

ABSTRACT

BACKGROUND: The application of the Gal4/UAS system to enhancer and gene trapping screens in zebrafish has greatly increased the ability to label and manipulate cell populations in multiple tissues, including the central nervous system (CNS). However the ability to select existing lines for specific applications has been limited by the lack of detailed expression analysis. RESULTS: We describe a Gal4 enhancer trap screen in which we used advanced image analysis, including three-dimensional confocal reconstructions and documentation of expression patterns at multiple developmental time points. In all, we have created and annotated 98 lines exhibiting a wide range of expression patterns, most of which include CNS expression. Expression was also observed in nonneural tissues such as muscle, skin epithelium, vasculature, and neural crest derivatives. All lines and data are publicly available from the Zebrafish International Research Center (ZIRC) from the Zebrafish Model Organism Database (ZFIN). CONCLUSIONS: Our detailed documentation of expression patterns, combined with the public availability of images and fish lines, provides a valuable resource for researchers wishing to study CNS development and function in zebrafish. Our data also suggest that many existing enhancer trap lines may have previously uncharacterized expression in multiple tissues and cell types.


Subject(s)
Animals, Genetically Modified/genetics , Central Nervous System/metabolism , DNA-Binding Proteins/genetics , Enhancer Elements, Genetic , Gene Expression Profiling/methods , Gene Expression Regulation, Developmental , Genes, Reporter , Imaging, Three-Dimensional/methods , Nerve Tissue Proteins/genetics , Saccharomyces cerevisiae Proteins/genetics , Transcription Factors/genetics , Zebrafish Proteins/genetics , Zebrafish/genetics , Animals , Animals, Genetically Modified/embryology , Central Nervous System/embryology , DNA Transposable Elements , Databases, Factual , Genes, Synthetic , Luminescent Proteins/analysis , Luminescent Proteins/genetics , Mutagenesis, Insertional , Nerve Tissue Proteins/biosynthesis , Neurons/metabolism , Organ Specificity , Zebrafish/embryology , Zebrafish Proteins/biosynthesis , Red Fluorescent Protein
16.
Dev Dyn ; 243(10): 1242-8, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25044784

ABSTRACT

The vertebrate eye acquires its basic form during the process of optic cup morphogenesis, during which the optic vesicle emerges from the brain neuroepithelium and, through a series of cell and tissue movements, transforms itself into the multilayered optic cup, containing neural retina (comprised of retinal progenitors), retinal pigmented epithelium, and the lens, which is derived from the overlying ectoderm. While great strides have been made to understand the developmental signals controlling specification, patterning, and differentiation of the optic cup, only in recent years have the cellular and molecular bases of optic cup morphogenesis begun to be unraveled. One critical component of the morphogenetic process is the extracellular matrix: the complex, glycoprotein-rich layer that surrounds the optic vesicle and lens. Though the extracellular matrix has long been visualized by classical histological techniques and postulated to play various roles in optic cup development, its functional role was uncertain. This is now beginning to change, as live imaging techniques, quantitative image analyses, molecular genetics and in vitro models yield new insights into the process of optic cup morphogenesis and the specific influences of particular extracellular matrix components and their associated signaling pathways.


Subject(s)
Extracellular Matrix/physiology , Eye/embryology , Morphogenesis , Vertebrates/embryology , Animals , Humans , Lens, Crystalline/cytology , Lens, Crystalline/embryology , Retina/embryology , Retinal Pigment Epithelium/embryology , Retinal Pigment Epithelium/physiology
17.
Development ; 139(2): 359-72, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22186726

ABSTRACT

Optic cup morphogenesis (OCM) generates the basic structure of the vertebrate eye. Although it is commonly depicted as a series of epithelial sheet folding events, this does not represent an empirically supported model. Here, we combine four-dimensional imaging with custom cell tracking software and photoactivatable fluorophore labeling to determine the cellular dynamics underlying OCM in zebrafish. Although cell division contributes to growth, we find it dispensable for eye formation. OCM depends instead on a complex set of cell movements coordinated between the prospective neural retina, retinal pigmented epithelium (RPE) and lens. Optic vesicle evagination persists for longer than expected; cells move in a pinwheel pattern during optic vesicle elongation and retinal precursors involute around the rim of the invaginating optic cup. We identify unanticipated movements, particularly of central and peripheral retina, RPE and lens. From cell tracking data, we generate retina, RPE and lens subdomain fate maps, which reveal novel adjacencies that might determine corresponding developmental signaling events. Finally, we find that similar movements also occur during chick eye morphogenesis, suggesting that the underlying choreography is conserved among vertebrates.


Subject(s)
Cell Movement/physiology , Eye/embryology , Morphogenesis/physiology , Signal Transduction/physiology , Zebrafish/embryology , Analysis of Variance , Animals , Cell Cycle/physiology , Chick Embryo , Image Processing, Computer-Assisted , Imaging, Three-Dimensional , Lens, Crystalline/physiology , Retina/cytology , Retina/physiology , Retinal Pigment Epithelium/cytology , Retinal Pigment Epithelium/physiology , Time Factors
18.
Dev Dyn ; 236(11): 3088-99, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17937395

ABSTRACT

Transgenesis is an important tool for assessing gene function. In zebrafish, transgenesis has suffered from three problems: the labor of building complex expression constructs using conventional subcloning; low transgenesis efficiency, leading to mosaicism in transient transgenics and infrequent germline incorporation; and difficulty in identifying germline integrations unless using a fluorescent marker transgene. The Tol2kit system uses site-specific recombination-based cloning (multisite Gateway technology) to allow quick, modular assembly of [promoter]-[coding sequence]-[3' tag] constructs in a Tol2 transposon backbone. It includes a destination vector with a cmlc2:EGFP (enhanced green fluorescent protein) transgenesis marker and a variety of widely useful entry clones, including hsp70 and beta-actin promoters; cytoplasmic, nuclear, and membrane-localized fluorescent proteins; and internal ribosome entry sequence-driven EGFP cassettes for bicistronic expression. The Tol2kit greatly facilitates zebrafish transgenesis, simplifies the sharing of clones, and enables large-scale projects testing the functions of libraries of regulatory or coding sequences.


Subject(s)
Animals, Genetically Modified , Cloning, Molecular/methods , DNA Transposable Elements , DNA, Recombinant/genetics , Gene Transfer Techniques , Zebrafish/genetics , Animals , Genetic Techniques , Genetic Vectors , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Plasmids/genetics , Recombination, Genetic , Transposases/metabolism , Zebrafish/metabolism
19.
Development ; 132(20): 4599-610, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16176947

ABSTRACT

During Xenopus development, convergent extension movements mediated by cell intercalation drive axial elongation. While many genes required for convergent extension have been identified, little is known of regulation of the cytoskeleton during these cell movements. Although microtubules are required for convergent extension, this applies only to initial stages of gastrulation, between stages 10 and 10.5. To examine the cytoskeleton more directly during convergent extension, we visualized actin and microtubules simultaneously in live explants using spinning disk confocal fluorescence microscopy. Microtubule depolymerization by nocodazole inhibits lamellipodial protrusions and cell-cell contact, thereby inhibiting convergent extension. However, neither taxol nor vinblastine, both of which block microtubule dynamics while stabilizing a polymer form of tubulin, inhibits lamellipodia or convergent extension. This suggests an unusual explanation: the mass of polymerized tubulin, not dynamics of the microtubule cytoskeleton, is crucial for convergent extension. Because microtubule depolymerization elicits striking effects on actin-based protrusions, the role of Rho-family GTPases was tested. The effects of nocodazole are partially rescued using dominant negative Rho, Rho-kinase inhibitor, or constitutively active Rac, suggesting that microtubules regulate small GTPases, possibly via a guanine-nucleotide exchange factor. We cloned full-length XLfc, a microtubule-binding Rho-GEF. Nucleotide exchange activity of XLfc is required for nocodazole-mediated inhibition of convergent extension; constitutively active XLfc recapitulates the effects of microtubule depolymerization. Morpholino knockdown of XLfc abrogates the ability of nocodazole to inhibit convergent extension. Therefore, we believe that XLfc is a crucial regulator of cell morphology during convergent extension, and microtubules limit its activity through binding to the lattice.


Subject(s)
Guanine Nucleotide Exchange Factors/metabolism , Microtubules/metabolism , Xenopus laevis/metabolism , Animals , Cell Shape , Gastrula/metabolism , Gene Expression Regulation, Developmental , Guanine Nucleotide Exchange Factors/genetics , Microtubules/drug effects , Microtubules/genetics , Nocodazole/pharmacology , Protein Binding , Protein Kinase Inhibitors/pharmacology , Pseudopodia/genetics , Pseudopodia/metabolism , Rho Guanine Nucleotide Exchange Factors , Time Factors , Xenopus laevis/embryology , Xenopus laevis/genetics , cdc42 GTP-Binding Protein/genetics , cdc42 GTP-Binding Protein/metabolism , rac GTP-Binding Proteins/genetics , rac GTP-Binding Proteins/metabolism , rho GTP-Binding Proteins/genetics , rho GTP-Binding Proteins/metabolism
20.
Curr Biol ; 14(1): 75-80, 2004 Jan 06.
Article in English | MEDLINE | ID: mdl-14711418

ABSTRACT

In metazoan oocytes, a metaphase arrest coordinates the completion of meiosis with fertilization. Vertebrate mos maintains the metaphase II arrest of mature oocytes and prevents DNA replication between the meiotic divisions. We identified a Drosophila homolog of mos and showed it to be the mos ortholog by two additional criteria. The dmos transcripts are present in Drosophila oocytes but not embryos, and injection of dmos into Xenopus embryos blocks mitosis and elevates active MAPK levels. In Drosophila, MAPK is activated in oocytes, consistent with a role in meiosis. We generated deletions of dmos and found that, as in vertebrates, dmos is responsible for the majority of MAPK activation. Unexpectedly, the oocytes that do mature complete meiosis normally and produce fertilized embryos that develop, although there is a reduction in female fertility and loss of some oocytes by apoptosis. Therefore, Drosophila contains a mos ortholog that activates a MAPK cascade during oogenesis and is nonessential for meiosis. This could be because there are redundant pathways regulating meiosis, because residual, low levels of active MAPK are sufficient, or because active MAPK is dispensable for meiosis in Drosophila. These results highlight the complexity of meiotic regulation that evolved to ensure accurate control over the reproductive process.


Subject(s)
Genes, mos/genetics , MAP Kinase Signaling System/genetics , Meiosis/physiology , Oocytes/metabolism , Oogenesis/physiology , Amino Acid Sequence , Animals , Blotting, Western , Drosophila , Female , Fertility/physiology , Gene Components , In Situ Hybridization , MAP Kinase Signaling System/physiology , Meiosis/genetics , Molecular Sequence Data , Sequence Alignment , Xenopus/embryology , Xenopus/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...