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










Publication year range
1.
Dev Cell ; 59(1): 141-155.e6, 2024 Jan 08.
Article in English | MEDLINE | ID: mdl-38091998

ABSTRACT

Morphogenetic movements during animal development involve repeated making and breaking of cell-cell contacts. Recent biophysical models of cell-cell adhesion integrate adhesion molecule interactions and cortical cytoskeletal tension modulation, describing equilibrium states for established contacts. We extend this emerging unified concept of adhesion to contact formation kinetics, showing that aggregating Xenopus embryonic cells rapidly achieve Ca2+-independent low-contact states. Subsequent transitions to cadherin-dependent high-contact states show rapid decreases in contact cortical F-actin levels but slow contact area growth. We developed a biophysical model that predicted contact growth quantitatively from known cellular and cytoskeletal parameters, revealing that elastic resistance to deformation and cytoskeletal network turnover are essential determinants of adhesion kinetics. Characteristic time scales of contact growth to low and high states differ by an order of magnitude, being at a few minutes and tens of minutes, respectively, thus providing insight into the timescales of cell-rearrangement-dependent tissue movements.


Subject(s)
Cadherins , Gastrula , Animals , Cell Adhesion , Xenopus laevis , Gastrula/metabolism , Cadherins/metabolism , Cell Adhesion Molecules
2.
Front Cell Dev Biol ; 10: 982477, 2022.
Article in English | MEDLINE | ID: mdl-36133924

ABSTRACT

The T-box family transcription factor Eomesodermin (Eomes) is present in all vertebrates, with many key roles in the developing mammalian embryo and immune system. Homozygous Eomes mutant mouse embryos exhibit early lethality due to defects in both the embryonic mesendoderm and the extraembryonic trophoblast cell lineage. In contrast, zebrafish lacking the predominant Eomes homologue A (Eomesa) do not suffer complete lethality and can be maintained. This suggests fundamental differences in either the molecular function of Eomes orthologues or the molecular configuration of processes in which they participate. To explore these hypotheses we initially analysed the expression of distinct Eomes isoforms in various mouse cell types. Next we compared the functional capabilities of these murine isoforms to zebrafish Eomesa. These experiments provided no evidence for functional divergence. Next we examined the functions of zebrafish Eomesa and other T-box family members expressed in early development, as well as its paralogue Eomesb. Though Eomes is a member of the Tbr1 subfamily we found evidence for functional redundancy with the Tbx6 subfamily member Tbx16, known to be absent from eutherians. However, Tbx16 does not appear to synergise with Eomesa cofactors Mixl1 and Gata5. Finally, we analysed the ability of Eomesa and other T-box factors to induce zebrafish left-right organiser progenitors (known as dorsal forerunner cells) known to be positively regulated by vgll4l, a gene we had previously shown to be repressed by Eomesa. Here we demonstrate that Eomesa indirectly upregulates vgll4l expression via interlocking feedforward loops, suggesting a role in establishment of left-right asymmetry. Conversely, other T-box factors could not similarly induce left-right organiser progenitors. Overall these findings demonstrate conservation of Eomes molecular function and participation in similar processes, but differential requirements across evolution due to additional co-expressed T-box factors in teleosts, albeit with markedly different molecular capabilities. Our analyses also provide insights into the role of Eomesa in left-right organiser formation in zebrafish.

4.
Elife ; 102021 03 23.
Article in English | MEDLINE | ID: mdl-33755014

ABSTRACT

In emerging epithelial tissues, cells undergo dramatic rearrangements to promote tissue shape changes. Dividing cells remain interconnected via transient cytokinetic bridges. Bridges are cleaved during abscission and currently, the consequences of disrupting abscission in developing epithelia are not well understood. We show that the Rab GTPase Rab25 localizes near cytokinetic midbodies and likely coordinates abscission through endomembrane trafficking in the epithelium of the zebrafish gastrula during epiboly. In maternal-zygotic Rab25a and Rab25b mutant embryos, morphogenic activity tears open persistent apical cytokinetic bridges that failed to undergo timely abscission. Cytokinesis defects result in anisotropic cell morphologies that are associated with a reduction of contractile actomyosin networks. This slows cell rearrangements and alters the viscoelastic responses of the tissue, all of which likely contribute to delayed epiboly. We present a model in which Rab25 trafficking coordinates cytokinetic bridge abscission and cortical actin density, impacting local cell shape changes and tissue-scale forces.


Subject(s)
Cell Movement/genetics , Zebrafish/physiology , rab GTP-Binding Proteins/genetics , Animals , Cytokinesis , Embryo, Nonmammalian/physiology , Epithelium/physiology , Gastrula/physiology , Zebrafish/genetics , Zebrafish Proteins , rab GTP-Binding Proteins/metabolism
5.
Mech Dev ; 163: 103625, 2020 09.
Article in English | MEDLINE | ID: mdl-32526279

ABSTRACT

The Brachyury gene encodes a transcription factor that is conserved across all animals. In non-chordate metazoans, brachyury is primarily expressed in ectoderm regions that are added to the endodermal gut during development, and often form a ring around the site of endoderm internalization in the gastrula, the blastopore. In chordates, this brachyury ring is conserved, but the gene has taken on a new role in the formation of the mesoderm. In this phylum, a novel type of mesoderm that develops into notochord and somites has been added to the ancestral lateral plate mesoderm. Brachyury contributes to a shift in cell fate from neural ectoderm to posterior notochord and somites during a major lineage segregation event that in Xenopus and in the zebrafish takes place in the early gastrula. In the absence of this brachyury function, impaired formation of posterior mesoderm indirectly affects the gastrulation movements of peak involution and convergent extension. These movements are confined to specific regions and stages, leaving open the question why brachyury expression in an extensive, coherent ring, before, during and after gastrulation, is conserved in the two species whose gastrulation modes differ considerably, and also in many other metazoan gastrulae of diverse structure.


Subject(s)
Ectoderm/growth & development , Fetal Proteins/genetics , Gastrula/growth & development , Morphogenesis/genetics , T-Box Domain Proteins/genetics , Animals , Endoderm/growth & development , Fetal Proteins/ultrastructure , Mesoderm/growth & development , Notochord/growth & development , T-Box Domain Proteins/ultrastructure , Xenopus laevis/genetics , Xenopus laevis/growth & development , Zebrafish/genetics , Zebrafish/growth & development
6.
Curr Top Dev Biol ; 136: 319-341, 2020.
Article in English | MEDLINE | ID: mdl-31959293

ABSTRACT

Epiboly is a conserved gastrulation movement describing the thinning and spreading of a sheet or multi-layer of cells. The zebrafish embryo has emerged as a vital model system to address the cellular and molecular mechanisms that drive epiboly. In the zebrafish embryo, the blastoderm, consisting of a simple squamous epithelium (the enveloping layer) and an underlying mass of deep cells, as well as a yolk nuclear syncytium (the yolk syncytial layer) undergo epiboly to internalize the yolk cell during gastrulation. The major events during zebrafish epiboly are: expansion of the enveloping layer and the internal yolk syncytial layer, reduction and removal of the yolk membrane ahead of the advancing blastoderm margin and deep cell rearrangements between the enveloping layer and yolk syncytial layer to thin the blastoderm. Here, work addressing the cellular and molecular mechanisms as well as the sources of the mechanical forces that underlie these events is reviewed. The contribution of recent findings to the current model of epiboly as well as open questions and future prospects are also discussed.


Subject(s)
Blastoderm/physiology , Body Patterning , Embryo, Nonmammalian/physiology , Epithelium/physiology , Gastrulation , Morphogenesis , Zebrafish/physiology , Animals , Blastoderm/cytology , Cell Movement , Embryo, Nonmammalian/cytology , Gastrula/cytology , Gastrula/physiology , Gene Expression Regulation, Developmental , Transcription Factors , Zebrafish/embryology , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism
7.
Dev Dyn ; 248(10): 997-1008, 2019 10.
Article in English | MEDLINE | ID: mdl-31390119

ABSTRACT

BACKGROUND: During zebrafish epiboly, the embryonic cell mass, or blastoderm, spreads to enclose the yolk cell. The blastoderm consists of an outer epithelial sheet, the enveloping layer (EVL), and the underlying deep cell layer (DEL). Studies have provided insights into the mechanisms of EVL and deep cell epiboly, but little is known about the interactions between the two cell layers and what role they may play during epiboly. RESULTS: We used live imaging to examine EVL basal protrusions. We identified them as filopodia based on f-actin content and localization of fluorescently tagged filopodial markers. A spatiotemporal analysis revealed that the largest number of EVL filopodia were present during early epiboly at the animal pole. In functional studies, expression of a constitutively active actin-bundling protein resulted in increased filopodial length and delayed gastrulation. CONCLUSIONS: We identified protrusions on the basal surface of EVL cells as filopodia and showed that they are present throughout the EVL during epiboly. The largest number of filopodia was at the animal pole during early epiboly, which is when and where deep cell radial intercalations occur to the greatest extent. These findings suggest that EVL filopodia may function during epiboly to promote deep cell rearrangements during epiboly initiation.


Subject(s)
Epithelium/ultrastructure , Pseudopodia/ultrastructure , Spatio-Temporal Analysis , Actins/metabolism , Animals , Blastoderm/cytology , Embryo, Nonmammalian , Gastrulation , Zebrafish
8.
Development ; 146(1)2019 01 02.
Article in English | MEDLINE | ID: mdl-30509968

ABSTRACT

In teleost fish, the multinucleate yolk syncytial layer functions as an extra-embryonic signaling center to pattern mesendoderm, coordinate morphogenesis and supply nutrients to the embryo. External yolk syncytial nuclei (e-YSN) undergo microtubule-dependent movements that distribute the nuclei over the large yolk mass. How e-YSN migration proceeds, and the role of the yolk microtubules, is not understood, but it is proposed that e-YSN are pulled vegetally as the microtubule network shortens from the vegetal pole. Live imaging revealed that nuclei migrate along microtubules, consistent with a cargo model in which e-YSN are moved down the microtubules by direct association with motor proteins. We found that blocking the plus-end directed microtubule motor kinesin significantly attenuated yolk nuclear movement. Blocking the outer nuclear membrane LINC complex protein Syne2a also slowed e-YSN movement. We propose that e-YSN movement is mediated by the LINC complex, which functions as the adaptor between yolk nuclei and motor proteins. Our work provides new insights into the role of microtubules in morphogenesis of an extra-embryonic tissue and further contributes to the understanding of nuclear migration mechanisms during development.


Subject(s)
Cell Movement , Cell Nucleus/metabolism , Giant Cells/cytology , Models, Biological , Zebrafish/embryology , Zebrafish/metabolism , Animals , Dyneins/metabolism , Embryo, Nonmammalian/cytology , Embryo, Nonmammalian/metabolism , Kinesins/metabolism , Microtubules/metabolism , Time-Lapse Imaging
9.
Dev Cell ; 47(3): 377-387.e4, 2018 11 05.
Article in English | MEDLINE | ID: mdl-30399336

ABSTRACT

Embryos have a striking ability to heal wounds rapidly and without scarring. Embryonic wound repair is a conserved process, driven by polarization of cell-cell junctions and the actomyosin cytoskeleton in the cells around the wound. However, the upstream signals that trigger cell polarization around wounds are unknown. We used quantitative in vivo microscopy in Drosophila and zebrafish embryos to identify reactive oxygen species (ROS) as a critical signal that orchestrates cell polarity around wounds. ROS promote trafficking of adherens junctions and accumulation of actin and myosin at the wound edge and are necessary for wound closure. We show that, in Drosophila, ROS drive wound healing in part through an ortholog of Src kinase, Src42A, which we identify as a redox sensor that promotes polarization of junctions and the cytoskeleton around wounds. We propose that ROS are a reparative signal that drives rapid embryonic wound healing in vertebrate and invertebrate species.


Subject(s)
Cell Polarity/physiology , Reactive Oxygen Species/metabolism , Wound Healing/physiology , Actins/metabolism , Actomyosin/metabolism , Adherens Junctions/metabolism , Animals , Cadherins/metabolism , Cytoskeleton/metabolism , Drosophila Proteins/metabolism , Drosophila melanogaster/embryology , Drosophila melanogaster/metabolism , Embryo, Nonmammalian/metabolism , Intercellular Junctions/metabolism , Myosins/metabolism , Oxidative Stress/physiology , Proto-Oncogene Proteins pp60(c-src)/metabolism , Zebrafish/embryology , Zebrafish/metabolism
10.
Curr Biol ; 27(15): 2260-2270.e5, 2017 Aug 07.
Article in English | MEDLINE | ID: mdl-28736167

ABSTRACT

In response to a pulling force, a material can elongate, hold fast, or fracture. During animal development, multi-cellular contraction of one region often stretches neighboring tissue. Such local contraction occurs by induced actomyosin activity, but molecular mechanisms are unknown for regulating the physical properties of connected tissue for elongation under stress. We show that cytohesins, and their Arf small G protein guanine nucleotide exchange activity, are required for tissues to elongate under stress during both Drosophila dorsal closure (DC) and zebrafish epiboly. In Drosophila, protein localization, laser ablation, and genetic interaction studies indicate that the cytohesin Steppke reduces tissue tension by inhibiting actomyosin activity at adherens junctions. Without Steppke, embryogenesis fails, with epidermal distortions and tears resulting from myosin misregulation. Remarkably, actomyosin network assembly is necessary and sufficient for local Steppke accumulation, where live imaging shows Steppke recruitment within minutes. This rapid negative feedback loop provides a molecular mechanism for attenuating the main tension generator of animal tissues. Such attenuation relaxes tissues and allows orderly elongation under stress.


Subject(s)
Drosophila Proteins/genetics , Drosophila melanogaster/genetics , GTP-Binding Protein Regulators/genetics , Guanine Nucleotide Exchange Factors/genetics , Signal Transduction , Zebrafish Proteins/genetics , Zebrafish/genetics , Animals , Drosophila Proteins/metabolism , Drosophila melanogaster/embryology , Drosophila melanogaster/metabolism , Embryo, Nonmammalian/embryology , Embryo, Nonmammalian/metabolism , GTP-Binding Protein Regulators/metabolism , Guanine Nucleotide Exchange Factors/metabolism , Zebrafish/embryology , Zebrafish/metabolism , Zebrafish Proteins/metabolism
11.
Dev Dyn ; 245(3): 244-58, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26434660

ABSTRACT

Tissue thinning and spreading, a morphogenetic movement termed epiboly, is used widely during animal development. In zebrafish, epiboly is a prominent cell movement during gastrulation, whereby a squamous epithelium (the enveloping layer), a multi-layer of loosely packed cells (the deep cells), and a yolk nuclear syncytium (the yolk syncytial layer) undergo coordinated expansion to engulf the yolk and close the blastopore. Elucidating the mechanisms that underlie epiboly is important not only for understanding animal development in general, but also for providing insights into fundamental cell behaviors including cell intercalation, cell adhesion, cell signaling, and epithelial morphogenesis. Here, recent work is reviewed with a focus on findings that advance our understanding of (1) the role of actomyosin motors in the yolk cell to drive epiboly, (2) the mechanisms that underlie the spreading of the epithelial enveloping layer, and (3) the regulation of deep cell movements by E-cadherin based adhesion. A discussion of how these new insights add to the current view of epiboly and future prospects is also presented. Overall, the study of zebrafish epiboly can provide general and broadly applicable insights into the genetic, molecular, and cellular control of morphogenesis.


Subject(s)
Cell Movement/physiology , Embryo, Nonmammalian/embryology , Morphogenesis/physiology , Zebrafish Proteins/metabolism , Zebrafish/embryology , Animals
12.
J Cell Biol ; 208(6): 839-56, 2015 Mar 16.
Article in English | MEDLINE | ID: mdl-25778923

ABSTRACT

Cleft-like boundaries represent a type of cell sorting boundary characterized by the presence of a physical gap between tissues. We studied the cleft-like ectoderm-mesoderm boundary in Xenopus laevis and zebrafish gastrulae. We identified the transcription factor Snail1 as being essential for tissue separation, showed that its expression in the mesoderm depends on noncanonical Wnt signaling, and demonstrated that it enables paraxial protocadherin (PAPC) to promote tissue separation through two novel functions. First, PAPC attenuates planar cell polarity signaling at the ectoderm-mesoderm boundary to lower cell adhesion and facilitate cleft formation. Second, PAPC controls formation of a distinct type of adhesive contact between mesoderm and ectoderm cells that shows properties of a cleft-like boundary at the single-cell level. It consists of short stretches of adherens junction-like contacts inserted between intermediate-sized contacts and large intercellular gaps. These roles of PAPC constitute a self/non-self-recognition mechanism that determines the site of boundary formation at the interface between PAPC-expressing and -nonexpressing cells.


Subject(s)
Cadherins/physiology , Transcription Factors/physiology , Xenopus Proteins/physiology , Actins/metabolism , Animals , Body Patterning , Cell Adhesion , Cell Polarity , Gastrula/embryology , Gastrula/metabolism , Mesoderm/cytology , Mesoderm/metabolism , Protocadherins , Receptors, G-Protein-Coupled/metabolism , Xenopus Proteins/metabolism , Xenopus laevis , Zebrafish , Zebrafish Proteins/physiology
13.
BMC Biol ; 12: 81, 2014 Oct 03.
Article in English | MEDLINE | ID: mdl-25277163

ABSTRACT

BACKGROUND: Nodal signalling is an absolute requirement for normal mesoderm and endoderm formation in vertebrate embryos, yet the transcriptional networks acting directly downstream of Nodal and the extent to which they are conserved is largely unexplored, particularly in vivo. Eomesodermin also plays a role in patterning mesoderm and endoderm in vertebrates, but its mechanisms of action, and how it interacts with the Nodal signalling pathway are still unclear. RESULTS: Using a combination of ChIP-seq and expression analysis we identify direct targets of Smad2, the effector of Nodal signalling in blastula stage zebrafish embryos, including many novel target genes. Through comparison of these data with published ChIP-seq data in human, mouse and Xenopus we show that the transcriptional network driven by Smad2 in mesoderm and endoderm is conserved in these vertebrate species. We also show that Smad2 and zebrafish Eomesodermin a (Eomesa) bind common genomic regions proximal to genes involved in mesoderm and endoderm formation, suggesting Eomesa forms a general component of the Smad2 signalling complex in zebrafish. Combinatorial perturbation of Eomesa and Smad2-interacting factor Foxh1 results in loss of both mesoderm and endoderm markers, confirming the role of Eomesa in endoderm formation and its functional interaction with Foxh1 for correct Nodal signalling. Finally, we uncover a novel, role for Eomesa in repressing ectodermal genes in the early blastula. CONCLUSION: Our data demonstrate that evolutionarily conserved developmental functions of Nodal signalling occur through maintenance of the transcriptional network directed by Smad2. This network is modulated by Eomesa in zebrafish which acts to promote mesoderm and endoderm formation in combination with Nodal signalling, whilst Eomesa also opposes ectoderm gene expression. Eomesa therefore regulates the formation of all three germ layers in the early zebrafish embryo.


Subject(s)
Body Patterning , Gene Expression Regulation, Developmental , Smad2 Protein/genetics , T-Box Domain Proteins/genetics , Zebrafish Proteins/genetics , Zebrafish/genetics , Animals , Ectoderm/embryology , Ectoderm/metabolism , Embryo, Nonmammalian/embryology , Embryo, Nonmammalian/metabolism , Endoderm/embryology , Endoderm/metabolism , Gene Regulatory Networks , Mesoderm/embryology , Mesoderm/metabolism , Signal Transduction , Smad2 Protein/metabolism , T-Box Domain Proteins/metabolism , Zebrafish/embryology , Zebrafish/metabolism , Zebrafish Proteins/metabolism
14.
Bioarchitecture ; 4(1): 31-4, 2014.
Article in English | MEDLINE | ID: mdl-24522009

ABSTRACT

Epiboly, the thinning and spreading of one tissue over another, is a widely employed morphogenetic movement that is essential for the development of many organisms. In the zebrafish embryo, epiboly describes the coordinated vegetal movement of the deep cells, enveloping layer (EVL) and yolk syncytial layer (YSL) to engulf the yolk cell. Recently, we showed that the large GTPase Dynamin plays a fundamental role in epiboly in the early zebrafish embryo. Because Dynamin plays a well-described role in vesicle scission during endocytosis, we predicted that Dynamin might regulate epiboly through participating in bulk removal of the yolk cell membrane ahead of the advancing margin, a proposed part of the epiboly motor. Unexpectedly, we found that Dynamin function was dispensable in the yolk cell and instead, it was required to maintain the epithelial integrity of the EVL during epiboly. Here, we present a model describing the maintenance of EVL integrity, which is required for the proper generation and transmission of tension during epiboly. Furthermore, we discuss the role of Dynamin-mediated regulation of ezrin-radixin-moesin (ERM) family proteins in the maintenance of epithelial integrity.


Subject(s)
Actomyosin/metabolism , Blastoderm/embryology , Dynamins/metabolism , Zebrafish/embryology , Animals
15.
Dev Biol ; 385(1): 52-66, 2014 Jan 01.
Article in English | MEDLINE | ID: mdl-24161849

ABSTRACT

Epiboly, the first morphogenetic cell movement that occurs in the zebrafish embryo, is the process by which the blastoderm thins and spreads to engulf the yolk cell. This process requires the concerted actions of the deep cells, the enveloping layer (EVL) and the extra-embryonic yolk syncytial layer (YSL). The EVL is mechanically coupled to the YSL which acts as an epiboly motor, generating the force necessary to draw the blastoderm towards the vegetal pole though actomyosin flow and contraction of the actomyosin ring. However, it has been proposed that the endocytic removal of yolk cell membrane just ahead of the advancing blastoderm may also play a role. To assess the contribution of yolk cell endocytosis in driving epiboly movements, we used a combination of drug- and dominant-negative-based approaches to inhibit Dynamin, a large GTPase with a well-characterized role in vesicle scission. We show that Dynamin-dependent endocytosis in the yolk cell is dispensable for epiboly of the blastoderm. However, global inhibition of Dynamin function revealed that Dynamin plays a fundamental role within the blastoderm during epiboly, where it maintains epithelial integrity and the transmission of tension across the EVL. The epithelial defects were associated with disrupted tight junctions and a striking reduction of cortically localized phosphorylated ezrin/radixin/moesin (P-ERM), key regulators of epithelial integrity in other systems. Furthermore, we show that Dynamin maintains EVL and promotes epiboly progression by antagonizing Rho A activity.


Subject(s)
Actomyosin/metabolism , Blastoderm/embryology , Dynamins/metabolism , Zebrafish/embryology , Adherens Junctions/metabolism , Animals , Blastoderm/growth & development , Cell Differentiation , Cytoskeletal Proteins/metabolism , Dynamins/genetics , Embryo, Nonmammalian/metabolism , Endocytosis , Membrane Proteins/metabolism , Microfilament Proteins/metabolism , Morphogenesis , Phosphorylation , Yolk Sac , Zebrafish/genetics , rho-Associated Kinases/antagonists & inhibitors
16.
Int J Dev Biol ; 54(8-9): 1213-28, 2010.
Article in English | MEDLINE | ID: mdl-20712002

ABSTRACT

Gastrulation involves of a series of coordinated cell movements to organize the germ layers and establish the major body axes of the embryo. One gastrulation movement is epiboly, which involves the thinning and spreading of a multilayered cell sheet. Epiboly plays a prominent role in zebrafish gastrulation and studies of zebrafish epiboly have provided insights into basic cellular properties and mechanisms of morphogenesis that are widely used in animal development. Although considerable progress has been made in identifying molecules that are required for epiboly, we still understand very little about how these factors cooperate to drive the process. Here, we review work on the molecular and cellular basis of zebrafish epiboly in order to identify unifying themes and to highlight some of the current open questions.


Subject(s)
Gastrula/embryology , Gastrulation , Zebrafish/embryology , Animals , Gastrula/cytology , Gastrula/metabolism , Gene Expression Regulation, Developmental , Microtubules/metabolism , Signal Transduction/genetics , Zebrafish/genetics , Zebrafish/metabolism , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism
17.
Dev Dyn ; 239(2): 715-22, 2010 Feb.
Article in English | MEDLINE | ID: mdl-20014098

ABSTRACT

Zebrafish epiboly results in the thinning and spreading of the blastoderm to cover the yolk cell and close the blastopore. The extra-embryonic yolk syncytial layer (YSL) tows the blastoderm vegetally during epiboly by means of its tight junction attachments to the enveloping layer (EVL). Claudins are the major transmembrane protein components of tight junctions. Here, we focus on the function of Claudin E (Cldne), which is expressed specifically in the EVL. Morpholino knock-down of cldne produced a highly penetrant epiboly delay. Our analysis suggested that the EVL margin, which is attached to the YSL, was under reduced tension in morphant embryos. We propose that local variation in the strength of EVL-YSL attachment in morphant embryos resulted in slow and uneven advancement of the EVL and blastoderm. Our work is the first to demonstrate that Claudins are important for zebrafish epiboly.


Subject(s)
Claudins/metabolism , Gastrulation , Tight Junctions/metabolism , Zebrafish/embryology , Animals , Gene Knockdown Techniques , Zebrafish/metabolism
18.
Development ; 136(10): 1675-85, 2009 May.
Article in English | MEDLINE | ID: mdl-19369398

ABSTRACT

The organizer is essential for dorsal-ventral (DV) patterning in vertebrates. Goosecoid (Gsc), a transcriptional repressor found in the organizer, elicits partial secondary axes when expressed ventrally in Xenopus, similar to an organizer transplant. Although gsc is expressed in all vertebrate organizers examined, knockout studies in mouse suggested that it is not required for DV patterning. Moreover, experiments in Xenopus and zebrafish suggest a role in head formation, although a function in axial mesoderm formation is less clear. To clarify the role of Gsc in vertebrate development, we used gain- and loss-of-function approaches in zebrafish. Ventral injection of low doses of gsc produced incomplete secondary axes, which we propose results from short-range repression of BMP signaling. Higher gsc doses resulted in complete secondary axes and long-range signaling, correlating with repression of BMP and Wnt signals. In striking contrast to Xenopus, the BMP inhibitor Chordin (Chd) is not required for Gsc function. Gsc produced complete secondary axes in chd null mutant embryos and gsc-morpholino knockdown in chd mutants enhanced the mutant phenotype, suggesting that Gsc has Chd-independent functions in DV patterning. Even more striking was that Gsc elicited complete secondary axes in the absence of three secreted BMP antagonists, Chd, Follistatin-like 1b and Noggin 1, suggesting that Gsc functions in parallel with secreted BMP inhibitors. Our findings suggest that Gsc has dose dependent effects on axis induction and provide new insights into molecularly distinct short- and long-range signaling activities of the organizer.


Subject(s)
Carrier Proteins/physiology , Follistatin-Related Proteins/physiology , Glycoproteins/physiology , Goosecoid Protein/physiology , Intercellular Signaling Peptides and Proteins/physiology , Zebrafish Proteins/physiology , Zebrafish/embryology , Animals , Animals, Genetically Modified , Body Patterning/physiology , Bone Morphogenetic Proteins/physiology , Embryo, Nonmammalian/physiology , Glycoproteins/genetics , Goosecoid Protein/genetics , Intercellular Signaling Peptides and Proteins/genetics , Mutation , Receptors, Notch/physiology , Signal Transduction/physiology , Zebrafish/physiology , Zebrafish Proteins/genetics
19.
Dev Dyn ; 237(3): 819-29, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18265014

ABSTRACT

The classic calpain system has been implicated in regulating a variety of cellular processes including cell adhesion, migration, and intracellular signaling; however, little is known regarding the function of this system in vivo. Two heterodimeric Ca(2+)-dependent cysteine proteases, mu-calpain (CAPN1) and m-calpain (CAPN2), and the endogenous inhibitor calpastatin (CAST) comprise the classic/ubiquitous calpain system in mammals. Recently, knockout of two murine classic calpain genes, Capn2 and Capn4/Capns1, revealed that components of the classic system are indispensable for preimplantation development. We identified four classic calpain catalytic subunit genes (capn1a, 1b, 2a, 2b), two regulatory subunit genes (capns1a, 1b), and calpastatin (cast) from the zebrafish. Our data suggest that the components of the classic mammalian system are both conserved and expanded in the teleost lineage. In contrast to the classic/ubiquitous mammalian system, zebrafish calpain system genes acquire unique, tissue-specific patterns of expression within the first 2 days of development.


Subject(s)
Calpain/genetics , Embryo, Nonmammalian/enzymology , Gene Expression Regulation, Developmental , Zebrafish/embryology , Animals , Calpain/metabolism , Catalytic Domain/genetics , Embryo, Nonmammalian/embryology , Evolution, Molecular , Phylogeny , Zebrafish/genetics , Zebrafish/metabolism
20.
Dev Biol ; 289(2): 482-93, 2006 Jan 15.
Article in English | MEDLINE | ID: mdl-16316643

ABSTRACT

To further our understanding of FOG gene function during cardiac development, we utilized zebrafish to examine FOG's role in the early steps of heart morphogenesis. We identified fragments of three fog genes in the zebrafish genomic database and isolated full-length coding sequences for each of these genes by using a combination of RT-PCR and 5'-RACE. One gene was similar to murine FOG-1 (fog1), while the remaining two were similar to murine FOG-2 (fog2a and fog2b). All Fog proteins were able to physically interact with GATA4 and function as transcriptional co-repressors. Whole-mount in situ hybridization revealed fog1 expression in the heart, the hematopoietic system, and the brain, while fog2a and fog2b expression was restricted to the brain. Injection of zebrafish embryos with a morpholino directed against fog1 resulted in embryos with a large pericardial effusion and an unlooped heart tube. This looping defect could be rescued by co-injection of mRNA encoding murine FOG-1, but not by mRNA encoding FOG-1 lacking the FOG repression motif. Taken together, these results demonstrate the importance of FOG proteins for zebrafish cardiac development and suggest a previously unappreciated role for FOG proteins in heart looping that is dependent on the FOG repression motif.


Subject(s)
Heart/embryology , Transcription Factors/genetics , Zebrafish Proteins/genetics , Zebrafish/metabolism , Animals , Base Sequence , Brain/embryology , Brain/metabolism , Conserved Sequence , GATA4 Transcription Factor/metabolism , Heart/physiology , Mice , Molecular Sequence Data , Mutation , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Protein Isoforms/genetics , Protein Isoforms/metabolism , Sequence Homology , Transcription Factors/metabolism , Transfection , Zebrafish/embryology , Zebrafish/genetics , Zebrafish Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...