Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Dev Neurobiol ; 75(9): 961-83, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25556989

ABSTRACT

MYO7A is an unconventional myosin involved in the structural organization of hair bundles at the apex of sensory hair cells (SHCs) where it serves mechanotransduction in the process of hearing and balance. Mutations of MYO7A are responsible for abnormal shaping of hair bundles, resulting in human deafness and murine deafness/circling behavior. Myo7aa, expressed in SHCs of the inner ear and lateral line of zebrafish, causes circling behavior and abnormal hair cell function when deficient in mariner mutant. This work identifies a new hair cell-specific enhancer, highly conserved between species, located within Intron 2-3 of zebrafish myosin 7a (myo7aa) gene. This enhancer is contained within a 761-bp DNA fragment that encompasses a newly identified Exon of myo7aa and whose activity does not depend on orientation. Compensation of mariner mutation by expression of mCherry-Myo7aa fusion protein under the control of this 761-bp DNA fragment results in recovery of balance, normal hair bundle shape and restored hair cell function. Two smaller adjacent fragments (344-bp and 431-bp), extracted from the 761-bp fragment, both show hair cell-specific enhancing activity, with apparently reduced intensity and coverage. These data should help understand the role of Myo7aa in sensory hair cell differentiation and function. They provide tools to decipher how myo7aa gene is expressed and regulated in SHCs by allowing the identification of potential transcription factors involved in this process. The discovered enhancer could represent a new target for the identification of deafness-causing mutations affecting human MYO7A.


Subject(s)
Enhancer Elements, Genetic , Exons , Hair Cells, Auditory/metabolism , Lateral Line System/cytology , Mechanoreceptors/metabolism , Myosins/genetics , Myosins/metabolism , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism , Animals , Animals, Genetically Modified , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Introns , Lateral Line System/embryology , Lateral Line System/metabolism , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Molecular Sequence Data , Mutation , Postural Balance/physiology , RNA, Messenger/metabolism , Reflex, Startle/physiology , Sequence Homology , Zebrafish , Red Fluorescent Protein
2.
Development ; 140(5): 1090-9, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23364327

ABSTRACT

Endoderm and mesoderm are both formed upon activation of Nodal signaling but how endoderm differentiates from mesoderm is still poorly explored. The sox-related gene casanova (sox32) acts downstream of the Nodal signal, is essential for endoderm development and requires the co-factor Pou2 (Pou5f1, Oct3, Oct4) in this process. Conversely, BMP signals have been shown to inhibit endoderm development by an as yet unexplained mechanism. In a search for Casanova regulators in zebrafish, we identified two of its binding partners as the transcription factors Pou2 and Vox, a member of the Vent group of proteins also involved in the patterning of the gastrula. In overexpression studies we show that vox and/or Vent group genes inhibit the capacity of Casanova to induce endoderm, even in the presence of its co-factor Pou2, and that Vox acts as a repressor in this process. We further show that vox, but not other members of the Vent group, is essential for defining the proper endodermal domain size at gastrulation. In this process, vox acts downstream of BMPs. Cell fate analysis further shows that Vox plays a key role downstream of BMP signals in regulating the capacity of Nodal to induce endoderm versus mesoderm by modulating the activity of the Casanova/Pou2 regulatory system.


Subject(s)
Endoderm/embryology , Homeodomain Proteins/metabolism , Homeodomain Proteins/physiology , Octamer Transcription Factor-3/metabolism , Repressor Proteins/metabolism , Repressor Proteins/physiology , SOX Transcription Factors/metabolism , Zebrafish Proteins/metabolism , Zebrafish Proteins/physiology , Animals , Animals, Genetically Modified , Bone Morphogenetic Proteins/genetics , Bone Morphogenetic Proteins/metabolism , Bone Morphogenetic Proteins/physiology , Down-Regulation/genetics , Embryo, Nonmammalian , Endoderm/growth & development , Endoderm/metabolism , Gene Expression Regulation, Developmental , Homeodomain Proteins/chemistry , Homeodomain Proteins/genetics , Nodal Signaling Ligands/genetics , Nodal Signaling Ligands/metabolism , Nodal Signaling Ligands/physiology , Octamer Transcription Factor-3/physiology , Protein Binding/physiology , Protein Interaction Domains and Motifs/genetics , Protein Interaction Domains and Motifs/physiology , Repressor Proteins/chemistry , Repressor Proteins/genetics , SOX Transcription Factors/physiology , Sequence Deletion , Tissue Distribution , Transcription Factors/genetics , Transcription Factors/metabolism , Transcription Factors/physiology , Zebrafish/embryology , Zebrafish/genetics , Zebrafish/metabolism , Zebrafish Proteins/chemistry , Zebrafish Proteins/genetics
3.
Proc Natl Acad Sci U S A ; 109(42): 16945-50, 2012 Oct 16.
Article in English | MEDLINE | ID: mdl-23027928

ABSTRACT

Collective cell migration is key to morphogenesis, wound healing, or cancer cell migration. However, its cellular bases are just starting to be unraveled. During vertebrate gastrulation, axial mesendoderm migrates in a group, the prechordal plate, from the embryonic organizer to the animal pole. How this collective migration is achieved remains unclear. Previous work has suggested that cells migrate as individuals, with collective movement resulting from the addition of similar individual cell behavior. Through extensive analyses of cell trajectories, morphologies, and polarization in zebrafish embryos, we reveal that all prechordal plate cells show the same behavior and rely on the same signaling pathway to migrate, as expected if they do so individually. However, by using cell transplants, we demonstrate that prechordal plate migration is a true collective process, as isolated cells do not migrate toward the animal pole. They are still polarized and motile but lose directionality. Directionality is restored upon contact with the endogenous prechordal plate. This contact dependent orientation relies on E-cadherin, Wnt-PCP signaling, and Rac1. Importantly, groups of cells also need contact with the endogenous plate to orient correctly, showing an instructive role of the plate in establishing directionality. Overall, our results lead to an original model of collective migration in which directional information is contained within the moving group rather than provided by extrinsic cues, and constantly maintained in cells by contacts with their neighbors. This self-organizing model could account for collective invasion of new territories, as observed in cancer strands, without requirement for any attractant in the colonized tissue.


Subject(s)
Cell Movement/physiology , Endoderm/physiology , Mesoderm/physiology , Morphogenesis/physiology , Signal Transduction/physiology , Animals , Cadherins/metabolism , Cell Polarity/physiology , Endoderm/cytology , In Situ Hybridization , Mesoderm/cytology , Time-Lapse Imaging , Wnt Signaling Pathway/physiology , Zebrafish
4.
Development ; 138(16): 3473-84, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21791527

ABSTRACT

Taste buds, the taste sensory organs, are conserved in vertebrates and composed of distinct cell types, including taste receptor, basal/presynaptic and support cells. Here, we characterize zebrafish taste bud development and show that compromised Fgf signaling in the larva results in taste bud reduction and disorganization. We determine that Fgf activity is required within pharyngeal endoderm for formation of Calb2b(+) cells and reveal miR-200 and Delta-Notch signaling as key factors in this process. miR-200 knock down shows that miR-200 activity is required for taste bud formation and in particular for Calb2b(+) cell formation. Compromised delta activity in mib(-/-) dramatically reduces the number of Calb2b(+) cells and increases the number of 5HT(+) cells. Conversely, larvae with increased Notch activity and ascl1a(-/-) mutants are devoid of 5HT(+) cells, but have maintained and increased Calb2b(+) cells, respectively. These results show that Delta-Notch signaling is required for intact taste bud organ formation. Consistent with this, Notch activity restores Calb2b(+) cell formation in pharyngeal endoderm with compromised Fgf signaling, but fails to restore the formation of these cells after miR-200 knock down. Altogether, this study provides genetic evidence that supports a novel model where Fgf regulates Delta-Notch signaling, and subsequently miR-200 activity, in order to promote taste bud cell type differentiation.


Subject(s)
MicroRNAs/genetics , Signal Transduction , Taste Buds/embryology , Taste Buds/metabolism , Zebrafish/embryology , Zebrafish/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Fibroblast Growth Factors/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , Receptors, Notch/metabolism , Taste Buds/growth & development , Transcription Factors , Zebrafish/growth & development , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism
5.
Development ; 138(9): 1783-94, 2011 May.
Article in English | MEDLINE | ID: mdl-21447554

ABSTRACT

The development of the different muscles within the somite is a complex process that involves the Hedgehog (Hh) signaling pathway. To specify the proper number of muscle cells and organize them spatially and temporally, the Hh signaling pathway needs to be precisely regulated at different levels, but only a few factors external to the pathway have been described. Here, we report for the first time the role of the STAR family RNA-binding protein Quaking A (QkA) in somite muscle development. We show in zebrafish that the loss of QkA function affects fast muscle fiber maturation as well as Hh-induced muscle derivative specification and/or morphogenesis. Mosaic analysis reveals that fast fiber maturation depends on the activity of QkA in the environment of fast fiber progenitors. We further show that Hh signaling requires QkA activity for muscle development. By an in silico approach, we screened the 3'UTRs of known Hh signaling component mRNAs for the Quaking response element and found the transcription factor Gli2a, a known regulator of muscle fate development. Using destabilized GFP as a reporter, we show that the gli2a mRNA 3'UTR is a functional QkA target. Consistent with this notion, the loss of QkA function rescued slow muscle fibers in yot mutant embryos, which express a dominant-negative Gli2a isoform. Thus, our results reveal a new mechanism to ensure muscle cell fate diversity by fine-tuning of the Hh signaling pathway via RNA-binding proteins.


Subject(s)
Hedgehog Proteins/physiology , Muscle Development/genetics , RNA-Binding Proteins/physiology , Zebrafish Proteins/physiology , Animals , Animals, Genetically Modified , Body Patterning/genetics , Body Patterning/physiology , Chromosome Mapping , Embryo, Nonmammalian , Genes, Recessive , Hedgehog Proteins/genetics , Hedgehog Proteins/metabolism , Morphogenesis/genetics , Morphogenesis/physiology , Muscle Development/physiology , Muscle Fibers, Fast-Twitch/metabolism , Muscle Fibers, Fast-Twitch/physiology , Muscle Fibers, Slow-Twitch/metabolism , Muscle Fibers, Slow-Twitch/physiology , Mutation/physiology , RNA-Binding Proteins/genetics , Signal Transduction/genetics , Signal Transduction/physiology , Zebrafish/embryology , Zebrafish/genetics , Zebrafish/metabolism , Zebrafish Proteins/genetics
6.
Curr Biol ; 18(4): 276-81, 2008 Feb 26.
Article in English | MEDLINE | ID: mdl-18291651

ABSTRACT

During gastrulation, dramatic movements rearrange cells into three germ layers expanded over the entire embryo [1-3]. In fish, both endoderm and mesoderm are specified as a belt at the embryo margin. Mesodermal layer expansion is achieved through the combination of two directed migrations. The outer ring of precursors moves toward the vegetal pole and continuously seeds mesodermal cells inside the embryo, which then reverse their movement in the direction of the animal pole [3-6]. Unlike mesoderm, endodermal cells internalize at once and must therefore adopt a different strategy to expand over the embryo [7, 8]. With live imaging of YFP-expressing zebrafish endodermal cells, we demonstrate that in contrast to mesoderm, internalized endodermal cells display a nonoriented/noncoordinated movement fit by a random walk that rapidly disperses them over the yolk surface. Transplantation experiments reveal that this behaviour is largely cell autonomous, induced by TGF-beta/Nodal, and dependent on the downstream effector Casanova. At midgastrulation, endodermal cells switch to a convergence movement. We demonstrate that this switch is triggered by environmental cues. These results uncover random walk as a novel Nodal-induced gastrulation movement and as an efficient strategy to transform a localized cell group into a layer expanded over the embryo.


Subject(s)
Cell Movement/physiology , Endoderm/cytology , Gastrulation/physiology , Zebrafish/embryology , Animals , Animals, Genetically Modified/embryology , Animals, Genetically Modified/physiology , Embryonic Induction/physiology , Endoderm/physiology , High Mobility Group Proteins/metabolism , Nodal Protein , SOX Transcription Factors , Transcription Factors/metabolism , Transforming Growth Factor beta/metabolism , Zebrafish/physiology , Zebrafish Proteins/metabolism
7.
PLoS One ; 3(1): e1434, 2008 Jan 16.
Article in English | MEDLINE | ID: mdl-18197245

ABSTRACT

The EGF-CFC factor Oep/Cripto1/Frl1 has been implicated in embryogenesis and several human cancers. During vertebrate development, Oep/Cripto1/Frl1 has been shown to act as an essential coreceptor in the TGFbeta/Nodal pathway, which is crucial for germ layer formation. Although studies in cell cultures suggest that Oep/Cripto1/Frl1 is also implicated in other pathways, in vivo it is solely regarded as a Nodal coreceptor. We have found that Rasl11b, a small GTPase belonging to a Ras subfamily of putative tumor suppressor genes, modulates Oep function in zebrafish independently of the Nodal pathway. rasl11b down regulation partially rescues endodermal and prechordal plate defects of zygotic oep(-/-) mutants (Zoep). Rasl11b inhibitory action was only observed in oep-deficient backgrounds, suggesting that normal oep expression prevents Rasl11b function. Surprisingly, rasl11b down regulation does not rescue mesendodermal defects in other Nodal pathway mutants, nor does it influence the phosphorylation state of the downstream effector Smad2. Thus, Rasl11b modifies the effect of Oep on mesendoderm development independently of the main known Oep output: the Nodal signaling pathway. This data suggests a new branch of Oep signaling that has implications for germ layer development, as well as for studies of Oep/Frl1/Cripto1 dysfunction, such as that found in tumors.


Subject(s)
Homeodomain Proteins/genetics , Monomeric GTP-Binding Proteins/genetics , Mutation , Transcription Factors/genetics , Zebrafish Proteins/genetics , Animals , Base Sequence , DNA Primers , Mutagenesis , Phenotype , Phosphorylation , Reverse Transcriptase Polymerase Chain Reaction , Smad2 Protein/metabolism , Zebrafish
8.
Gene Expr Patterns ; 6(8): 835-42, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16545988

ABSTRACT

The secreted frizzled-related proteins (Sfrp) are a family of soluble proteins with diverse biological functions having the capacity to bind Wnt ligands, to modulate Wnt signalling, and to signal directly via the Wnt receptor, Frizzled. In an enhancer trap screen for embryonic expression in zebrafish we identified an sfrp1 gene. Previous studies suggest an important role for sfrp1 in eye development, however, no data have been reported using the zebrafish model. In this paper, we describe duplicate sfrp1 genes in zebrafish and present a detailed analysis of the expression profile of both genes. Whole mount in situ hybridisation analyses of sfrp1a during embryonic and larval development revealed a dynamic expression profile, including: the central nervous system, where sfrp1a was regionally expressed throughout the brain and developing eye; the posterior gut, from the time of endodermal cell condensation; the lateral line, where sfrp1a was expressed in the migrating primordia and interneuromast cells that give rise to the sensory organs. Other sites included the blastoderm, segmenting mesoderm, olfactory placode, developing ear, pronephros and fin-bud. We have also analysed sfrp1b expression during embryonic development. Surprisingly this gene exhibited a divergent expression profile being limited to the yolk syncytium under the elongating tail-bud, which later covered the distal yolk extension, and transiently in the tail-bud mesenchyme. Overall, our studies provide a basis for future analyses of these developmentally important factors using the zebrafish model.


Subject(s)
Central Nervous System/embryology , Central Nervous System/metabolism , Gastrula/metabolism , Proteins/genetics , Zebrafish Proteins/metabolism , Zebrafish/embryology , Amino Acid Sequence , Animals , Cleavage Stage, Ovum/metabolism , Embryo, Nonmammalian , Eye/embryology , Eye/metabolism , Gene Duplication , Gene Expression Regulation, Developmental , Intracellular Signaling Peptides and Proteins , Mesoderm/metabolism , Molecular Sequence Data , Phylogeny , Proteins/metabolism , Sequence Homology, Amino Acid , Zebrafish/genetics , Zebrafish/metabolism , Zebrafish Proteins/genetics
9.
Med Sci (Paris) ; 20(5): 557-61, 2004 May.
Article in French | MEDLINE | ID: mdl-15190475

ABSTRACT

Endothelial cells (EC) of the vertebrate cardiovascular system (CVS) are bona fide, yet enigmatic mechanoreceptors. When cultured in vitro and exposed to fluid forces, EC modify their physiological behaviour at the structural, metabolical and gene expression levels in response to the mechanical stimulus. However, as a direct consequence of the hypoxic bias (and often the lethality) that results from blocking blood flow in most animal systems, the in vivo role of EC mechanosensation (ECMS) remains poorly understood. The zebrafish has recently emerged as an alternative genetic model for the study of vertebrate development. Its striking ability to survive until larval stages in the absence of blood circulation circumveys the usual caveats that are inherent to CVS research, and offers the exciting opportunity to dissect the function of ECMS in vivo. Two groups have already uncovered an essential role for ECMS in zebrafish organogenesis, particularly in heart morphogenesis. In embryos in which intracardiac blood flow is genetically or physically compromised, several features of the normally developing heart, including valve formation, are specifically disrupted. In addition, impressive imaging studies of zebrafish hemodynamics demonstrate that the shear stress exerted upon the cardiac endothelium is largely in the range of the stimulus that in vitro activates cytoskelettal remodeling and gene expression changes in EC. Hence the cardiac phenotypes observed in vivo may indeed directly result from a lack of ECMS-dependent EC activity. These data shed first light on the role of ECMS in vivo. Notably, they also suggest that a number of human congenital cardiomyopathies may arise through abnormal fetal hemodynamics and/or EC sensory activity. Finally, these discoveries reinforce the too often neglected role of epigenetic factors (in this case, fluid forces) in the regulation of animal development.


Subject(s)
Hemodynamics , Mechanotransduction, Cellular/physiology , Organogenesis/physiology , Animals , Endothelium, Vascular/cytology , Endothelium, Vascular/physiology , Heart/embryology , Kidney/enzymology
10.
Development ; 131(3): 629-41, 2004 Feb.
Article in English | MEDLINE | ID: mdl-14711879

ABSTRACT

Interactions between Nodal/Activin and Fibroblast growth factor (Fgf) signalling pathways have long been thought to play an important role in mesoderm formation. However, the molecular and cellular processes underlying these interactions have remained elusive. Here, we address the epistatic relationships between Nodal and Fgf pathways during early embryogenesis in zebrafish. First, we find that Fgf signalling is required downstream of Nodal signals for inducing the Nodal co-factor One-eyed-pinhead (Oep). Thus, Fgf is likely to be involved in the amplification and propagation of Nodal signalling during early embryonic stages. This could account for the previously described ability of Fgf to render cells competent to respond to Nodal/Activin signals. In addition, overexpression data shows that Fgf8 and Fgf3 can take part in this process. Second, combining zygotic mutations in ace/fgf8 and oep disrupts mesoderm formation, a phenotype that is not produced by either mutation alone and is consistent with our model of an interdependence of Fgf8 and Nodal pathways through the genetic regulation of the Nodal co-factor Oep and the cell propagation of Nodal signalling. Moreover, mesodermal cell populations are affected differentially by double loss-of-function of Zoep;ace. Most of the dorsal mesoderm undergoes massive cell death by the end of gastrulation, in contrast to either single-mutant phenotype. However, some mesoderm cells are still able to undergo myogenic differentiation in the anterior trunk of Zoep;ace embryos, revealing a morphological transition at the level of somites 6-8. Further decreasing Oep levels by removing maternal oep products aggravates the mesodermal defects in double mutants by disrupting the fate of the entire mesoderm. Together, these results demonstrate synergy between oep and fgf8 that operates with regional differences and is involved in the induction, maintenance, movement and survival of mesodermal cell populations.


Subject(s)
Embryo, Nonmammalian/metabolism , Fibroblast Growth Factors/metabolism , Mesoderm/metabolism , Transforming Growth Factor beta/metabolism , Up-Regulation/physiology , Zebrafish Proteins , Animals , Apoptosis/physiology , Body Patterning/physiology , Fibroblast Growth Factor 3 , Fibroblast Growth Factor 8 , Fibroblast Growth Factors/genetics , Gastrula/physiology , Nodal Protein , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Signal Transduction , Transforming Growth Factor beta/genetics , Zebrafish/embryology , Zebrafish/metabolism
11.
Proc Natl Acad Sci U S A ; 100(6): 3315-20, 2003 Mar 18.
Article in English | MEDLINE | ID: mdl-12601179

ABSTRACT

In vertebrate embryos, maternal determinants are thought to preestablish the dorsoventral axis by locally activating zygotic ventral- and dorsal-specifying genes, e.g., genes encoding bone morphogenetic proteins (BMPs) and BMP inhibitors, respectively. Whereas the canonical Wntbeta-catenin pathway fulfills this role dorsally, the existence of a reciprocal maternal ventralizing signal remains hypothetical. Maternal noncanonical WntCa(2+) signaling may promote ventral fates by suppressing Wntbeta-catenin dorsalizing signals; however, whether any maternal determinant is directly required for the activation of zygotic ventral-specifying genes is unknown. Here, we show that such a function is achieved, in part, in the zebrafish embryo by the maternally encoded transforming growth factor beta (TGF-beta) signaling molecule, Radar. Loss-of-function experiments, together with epistasis analyses, identify maternal Radar as an upstream activator of bmps expression. Maternal induction of bmps by Radar is essential for zebrafish development as its removal results in larval-lethal dorsalized phenotypes. Double-morphant analyses further suggest that Radar functions through the TGF-beta receptor Alk8 to initiate the expression of bmp genes. Our results support the existence of a previously uncharacterized maternal ventralizing pathway. They might further indicate that maternal TGF-betaRdr and WntCa(2+) pathways complementarily specify ventral cell fates, with the former triggering bmps expression and the latter indirectly repressing genes encoding BMP antagonists.


Subject(s)
Bone Morphogenetic Proteins/genetics , Bone Morphogenetic Proteins/physiology , Zebrafish Proteins , Zebrafish/embryology , Zebrafish/genetics , Animals , Base Sequence , Body Patterning , Embryonic Induction , Female , Gene Expression Regulation, Developmental , Growth Differentiation Factor 6 , In Situ Hybridization , Mutation , Phenotype , RNA, Messenger/genetics , RNA, Messenger/metabolism
12.
Sci STKE ; 2002(158): pe47, 2002 Nov 12.
Article in English | MEDLINE | ID: mdl-12429886

ABSTRACT

Within a multicellular organism, communication between cells is essential during development to ensure proper execution of cell migration, cell fate decisions, and differentiation events. It is also essential in the adult for the coordination of many physiological functions. Cell-to-cell communications often rely on the interaction of cell surface receptors with soluble or membrane-bound ligands. Receptors or ligands may interact with additional partners to trigger specific signaling cascades inside the cell. In most cases, partners act in a specific configuration, either as a diffusible "co-ligand" or a membrane-bound co-receptor. Here, we examine the case of Cripto, a signaling molecule that has prominent functions during vertebrate development. Conflicting results have suggested that Cripto has the unusual capacity to act both as a secreted ligand and as a cell surface component to control a single signaling pathway. Here, we review the recent experiments that attempt to reconcile those results. Furthermore, three reports have described the fact that Cripto is modified by the addition of sugar residues, including a rare case of fucosylation. These modifications are essential for Cripto function, suggesting that, as is the case for other key developmental or physiological regulators such as Notch or selectins, the activity of Cripto may be controlled by the extent of its glycosylation or fucosylation (or both).


Subject(s)
Epidermal Growth Factor , Membrane Glycoproteins , Neoplasm Proteins/physiology , Signal Transduction/physiology , Animals , Biomarkers, Tumor/physiology , GPI-Linked Proteins , Growth Substances/physiology , Humans , Intercellular Signaling Peptides and Proteins
13.
Proc Natl Acad Sci U S A ; 99(25): 16297-302, 2002 Dec 10.
Article in English | MEDLINE | ID: mdl-12444253

ABSTRACT

Cell migration plays an essential role in many morphogenetic processes, and its deregulation has many dramatic consequences. Yet how migration is controlled during normal development is still a largely unresolved question. We examined this process in the case of the posterior lateral line (PLL), a mechanosensory system present in fish and amphibians. In zebrafish, the embryonic PLL comprises seven to eight sense organs (neuromasts) aligned from head to tail along the flank of the animal and is formed by a primordium that originates from a cephalic placode. This primordium migrates along a stereotyped pathway toward the tip of the tail and deposits in its wake discrete groups of cells, each of which will become a neuromast. We show that a trail of SDF1-like chemokine is present along the pathway of the primordium and that a CXCR4-like chemokine receptor is expressed by the migrating cells. The inactivation of either the ligand or its receptor blocks migration, whereas in mutants in which the normal SDF1 trail is absent, the primordium path is redirected to the next, more ventral sdf1 expression domain. In all cases, the sensory axons remain associated to the primordium, indicating that the extension of the neurites to form the PLL nerve depends on the movement of the primordium. We conclude that both the formation and the innervation of this system depend on the SDF1-CXCR4 system, which has also been implicated in several migration events in humans, including metastasis formation and lymphocyte homing.


Subject(s)
Chemokines, CXC/physiology , Neurons, Afferent/cytology , Receptors, CXCR4/physiology , Sense Organs/embryology , Zebrafish/genetics , Animals , Axons/ultrastructure , Cell Lineage , Cell Movement , Chemokine CXCL12 , Chemokines, CXC/analysis , Chemokines, CXC/genetics , Embryonic Induction , Neurites/ultrastructure , Receptors, CXCR4/analysis , Receptors, CXCR4/genetics , Sense Organs/innervation , Zebrafish/embryology
14.
Development ; 129(19): 4457-68, 2002 Oct.
Article in English | MEDLINE | ID: mdl-12223404

ABSTRACT

The vertebrate head skeleton is derived in part from neural crest cells, which physically interact with head ectoderm, mesoderm and endoderm to shape the pharyngeal arches. The cellular and molecular nature of these interactions is poorly understood, and we explore here the function of endoderm in this process. By genetic ablation and reintroduction of endoderm in zebrafish, we show that it is required for the development of chondrogenic neural crest cells, including their identity, survival and differentiation into arch cartilages. Using a genetic interference approach, we further identify Fgf3 as a critical component of endodermal function that allows the development of posterior arch cartilages. Together, our results reveal for the first time that the endoderm provides differential cues along the anteroposterior axis to control ventral head skeleton development and demonstrate that this function is mediated in part by Fgf3.


Subject(s)
Fibroblast Growth Factors/metabolism , Proto-Oncogene Proteins/metabolism , Signal Transduction , Skull/embryology , Zebrafish Proteins , Animals , Branchial Region/embryology , Cartilage, Articular/embryology , Cell Differentiation , Endoderm , Fibroblast Growth Factor 3 , Head/embryology , High Mobility Group Proteins/genetics , High Mobility Group Proteins/metabolism , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Morphogenesis , Neural Crest/cytology , SOX Transcription Factors , Transcription Factors/genetics , Transcription Factors/metabolism , Zebrafish/embryology , Zebrafish/metabolism
15.
Development ; 129(13): 3055-65, 2002 Jul.
Article in English | MEDLINE | ID: mdl-12070082

ABSTRACT

Despite its evolutionary conservation and functional importance, little is known of the signaling pathways that underlie development of the hypothalamus. Although mutations affecting Nodal and Hedgehog signaling disrupt hypothalamic development, the time and site of action and the exact roles of these pathways remain very poorly understood. Unexpectedly, we show here that cell-autonomous reception of Nodal signals is neither required for the migration of hypothalamic precursors within the neural plate, nor for further development of the anterior-dorsal hypothalamus. Nodal signaling is, however, cell-autonomously required for establishment of the posterior-ventral hypothalamus. Conversely, Hedgehog signaling antagonizes the development of posterior-ventral hypothalamus, while promoting anterior-dorsal hypothalamic fates. Besides their distinct roles in the regionalization of the diencephalon, we reveal cooperation between Nodal and Hedgehog pathways in the maintenance of the anterior-dorsal hypothalamus. Finally we show that it is the prechordal plate and not the head endoderm that provides the early signals essential for establishment of the hypothalamus.


Subject(s)
Hypothalamus/embryology , Trans-Activators/genetics , Transforming Growth Factor beta/genetics , Zebrafish Proteins , Zebrafish/embryology , Animals , Embryo, Nonmammalian , Embryonic Induction , Endoderm , Female , Gene Expression Regulation, Developmental , Hedgehog Proteins , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Hypothalamus/metabolism , Mutation , Nodal Protein , Trans-Activators/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Transforming Growth Factor beta/metabolism , Zebrafish/genetics
16.
Dev Biol ; 241(2): 273-88, 2002 Jan 15.
Article in English | MEDLINE | ID: mdl-11784111

ABSTRACT

Nodal signalling is essential for many developmental events during vertebrate development, including the establishment of left-right asymmetry, of dorsoventral axis of the central nervous system, and endoderm and mesoderm formation. The zebrafish TGFbeta-related type I receptor, TARAM-A (Tar), is expressed in the prospective mesendodermal territory and, when activated, can transfate early blastomeres into endoderm, suggesting that Nodal and Tar may represent similar signalling pathways. We have analysed the functional relationships between those two pathways in zebrafish. We first demonstrate that tar and the zebrafish nodal genes cyc and sqt functionally interact. We also show that a dominant-negative isoform of Tar, TarMR, interferes specifically with the function of Cyc and Sqt in vitro, but does not interfere with the function of BMP2, another TGFbeta-related molecule. TarMR interferes also with Nodal signalling in vivo since it enhances the phenotype of embryos with weakened Nodal signalling. Overexpression of tarMR in wild-type embryos interfered with the formation of endoderm-derived structures. Conversely, overexpression of tar enlarged the presumptive mesendodermal region at the onset of gastrulation. Together, our results point to Tar as an essential factor for endoderm formation and an important modulator of Nodal signalling, potentially representing one of the Nodal receptors. (c)2001 Elsevier Science.


Subject(s)
Body Patterning/physiology , Endoderm/physiology , Gene Expression Regulation, Developmental , Mesoderm/physiology , Protein Serine-Threonine Kinases , Receptors, Transforming Growth Factor beta/physiology , Signal Transduction/physiology , Transforming Growth Factor beta/physiology , Zebrafish Proteins , Activins/metabolism , Animals , Biomarkers , Body Patterning/genetics , Bone Morphogenetic Proteins/physiology , Embryo, Nonmammalian/physiology , Genes, Dominant , Homeodomain Proteins/genetics , Homeodomain Proteins/physiology , Intracellular Signaling Peptides and Proteins , Mutagenesis , Nodal Protein , Nodal Signaling Ligands , Phenotype , Receptors, Transforming Growth Factor beta/genetics , Recombinant Fusion Proteins/physiology , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/genetics , Transcription Factors/genetics , Transcription Factors/physiology , Transforming Growth Factor beta/genetics , Zebrafish/embryology , Zebrafish/genetics
17.
Development ; 129(2): 275-86, 2002 Jan.
Article in English | MEDLINE | ID: mdl-11807021

ABSTRACT

Endoderm originates from a large endomesodermal field requiring Nodal signalling. The mechanisms that ensure segregation of endoderm from mesoderm are not fully understood. We first show that the timing and dose of Nodal activation are crucial for endoderm formation and the endoderm versus mesoderm fate choice, because sustained Nodal signalling is required to ensure endoderm formation but transient signalling is sufficient for mesoderm formation. In zebrafish, downstream of Nodal signals, three genes encoding transcription factors (faust, bonnie and clyde and the recently identified gene casanova) are required for endoderm formation and differentiation. However their positions within the pathway are not completely established. In the present work, we show that casanova is the earliest specification marker for endodermal cells and that its expression requires bonnie and clyde. Furthermore, we have analysed the molecular activities of casanova on endoderm formation and found that it can induce endodermal markers and repress mesodermal markers during gastrulation, as well as change the fate of marginal blastomeres to endoderm. Overexpression of casanova also restores endoderm markers in the absence of Nodal signalling. In addition, casanova efficiently restores later endodermal differentiation in these mutants, but this process requires, in addition, a partial activation of Nodal signalling.


Subject(s)
Endoderm/physiology , Gene Expression Regulation, Developmental/physiology , High Mobility Group Proteins/metabolism , Signal Transduction/physiology , Transcription Factors/metabolism , Transforming Growth Factor beta/metabolism , Zebrafish Proteins , Zebrafish/embryology , Animals , Biomarkers , Cell Differentiation/physiology , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , GATA5 Transcription Factor , High Mobility Group Proteins/genetics , Homeodomain Proteins/metabolism , Immunohistochemistry , In Situ Hybridization , Microinjections , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Nodal Protein , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Ribosomal Proteins/genetics , Ribosomal Proteins/metabolism , SOX Transcription Factors , Transcription Factors/genetics , Transforming Growth Factor beta/genetics , Zebrafish/genetics , Zebrafish/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...