Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
1.
Dev Biol ; 313(2): 568-83, 2008 Jan 15.
Article in English | MEDLINE | ID: mdl-18068699

ABSTRACT

Neural crest-derived pigment cell development has been used extensively to study cell fate specification, migration, proliferation, survival and differentiation. Many of the genes and regulatory mechanisms required for pigment cell development are conserved across vertebrates. The zebrafish mutant colgate (col)/histone deacetylase1 (hdac1) has reduced numbers, delayed differentiation and decreased migration of neural crest-derived melanophores and their precursors. In hdac1(col) mutants normal numbers of premigratory neural crest cells are induced. Later, while there is only a slight reduction in the number of neural crest cells in hdac1(col) mutants, there is a severe reduction in the number of mitfa-positive melanoblasts suggesting that hdac1 is required for melanoblast specification. Concomitantly, there is a significant increase in and prolonged expression of foxd3 in neural crest cells in hdac1(col) mutants. We found that partially reducing Foxd3 expression in hdac1(col) mutants rescues mitfa expression and the melanophore defects in hdac1(col) mutants. Furthermore, we demonstrate the ability of Foxd3 to physically interact at the mitfa promoter. Because mitfa is required for melanoblast specification and development, our results suggest that hdac1 is normally required to suppress neural crest foxd3 expression thus de-repressing mitfa resulting in melanogenesis by a subset of neural crest-derived cells.


Subject(s)
Forkhead Transcription Factors/metabolism , Gene Expression Regulation, Developmental , Histone Deacetylases/physiology , Microphthalmia-Associated Transcription Factor/metabolism , Zebrafish Proteins/metabolism , Zebrafish Proteins/physiology , Zebrafish/physiology , Animals , Base Sequence , Binding Sites , Cell Movement , Electrophoretic Mobility Shift Assay , Embryo, Nonmammalian , Histone Deacetylase 1 , Histone Deacetylases/genetics , Histone Deacetylases/metabolism , In Situ Hybridization , Melanophores/cytology , Melanophores/metabolism , Melanophores/physiology , Microinjections , Models, Biological , Molecular Sequence Data , Mutation , Neural Crest/cytology , Neural Crest/embryology , Oligonucleotides, Antisense/pharmacology , Promoter Regions, Genetic , Protein Binding , Zebrafish/embryology , Zebrafish/genetics , Zebrafish/metabolism , Zebrafish Proteins/genetics
2.
Curr Biol ; 15(7): 667-71, 2005 Apr 12.
Article in English | MEDLINE | ID: mdl-15823540

ABSTRACT

Development of the adult form requires coordinated growth and patterning of multiple traits in response to local gene activity as well as to global endocrine and physiological effectors. An excellent example of such coordination is the skeleton. Skeletal development depends on the differentiation and morphogenesis of multiple cell types to generate elements with distinct forms and functions throughout the body. We show that zebrafish touchtone/nutria mutants exhibit severe growth retardation and gross alterations in skeletal development in addition to embryonic melanophore and touch-response defects. These alterations include accelerated endochondral ossification but delayed intramembranous ossification, as well as skeletal deformities. We show that the touchtone/nutria phenotype results from mutations in trpm7, which encodes a transient receptor potential (TRP) family member that functions as both a cation channel and kinase. We find trpm7 expression in the mesonephric kidney and show that mutants develop kidney stones, indicating renal dysfunction. These results identify a requirement for trpm7 in growth and skeletogenesis and highlight the potential of forward genetic approaches to uncover physiological mechanisms contributing to the development of adult form.


Subject(s)
Ion Channels/genetics , Kidney Calculi/veterinary , Osteogenesis/genetics , Protein Kinases/genetics , Zebrafish , Abnormalities, Multiple/genetics , Abnormalities, Multiple/metabolism , Animals , Base Sequence , Bone and Bones/anatomy & histology , Chromosome Mapping , DNA, Complementary/genetics , Histological Techniques , In Situ Hybridization , Ion Channels/metabolism , Kidney/metabolism , Kidney Calculi/genetics , Larva/metabolism , Molecular Sequence Data , Protein Kinases/metabolism , Protein Serine-Threonine Kinases , Sequence Analysis, DNA , TRPM Cation Channels , Zebrafish Proteins/genetics
3.
Mech Dev ; 124(9-10): 682-98, 2007.
Article in English | MEDLINE | ID: mdl-17716875

ABSTRACT

Vertebrate gastrulation involves the coordinated movements of populations of cells. These movements include cellular rearrangements in which cells polarize along their medio-lateral axes leading to cell intercalations that result in elongation of the body axis. Molecular analysis of this process has implicated the non-canonical Wnt/Frizzled signaling pathway that is similar to the planar cell polarity pathway (PCP) in Drosophila. Here we describe a zebrafish mutant, colgate (col), which displays defects in the extension of the body axis and the migration of branchiomotor neurons. Activation of the non-canonical Wnt/PCP pathway in these mutant embryos by overexpressing DeltaNdishevelled, rho kinase2 and van gogh-like protein 2 (vangl2) rescues the extension defects suggesting that col acts as a positive regulator of the non-canonical Wnt/PCP pathway. Further, we show that col normally regulates the caudal migration of nVII facial hindbrain branchiomotor neurons and that the mutant phenotype can be rescued by misexpression of vangl2 independent of the Wnt/PCP pathway. We cloned the col locus and found that it encodes histone deacetylase1 (hdac1). Our previous results and studies by others have implicated hdac1 in repressing the canonical Wnt pathway. Here, we demonstrate novel roles for zebrafish hdac1 in activating non-canonical Wnt/PCP signaling underlying axial extension and in promoting Wnt-independent caudal migration of a subset of hindbrain branchiomotor neurons.


Subject(s)
Axons/physiology , Body Patterning/physiology , Cell Movement/physiology , Histone Deacetylases/physiology , Motor Neurons/physiology , Signal Transduction/physiology , Wnt Proteins/physiology , Zebrafish Proteins/physiology , Zebrafish/embryology , Animals , Axons/enzymology , Body Patterning/genetics , Cell Movement/genetics , Cell Polarity/genetics , Cell Polarity/physiology , Histone Deacetylase 1 , Histone Deacetylases/genetics , Mutation , Rhombencephalon/cytology , Rhombencephalon/embryology , Rhombencephalon/enzymology , Signal Transduction/genetics , Wnt Proteins/genetics , Zebrafish/genetics , Zebrafish Proteins/genetics
4.
Mech Dev ; 121(11): 1353-64, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15454265

ABSTRACT

The specification, differentiation and maintenance of diverse cell types are of central importance to the development of multicellular organisms. The neural crest of vertebrate animals gives rise to many derivatives, including pigment cells, peripheral neurons, glia and elements of the craniofacial skeleton. The development of neural crest-derived pigment cells has been studied extensively to elucidate mechanisms involved in cell fate specification, differentiation, migration and survival. This analysis has been advanced considerably by the availability of large numbers of mouse and, more recently, zebrafish mutants with defects in pigment cell development. We have identified the zebrafish mutant touchtone (tct), which is characterized by the selective absence of most neural crest-derived melanophores. We find that although wild-type numbers of melanophore precursors are generated in the first day of development and migrate normally in tct mutants, most differentiated melanophores subsequently fail to appear. We demonstrate that the failure in melanophore differentiation in tct mutant embryos is due at least in part to the death of melanoblasts and that tct function is required cell autonomously by melanoblasts. The tct locus is located on chromosome 18 in a genomic region apparently devoid of genes known to be involved in melanophore development. Thus, zebrafish tct may represent a novel as well as selective regulator of melanoblast development within the neural crest lineage. Further, our results suggest that, like other neural crest-derived sublineages, melanogenic precursors constitute a heterogeneous population with respect to genetic requirements for development.


Subject(s)
Melanophores/cytology , Neural Crest/growth & development , Zebrafish Proteins/physiology , Zebrafish/embryology , Alleles , Animals , Apoptosis , Caspase 3 , Caspases/analysis , Cell Differentiation/genetics , Cell Differentiation/physiology , Cell Lineage/genetics , Cell Lineage/physiology , Chromosome Mapping , Embryo, Nonmammalian/cytology , Embryo, Nonmammalian/physiology , Melanophores/chemistry , Melanophores/physiology , Mutation/genetics , Protein Serine-Threonine Kinases , TRPM Cation Channels , Zebrafish/genetics , Zebrafish Proteins/genetics
5.
J Comp Neurol ; 446(3): 267-75, 2002 May 06.
Article in English | MEDLINE | ID: mdl-11932942

ABSTRACT

The trunk neural crest of vertebrate embryos gives rise to dorsal root ganglion (DRG) sensory neurons and autonomic sympathetic neurons, among other derivatives. We have examined the development of DRG and sympathetic neurons during development in the zebrafish. We found that sensory neurons differentiate rapidly and that their overt neuronal differentiation significantly precedes that of sympathetic neurons in the trunk. Sympathetic neurons in different regions differentiate at different times. The most rostral population, which we call the cervical ganglion, differentiates several days before trunk sympathetic neurons. After undergoing overt neuronal differentiation, sympathetic neurons subsequently express the adrenergic differentiation markers dopamine beta-hydroxylase and tyrosine hydroxylase. A second population of adrenergic nonneuronal cells initially localized with cervical sympathetic neurons appears to represent adrenal chromaffin cells. In more mature fish, these cells were present in clusters within the kidneys. Individual DRG and sympathetic ganglia initially contain few neurons. However, the number of neurons in DRG and sympathetic ganglia increases continuously at least up to 4 weeks of age. Analysis of phosphohistone H3 expression and bromodeoxyuridine incorporation studies suggests that the increases in DRG and sympathetic ganglion neuronal cell number are due wholly or in part to the division of neuronal cells within the ganglia.


Subject(s)
Ganglia, Spinal/cytology , Ganglia, Spinal/growth & development , Ganglia, Sympathetic/cytology , Ganglia, Sympathetic/growth & development , Animals , Antimetabolites , Bromodeoxyuridine , Cell Differentiation , Cell Division/physiology , Chromaffin Cells/physiology , Dopamine beta-Hydroxylase/biosynthesis , Embryo, Nonmammalian , Female , Ganglia, Spinal/embryology , Ganglia, Sympathetic/embryology , Immunohistochemistry , In Situ Hybridization , Neurons/physiology , RNA, Messenger/biosynthesis , Tyrosine 3-Monooxygenase/biosynthesis , Zebrafish
6.
PLoS One ; 8(5): e63218, 2013.
Article in English | MEDLINE | ID: mdl-23667588

ABSTRACT

The regulation of gene expression is accomplished by both genetic and epigenetic means and is required for the precise control of the development of the neural crest. In hdac1(b382) mutants, craniofacial cartilage development is defective in two distinct ways. First, fewer hoxb3a, dlx2 and dlx3-expressing posterior branchial arch precursors are specified and many of those that are consequently undergo apoptosis. Second, in contrast, normal numbers of progenitors are present in the anterior mandibular and hyoid arches, but chondrocyte precursors fail to terminally differentiate. In the peripheral nervous system, there is a disruption of enteric, DRG and sympathetic neuron differentiation in hdac1(b382) mutants compared to wildtype embryos. Specifically, enteric and DRG-precursors differentiate into neurons in the anterior gut and trunk respectively, while enteric and DRG neurons are rarely present in the posterior gut and tail. Sympathetic neuron precursors are specified in hdac1(b382) mutants and they undergo generic neuronal differentiation but fail to undergo noradrenergic differentiation. Using the HDAC inhibitor TSA, we isolated enzyme activity and temporal requirements for HDAC function that reproduce hdac1(b382) defects in craniofacial and sympathetic neuron development. Our study reveals distinct functional and temporal requirements for zebrafish hdac1 during neural crest-derived craniofacial and peripheral neuron development.


Subject(s)
Face/embryology , Histone Deacetylase 1/metabolism , Neural Crest/pathology , Neurons/metabolism , Skull/embryology , Zebrafish Proteins/metabolism , Zebrafish/embryology , Zebrafish/metabolism , Animals , Branchial Region/abnormalities , Branchial Region/embryology , Branchial Region/pathology , Cell Differentiation/drug effects , Craniofacial Abnormalities/embryology , Craniofacial Abnormalities/pathology , Embryo, Nonmammalian/drug effects , Embryo, Nonmammalian/metabolism , Embryo, Nonmammalian/pathology , Face/abnormalities , Face/pathology , Histone Deacetylase 1/genetics , Hydroxamic Acids/pharmacology , Hyoid Bone/abnormalities , Hyoid Bone/drug effects , Hyoid Bone/embryology , Hyoid Bone/pathology , Mandible/abnormalities , Mandible/drug effects , Mandible/embryology , Mandible/pathology , Mutation/genetics , Neural Crest/drug effects , Neural Crest/embryology , Neural Crest/metabolism , Neurons/drug effects , Neurons/pathology , Peripheral Nervous System/drug effects , Peripheral Nervous System/embryology , Peripheral Nervous System/pathology , Phenotype , Skull/abnormalities , Skull/pathology , Stem Cells/drug effects , Stem Cells/metabolism , Stem Cells/pathology , Sympathetic Nervous System/drug effects , Sympathetic Nervous System/metabolism , Sympathetic Nervous System/pathology , Time Factors , Zebrafish Proteins/genetics
7.
Int J Dev Biol ; 56(4): 223-37, 2012.
Article in English | MEDLINE | ID: mdl-22562198

ABSTRACT

The functions of gene regulatory networks that control embryonic cell diversification occur on a background of constitutively active molecular machinery necessary for the elaboration of genetic interactions. The essential roles of subsets of such "housekeeping" genes in the regulation of specific aspects of development have become increasingly clear. Pre-mRNA processing is essential for production of functional transcripts by, for example, excision of introns. We have cloned the zebrafish toast(b460) locus and found that it encodes splicing factor 3b, subunit 1 (sf3b1). The sf3b1(b460) mutation causes aberrant splicing of sf3b1 resulting in functional and predicted non-functional transcripts and a 90% reduction in full-length Sf3b1 protein. The sf3b1(b460) mutation was isolated in a mutagenesis screen based on the absence of neural crest-derived melanophores. Further analysis revealed specific earlier defects in neural crest development, whereas the early development of other ectodermal populations appears unaffected. The expression of essential transcriptional regulators of neural crest development are severely disrupted in sf3b1(b460) mutants, due in part to defects in pre-mRNA processing of a subset of these factors, leading to defects in neural crest sublineage specification, survival and migration. Misexpression of a subset of these factors rescues aspects of neural crest development in mutant embryos. Our results indicate that although sf3b1 is a ubiquitously essential gene, the degree to which it is required exhibits tissue-type specificity during early embryogenesis. Further, the developmental defects caused by the sf3b1(b460) mutation provide insights into genetic interactions among members of the gene regulatory network controlling neural crest development.


Subject(s)
Mutation , Neural Crest/metabolism , RNA Precursors/genetics , RNA-Binding Proteins/genetics , Zebrafish Proteins/genetics , Amino Acid Sequence , Animals , Apoptosis , Base Sequence , Blotting, Western , Embryo, Nonmammalian/cytology , Embryo, Nonmammalian/embryology , Embryo, Nonmammalian/metabolism , Gene Expression Regulation, Developmental , Immunohistochemistry , In Situ Hybridization , Melanophores/cytology , Melanophores/metabolism , Molecular Sequence Data , Neural Crest/cytology , Neural Crest/embryology , RNA Precursors/metabolism , RNA Splice Sites/genetics , RNA Splicing , RNA Splicing Factors , RNA-Binding Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Sequence Homology, Amino Acid , Zebrafish/embryology , Zebrafish/genetics , Zebrafish/metabolism , Zebrafish Proteins/metabolism
8.
Methods Cell Biol ; 100: 127-52, 2010.
Article in English | MEDLINE | ID: mdl-21111216

ABSTRACT

The combined experimental attributes of the zebrafish model system, which accommodates cellular, molecular, and genetic approaches, make it particularly well-suited for determining the mechanisms underlying normal vertebrate development as well as disease states, such as cancer. In this chapter, we describe the advantages of the zebrafish system for identifying genes and their functions that participate in the regulation of the development of the peripheral sympathetic nervous system (PSNS). The zebrafish model is a powerful system for identifying new genes and pathways that regulate PSNS development, which can then be used to genetically dissect PSNS developmental processes, such as tissue size and cell numbers, which in the past haves proved difficult to study by mutational analysis in vivo. We provide a brief review of our current understanding of genetic pathways important in PSNS development, the rationale for developing a zebrafish model, and the current knowledge of zebrafish PSNS development. Finally, we postulate that knowledge of the genes responsible for normal PSNS development in the zebrafish will help in the identification of molecular pathways that are dysfunctional in neuroblastoma, a highly malignant cancer of the PSNS.


Subject(s)
Neurogenesis , Sympathetic Nervous System/embryology , Animals , Neuroblastoma , Organogenesis , Zebrafish
9.
Development ; 136(12): 1987-94, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19439494

ABSTRACT

The neural crest generates multiple cell types during embryogenesis but the mechanisms regulating neural crest cell diversification are incompletely understood. Previous studies using mutant zebrafish indicated that foxd3 and tfap2a function early and differentially in the development of neural crest sublineages. Here, we show that the simultaneous loss of foxd3 and tfap2a function in zebrafish foxd3(zdf10);tfap2a(low) double mutant embryos globally prevents the specification of developmentally distinct neural crest sublineages. By contrast, neural crest induction occurs independently of foxd3 and tfap2a function. We show that the failure of neural crest cell diversification in double mutants is accompanied by the absence of neural crest sox10 and sox9a/b gene expression, and that forced expression of sox10 and sox9a/b differentially rescues neural crest sublineage specification and derivative differentiation. These results demonstrate the functional necessity for foxd3 and tfap2a for neural crest sublineage specification and that this requirement is mediated by the synergistic regulation of the expression of SoxE family genes. Our results identify a genetic regulatory pathway functionally discrete from the process of neural crest induction that is required for the initiation of neural crest cell diversification during embryonic development.


Subject(s)
Cell Lineage/physiology , Neural Crest/physiology , Zebrafish/embryology , Animals , Embryo, Nonmammalian/physiology , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Gene Expression Regulation, Developmental , Mutation , Neural Crest/embryology , SOX9 Transcription Factor/genetics , SOX9 Transcription Factor/metabolism , Transcription Factor AP-2/genetics , Transcription Factor AP-2/metabolism , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism
10.
PLoS One ; 3(7): e2845, 2008 Jul 30.
Article in English | MEDLINE | ID: mdl-18665240

ABSTRACT

The development of neural crest-derived pigment cells has been studied extensively as a model for cellular differentiation, disease and environmental adaptation. Neural crest-derived chromatophores in the zebrafish (Danio rerio) consist of three types: melanophores, xanthophores and iridiphores. We have identified the zebrafish mutant endzone (enz), that was isolated in a screen for mutants with neural crest development phenotypes, based on an abnormal melanophore pattern. We have found that although wild-type numbers of chromatophore precursors are generated in the first day of development and migrate normally in enz mutants, the numbers of all three chromatophore cell types that ultimately develop are reduced. Further, differentiated melanophores and xanthophores subsequently lose dendricity, and iridiphores are reduced in size. We demonstrate that enz function is required cell autonomously by melanophores and that the enz locus is located on chromosome 7. In addition, zebrafish enz appears to selectively regulate chromatophore development within the neural crest lineage since all other major derivatives develop normally. Our results suggest that enz is required relatively late in the development of all three embryonic chromatophore types and is normally necessary for terminal differentiation and the maintenance of cell size and morphology. Thus, although developmental regulation of different chromatophore sublineages in zebrafish is in part genetically distinct, enz provides an example of a common regulator of neural crest-derived chromatophore differentiation and morphology.


Subject(s)
Cell Differentiation , Chromatophores/metabolism , Melanophores/metabolism , Animals , Body Patterning , Cell Size , Chromosome Mapping , Models, Biological , Models, Genetic , Mutation , Neural Crest/metabolism , Phenotype , Time Factors , Zebrafish
11.
Development ; 133(20): 4015-24, 2006 Oct.
Article in English | MEDLINE | ID: mdl-17008447

ABSTRACT

The basic helix-loop-helix transcription factor Hand2, together with Ascl1, Phox2a, Phox2b and Gata2/Gata3, is induced by bone morphogenetic proteins in neural crest-derived precursor cells during sympathetic neuron generation. Hand2 overexpression experiments and the analysis of its function at the Dbh promotor implicated Hand2 in the control of noradrenergic gene expression. Using the zebrafish hand2 deletion mutant hands off, we have now investigated the physiological role of hand2 in the development of sympathetic ganglia. In hands off mutant embryos, sympathetic precursor cells aggregate to form normal sympathetic ganglion primordia characterized by the expression of phox2b, phox2a and the achaete-scute family member zash1a/ascl1. The expression of the noradrenergic marker genes th and dbh is strongly reduced, as well as the transcription factors gata2 and tfap2a (Ap-2alpha). By contrast, generic neuronal differentiation seems to be unaffected, as the expression of elavl3 (HuC) is not reduced in hands off sympathetic ganglia. These results demonstrate in vivo an essential and selective function of hand2 for the noradrenergic differentiation of sympathetic neurons, and implicates tfap2a and gata2 as downstream effectors.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/physiology , Ganglia, Sympathetic/embryology , Gene Expression Regulation, Developmental , Neurons/cytology , Zebrafish Proteins/physiology , Zebrafish/embryology , Animals , Basic Helix-Loop-Helix Transcription Factors/analysis , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Differentiation/genetics , GATA2 Transcription Factor/genetics , GATA2 Transcription Factor/metabolism , Ganglia, Sympathetic/chemistry , Ganglia, Sympathetic/cytology , Gene Deletion , Homeodomain Proteins/analysis , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Neurons/chemistry , Neurons/metabolism , Norepinephrine/metabolism , Transcription Factor AP-2/analysis , Transcription Factor AP-2/genetics , Transcription Factor AP-2/metabolism , Transcription Factors/analysis , Transcription Factors/genetics , Transcription Factors/metabolism , Zebrafish/genetics , Zebrafish Proteins/analysis , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism
12.
Dev Biol ; 292(1): 174-88, 2006 Apr 01.
Article in English | MEDLINE | ID: mdl-16499899

ABSTRACT

The vertebrate neural crest is a pluripotent cell population that generates a large variety of cell types, including peripheral neurons, cartilage and pigment cells. Mechanisms that control the patterning of the neural crest toward specific cell fates remain only partially understood. Zebrafish homozygous for the sympathetic mutation 1 (sym1) have defects in a subset of neural crest derivatives, such as peripheral neurons, glia and cartilage, but retain normal numbers of melanocytes. The sym1 mutation is a nucleotide deletion that disrupts the forkhead DNA-binding domain of the foxd3 gene, which encodes a conserved winged-helix transcription factor. We show that sym1 mutants have normal numbers of premigratory neural crest cells, but these cells express reduced levels of snai1b and sox10, implicating foxd3 as an essential regulator of these transcription factors in the premigratory neural crest. The onset of neural crest migration is also delayed in sym1 mutants, and there is a reduction in the number of migratory trunk neural crest cells, particularly along the medial migration pathway. TUNEL analysis revealed aberrant apoptosis localized to the hindbrain neural crest at the 15-somite stage, indicating a critical role for foxd3 in the survival of a subpopulation of neural crest cells. These results show that foxd3 selectively specifies premigratory neural crest cells for a neuronal, glial or cartilage fate, by inducing the expression of lineage-associated transcription factors in these cells and regulating their subsequent migration.


Subject(s)
Cell Movement/physiology , Forkhead Transcription Factors/physiology , Neural Crest/cytology , Neural Crest/embryology , Zebrafish Proteins/physiology , Zebrafish/genetics , Animals , Cell Lineage/genetics , Cell Lineage/physiology , Cell Movement/genetics , Cell Survival/genetics , Cell Survival/physiology , Female , Forkhead Transcription Factors/deficiency , Forkhead Transcription Factors/genetics , Neural Crest/physiology , Sympathetic Nervous System/cytology , Sympathetic Nervous System/physiology , Transcription Factors/biosynthesis , Transcription Factors/physiology , Zebrafish/embryology , Zebrafish/physiology , Zebrafish Proteins/deficiency , Zebrafish Proteins/genetics
13.
Dev Biol ; 267(1): 165-80, 2004 Mar 01.
Article in English | MEDLINE | ID: mdl-14975724

ABSTRACT

The establishment of the vertebrate body plan involves patterning of the ectoderm, mesoderm, and endoderm along the dorsoventral and antero-posterior axes. Interactions among numerous signaling molecules from several multigene families, including Wnts, have been implicated in regulating these processes. Here we provide evidence that the zebrafish colgate(b382) (col) mutation results in increased Wnt signaling that leads to defects in dorsal and anterior development. col mutants display early defects in dorsoventral patterning manifested by a decrease in the expression of dorsal shield-specific markers and ectopic expression of ventrolaterally expressed genes during gastrulation. In addition to these early patterning defects, col mutants display a striking regional posteriorization within the neuroectoderm, resulting in a reduction in anterior fates and an expansion of posterior fates within the forebrain and midbrain-hindbrain regions. We are able to correlate these phenotypes to the overactivation of Wnt signaling in col mutants. The early dorsal and anterior patterning phenotypes of the col mutant embryos are selectively rescued by inactivation of Wnt8 function by morpholino translational interference. In contrast, the regionalized neuroectoderm posterioriorization phenotype is selectively rescued by morpholino-mediated inactivation of Wnt8b. These results suggest that col-mediated antagonism of early and late Wnt-signaling activity during gastrulation is normally required sequentially for both early dorsoventral patterning and the specification and patterning of regional fates within the anterior neuroectoderm.


Subject(s)
Cell Lineage , Proto-Oncogene Proteins/metabolism , Signal Transduction/physiology , Zebrafish Proteins , Zebrafish/embryology , Animals , Immunohistochemistry , In Situ Hybridization , Phenotype , Wnt Proteins
14.
Development ; 130(2): 321-30, 2003 Jan.
Article in English | MEDLINE | ID: mdl-12466199

ABSTRACT

Clonal and lineage analyses have demonstrated that although some neural crest cells have the ability to generate multiple cell types and display self-renewal ability, other crest cells generate a single or limited repertoire of cell types. However, it is not yet clear when, and in what order, crest cells become specified to adopt a particular fate. We report that the receptor tyrosine kinases TrkC and C-Kit are expressed by distinct neural crest subpopulations in vitro. We then analyzed the lineages of individual receptor-expressing crest cells and found that TrkC-expressing cells that have just emerged from the neural tube give rise to clones containing neurons or glial cells, or both, but never produce melanocytes. A short time later, TrkC-expressing cells only generate pure neuronal clones. By contrast, from their earliest appearance in neural tube outgrowths, C-Kit-expressing cells invariably give rise to clones containing only melanocytes. Our results directly demonstrate that distinct neurogenic and melanogenic sublineages diverge before or soon after crest cells emerge from the neural tube, that fate-restricted precursors are present in nascent neural crest populations and that these sublineages can be distinguished by their cell type-specific expression of receptor tyrosine kinases.


Subject(s)
Cell Lineage , Neural Crest/embryology , Proto-Oncogene Proteins c-kit/metabolism , Receptor, trkC/metabolism , Stem Cells/physiology , Animals , Cells, Cultured , Clone Cells/metabolism , Embryo, Nonmammalian/anatomy & histology , Gene Expression Regulation, Developmental , Immunohistochemistry , Melanocytes/physiology , Neural Crest/cytology , Quail , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL