Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 99
Filter
Add more filters










Publication year range
1.
Development ; 128(19): 3675-83, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11585794

ABSTRACT

Recent evidence indicates that acquisition of artery or vein identity during vascular development is governed, in part, by genetic mechanisms. The artery-specific expression of a number of Notch signaling genes in mouse and zebrafish suggests that this pathway may play a role in arterial-venous cell fate determination during vascular development. We show that loss of Notch signaling in zebrafish embryos leads to molecular defects in arterial-venous differentiation, including loss of artery-specific markers and ectopic expression of venous markers within the dorsal aorta. Conversely, we find that ectopic activation of Notch signaling leads to repression of venous cell fate. Finally, embryos lacking Notch function exhibit defects in blood vessel formation similar to those associated with improper arterial-venous specification. Our results suggest that Notch signaling is required for the proper development of arterial and venous blood vessels, and that a major role of Notch signaling in blood vessels is to repress venous differentiation within developing arteries. Movies available on-line


Subject(s)
Arteries/embryology , Embryonic Induction , Proto-Oncogene Proteins/metabolism , Receptors, Cell Surface , Veins/embryology , Zebrafish Proteins , Zebrafish/embryology , Animals , Base Sequence , Basic Helix-Loop-Helix Transcription Factors , Biomarkers , Cell Differentiation/physiology , Ephrin-B2 , Female , Gene Expression Regulation, Developmental , Intracellular Signaling Peptides and Proteins , Membrane Proteins/genetics , Membrane Proteins/metabolism , Microinjections , Molecular Sequence Data , Mutation , Proteins/metabolism , Proto-Oncogene Proteins/genetics , Receptor Protein-Tyrosine Kinases/metabolism , Receptors, Growth Factor/metabolism , Signal Transduction , Vascular Endothelial Growth Factor Receptor-3 , Zebrafish/genetics
2.
Development ; 128(7): 1099-107, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11245575

ABSTRACT

The Gal4-UAS technique has been used to misexpress a constitutively active Notch receptor variant (notch1a-intra) in the developing zebrafish retina. This is the first study to use this technique to misexpress genes and assess their function in neural development of the zebrafish. Expression of activated Notch1a either ubiquitously, driven by a heat-shock70 promoter, or in a spatially regulated manner, controlled by the deltaD promoter, causes a block in neuronal differentiation that affects all cell types. Developing cells take on either a glial fate or remain undifferentiated. A large number of cells eventually undergo apoptosis. These phenotypic effects of activated Notch1a are expressed cell autonomously. Cells within central regions of the retina adopt a glial fate if they express activated Notch1a in a time window that extends from 27 to 48 hours postfertilization. This period corresponds mainly to the time of origin of ganglion cells in the normal retina. Activation of notch1a at later stages results in defects in cell type specification that remain restricted to the ciliary marginal zone, whereas neuronal types are specified normally within the central region. These observations indicate that glial differentiation is initiated by Notch1a-intra expressing cells, which become postmitotic in the same time window. Our results strongly suggest that Notch1a instructs a certain cell population to enter gliogenesis, and keeps the remaining cells in an undifferentiated state. Some or all of these cells will eventually succumb to apoptosis.


Subject(s)
Membrane Proteins/physiology , Receptors, Cell Surface , Retinal Ganglion Cells/cytology , Transcription Factors , Animals , Apoptosis , Biomarkers , Cell Differentiation , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mitosis/physiology , Receptor, Notch1 , Retinal Ganglion Cells/metabolism , Retinal Ganglion Cells/physiology , Zebrafish
3.
Dev Genes Evol ; 211(12): 603-10, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11819118

ABSTRACT

Abstract. We have generated transgenic zebrafish lines expressing a fusion of a histone variant, H2A.F/Z, to the green fluorescent protein (GFP) of the jellyfish Aequorea victoria. Here, we describe the molecular cloning, partial characterisation and expression of the zebrafish H2A.F/Z histone gene, as well as the construction of the transgene and its transformation into the zebrafish germ line. No abnormality can be detected in transgenic fish expressing the H2A.F/Z:GFP fusion protein. The nuclear localisation of the fusion protein correlates with the start of zygotic transcription, in that it is present in the unfertilised egg and in the cytoplasm of cells after the first cleavages, being found in some nuclei after the seventh or eighth cleavage, whereas all nuclei from the 1,000-cell stage on, i.e. after midblastula transition, contain protein. In addition to these data, we present a few examples of the many possible applications of this transgenic line for developmental studies in vivo. Electronic supplementary material to this paper can be obtained by using the Springer LINK server located at http://dx.doi.org/10.1007/s00427-001-0196-x


Subject(s)
Animals, Genetically Modified/genetics , Histones/genetics , Zebrafish/embryology , Zebrafish/genetics , Animals , Animals, Genetically Modified/embryology , Animals, Genetically Modified/physiology , Genes, Reporter , Green Fluorescent Proteins , Histones/physiology , Luminescent Proteins , Molecular Sequence Data , Recombinant Fusion Proteins/genetics
4.
Mech Dev ; 95(1-2): 23-34, 2000 Jul.
Article in English | MEDLINE | ID: mdl-10906447

ABSTRACT

The Delta1 gene encodes one of the Notch ligands in mice. Delta1 is expressed during early embryogenesis in a complex and dynamic pattern in the paraxial mesoderm and neuroectoderm, and is essential for normal somitogenesis and neuronal differentiation. Molecular components thought to act in response to ligand binding and Notch activation have been identified in different species. In contrast, little is known about the transcriptional regulation of Notch receptors and their ligands. As a first step to identify upstream factors regulating Delta1 expression in different tissues, we searched for cis-regulatory regions in the Delta1 promoter able to direct heterologous gene expression in a tissue specific manner in transgenic mice. Our results show that a 4.3 kb genomic DNA fragment of the Delta1 gene is sufficient in a lacZ reporter transgene to reproduce most aspects of Delta1 expression from the primitive streak stage to early organogenesis. Using a minimal Delta1 promoter we also show that this upstream region contains distinct regulatory modules that individually direct tissue-specific transgene expression in subdomains of the endogenous expression pattern. It appears that expression in the paraxial mesoderm depends on the interaction of multiple positive and negative regulatory elements. We also find that at least some regulatory sequences required for transgene expression in subdomains of the neural tube have been maintained during the evolution of mammals and teleost fish, suggesting that part of the regulatory network that controls expression of Delta genes may be conserved.


Subject(s)
Gene Expression Regulation, Developmental/physiology , Membrane Proteins/physiology , Mesoderm/cytology , Nervous System/cytology , Nervous System/embryology , Animals , Base Sequence , Cell Communication/physiology , Cell Differentiation/physiology , Embryonic and Fetal Development , Homeodomain Proteins/physiology , Intracellular Signaling Peptides and Proteins , Mesoderm/physiology , Mice , Mice, Transgenic , Molecular Sequence Data , Nervous System Physiological Phenomena
5.
Mech Dev ; 86(1-2): 17-28, 1999 Aug.
Article in English | MEDLINE | ID: mdl-10446262

ABSTRACT

We describe the characterization of the zebrafish homologue of the human gene DLG3. The zebrafish dlg3 gene encodes a membrane-associated guanylate kinase containing a single PDZ domain. This gene was cloned using a gene-trap construct inserted in the gene's first intron. The insertion co-segregates with a viable mutation called humpback (hmp), which leads to formation of ankylotic vertebrae in adult fishes. Insertion and mutation have both been mapped to chromosome 12, in a segment which is syntenic with region p12 to q12 of human chromosome 17. The hmp mutant phenotype, however, appears to be due to two point mutations in the guanylate kinase domain rather than to the transgene insertion itself. The results of this study are discussed in the light of the possible function of the guanylate kinase domain.


Subject(s)
Ankylosis/genetics , Fish Diseases/genetics , Membrane Proteins/genetics , Nucleoside-Phosphate Kinase/genetics , Spine/abnormalities , Zebrafish/genetics , Animals , Animals, Genetically Modified , Ankylosis/veterinary , Chromosome Mapping , Cloning, Molecular , DNA Transposable Elements , Genes, Recessive , Guanylate Kinases , Humans , Membrane Proteins/metabolism , Molecular Sequence Data , Mutation , Nucleoside-Phosphate Kinase/metabolism , Phosphoproteins , Transcription, Genetic , Transgenes , Zebrafish Proteins , Zonula Occludens-1 Protein
6.
Development ; 126(13): 3005-14, 1999 Jul.
Article in English | MEDLINE | ID: mdl-10357943

ABSTRACT

During vertebrate embryonic development, the paraxial mesoderm becomes subdivided into metameric units known as somites. In the zebrafish embryo, genes encoding homologues of the proteins of the Drosophila Notch signalling pathway are expressed in the presomitic mesoderm and expression is maintained in a segmental pattern during somitogenesis. This expression pattern suggests a role for these genes during somite development. We misexpressed various zebrafish genes of this group by injecting mRNA into early embryos. RNA encoding a constitutively active form of notch1a (notch1a-intra) and a truncated variant of deltaD [deltaD(Pst)], as well as transcripts of deltaC and deltaD, the hairy-E(spl) homologues her1 and her4, and groucho2 were tested for their effects on somite formation, myogenesis and on the pattern of transcription of putative downstream genes. In embryos injected with any of these RNAs, with the exception of groucho2 RNA, the paraxial mesoderm differentiated normally into somitic tissue, but failed to segment correctly. Activation of notch results in ectopic activation of her1 and her4. This misregulation of the expression of her genes might be causally related to the observed mesodermal defects, as her1 and her4 mRNA injections led to effects similar to those seen with notch1a-intra. deltaC and deltaD seem to function after subdivision of the presomitic mesoderm, since the her gene transcription pattern in the presomitic mesoderm remains essentially normal after misexpression of delta genes. Whereas notch signalling alone apparently does not affect myogenesis, zebrafish groucho2 is involved in differentiation of mesodermal derivatives.


Subject(s)
Caenorhabditis elegans Proteins , Helminth Proteins/genetics , Membrane Proteins/genetics , Proteins/genetics , Somites/cytology , Somites/metabolism , Zebrafish Proteins , Zebrafish/genetics , Animals , Basic Helix-Loop-Helix Transcription Factors , Cell Differentiation , DNA-Binding Proteins/genetics , Gene Expression Regulation, Developmental , Muscles/embryology , RNA, Messenger/metabolism , Receptors, Notch , Repressor Proteins/genetics , Signal Transduction/genetics , Zebrafish/embryology
7.
Development ; 126(9): 1811-21, 1999 May.
Article in English | MEDLINE | ID: mdl-10101116

ABSTRACT

her4 encodes a zebrafish bHLH protein of the hairy-E(spl) family. The gene is transcribed in a complex pattern in the developing nervous system and in the hypoblast. During early neurogenesis, her4 expression domains include the regions of the neural plate from which primary neurons arise, suggesting that the gene is involved in directing their development. Indeed, misexpression of specific her4 variants leads to a reduction in the number of primary neurons formed. The amino-terminal region of her4, including the basic domain, and the region between the putative helix IV and the carboxy-terminal tetrapeptide wrpw are essential for this effect, since her4 variants lacking either of these regions are non-functional. However, the carboxy-terminal wrpw itself is dispensable. We have examined the interrelationships between deltaD, deltaA, notch1, her4 and neurogenin1 by means of RNA injections. her4 is involved in a regulatory feedback loop which modulates the activity of the proneural gene neurogenin, and as a consequence, of deltaA and deltaD. Activation of notch1 leads to strong activation of her4, to suppression of neurogenin transcription and, ultimately, to a reduction in the number of primary neurons. These results suggest that her4 acts as a target of notch-mediated signals that regulate primary neurogenesis.


Subject(s)
DNA-Binding Proteins/genetics , Drosophila Proteins , Embryo, Nonmammalian/physiology , Gene Expression Regulation, Developmental , Insect Proteins/genetics , Membrane Proteins/metabolism , Nervous System/embryology , Neurons/physiology , Repressor Proteins , Xenopus Proteins , Zebrafish Proteins , Zebrafish/embryology , Animals , Basic Helix-Loop-Helix Transcription Factors , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/physiology , Drosophila/genetics , Embryonic Induction , Feedback , Helix-Loop-Helix Motifs , Homeodomain Proteins/genetics , LIM-Homeodomain Proteins , Nerve Tissue Proteins/genetics , Protein Structure, Secondary , Receptors, Notch , Signal Transduction , Trans-Activators/metabolism , Transcription Factors , Transcription, Genetic , Zebrafish/genetics
8.
Dev Genes Evol ; 209(3): 135-44, 1999 Mar.
Article in English | MEDLINE | ID: mdl-10079356

ABSTRACT

To analyse the proliferative abilities of cells within particular regions of the zebrafish neural plate, injections of fluorescein-dextran were made into single cells at either medial or intermediary positions in the neural plate region of two-somite stage embryos. The resulting cell clones were analysed in 3. 5-day-old embryos. Clones with similar compositions were found among those derived from injections in both regions, and these were grouped into classes. 78 clones 29 obtained following injections in the medial region, and 22 of 59 cell clones derived from injections in the intermediary region, were classifiable into 9 and 10 different classes, respectively, each comprising a variable number of clones. Several identified cell types, as well as each of the clone classes themselves, were specific for the regions of the neural plate from which they derived, i.e. they were not represented among the clones derived from the other region. These results suggest that the composition of the lineages derived from particular cells is constant in different animals.


Subject(s)
Spinal Cord/cytology , Spinal Cord/embryology , Zebrafish/embryology , Animals , Cell Count , Cell Division , Clone Cells/cytology , Dextrans/administration & dosage , Fluoresceins/administration & dosage , Microinjections
9.
Mech Dev ; 80(2): 153-8, 1999 Feb.
Article in English | MEDLINE | ID: mdl-10072782

ABSTRACT

The most common way to analyze the function of cloned genes in zebrafish is to misexpress the gene product or an altered variant of it by mRNA injection. However, mRNA injection has several disadvantages. The GAL4-UAS system for targeted gene expression allows one to overcome some of these disadvantages. To test the GAL4-UAS system in zebrafish, we generated two different kinds of stable transgenic lines, carrying activator and effector constructs, respectively. In the activator lines the gene for the yeast transcriptional activator GAL4 is under the control of a given promoter, while in the effectors the gene of interest is fused to the sequence of the DNA-binding motif of GAL4 (UAS). Crosses of animals from the activator and effector lines show that effector genes are transcribed with the spatial pattern of the activators. This work smoothes the way for a novel method of misexpression of gene products in zebrafish in order to analyze the function of genes in developmental processes.


Subject(s)
Fungal Proteins/genetics , Gene Expression Regulation, Developmental , Genes, Reporter , Promoter Regions, Genetic , Receptors, Cell Surface , Regulatory Sequences, Nucleic Acid , Saccharomyces cerevisiae Proteins , Transcription Factors/genetics , Transcription, Genetic , Zebrafish/genetics , Actins/genetics , Adenovirus E1B Proteins/genetics , Animals , Animals, Genetically Modified , Cloning, Molecular , Crosses, Genetic , DNA-Binding Proteins , Embryo, Nonmammalian/metabolism , Enhancer Elements, Genetic , Genes, myc , In Situ Hybridization , Membrane Proteins/biosynthesis , Membrane Proteins/genetics , Pilot Projects , Proto-Oncogene Proteins c-myc/biosynthesis , Receptor, Notch1 , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/physiology , Simian virus 40/genetics , Simplexvirus/enzymology , Simplexvirus/genetics , Thymidine Kinase/genetics , Transgenes , Viral Proteins/genetics , Zebrafish/embryology
10.
Dev Genes Evol ; 209(2): 126-31, 1999 Feb.
Article in English | MEDLINE | ID: mdl-10022956

ABSTRACT

In a search for novel, developmentally regulated genes we screened randomly picked cDNA clones, obtained from zebrafish mRNA, by in situ hybridization with digoxigenin-labelled riboprobes. Out of 150 clones tested, 1 codes for a new beta-globin gene and is expressed during embryogenesis. Here we describe its pattern of expression and its use as a marker for early zebrafish erythropoiesis.


Subject(s)
Gene Expression Regulation, Developmental , Globins/genetics , Zebrafish/embryology , Zebrafish/genetics , Animals , Cell Lineage , DNA, Complementary/genetics , Transcription, Genetic
11.
Int J Dev Biol ; 42(3): 291-7, 1998.
Article in English | MEDLINE | ID: mdl-9654011

ABSTRACT

The Drosophila achaete-scute complex consists of four genes encoding transcription factors of the bHLH family. Due to their intricate organization, these genes have occupied geneticists and developmental biologists for many years. Here, genetic studies on the complex are discussed from a historical point of view.


Subject(s)
DNA-Binding Proteins/genetics , Drosophila Proteins , Drosophila/genetics , Gene Expression Regulation, Developmental , Helix-Loop-Helix Motifs/genetics , Transcription Factors/genetics , Alleles , Animals , Basic Helix-Loop-Helix Transcription Factors , Central Nervous System/embryology , Central Nervous System/growth & development , DNA-Binding Proteins/physiology , Genes, Insect , Phenotype , Transcription Factors/physiology
12.
Development ; 124(16): 3123-34, 1997 Aug.
Article in English | MEDLINE | ID: mdl-9272953

ABSTRACT

The klumpfuss (klu) transcription unit in Drosophila gives rise to two different transcripts of 4.5 and 4.9 kb, both of which encode a putative transcription factor with four zinc-finger motifs of the C2H2 class. Zinc-finger 2-4 are homologous to those of the proteins of the EGR transcription factor family. As in the case of the most divergent member of the family, the Wilms' tumor suppressor gene (WT-1), klu contains an additional zinc finger, which is only distantly related. Loss of klumpfuss function is semilethal and causes a variety of defects in bristles and legs of adults, as well as in mouth hooks and brains of larvae. Analysis of the mutants indicates that klumpfuss is required for proper specification and differentiation of a variety of cells, including the sensory organ mother cells and those of the distal parts of tarsal segments.


Subject(s)
Drosophila Proteins , Drosophila/growth & development , Drosophila/genetics , Genes, Insect/genetics , Transcription Factors/genetics , Amino Acid Sequence , Animals , Basic Helix-Loop-Helix Transcription Factors , Cell Differentiation , Cloning, Molecular , DNA/analysis , DNA-Binding Proteins/genetics , Epidermis/growth & development , Extremities/growth & development , Gene Expression Regulation, Developmental , Molecular Sequence Data , Mutation , Phenotype , RNA, Messenger/analysis , Restriction Mapping , Sequence Analysis, DNA , Transcription Factors/physiology , Zinc Fingers
13.
Mech Dev ; 65(1-2): 175-85, 1997 Jul.
Article in English | MEDLINE | ID: mdl-9256354

ABSTRACT

We describe the cloning, sequencing and pattern of transcript distribution during embryogenesis of a zebrafish Id homologue that we have called Id6. Transcription of the gene is spatially regulated, and its pattern of transcription shows considerable overlaps with those of other zebrafish genes with homology to Drosophila neurogenic genes, such as Notch and Delta. Since all these genes are coexpressed in particular cells, they may function together in a single genetic circuit in zebrafish as they do in Drosophila. A zebrafish homologue of Drosophila AS-C proteins can activate transcription of a CAT reporter gene by binding to an E-box in mouse 3T3 cells, either alone or in conjunction with ZfE12. The activation of transcription is inhibited in the presence of Id6. This indicates that the zebrafish gene described here is a genuine member of the Id family, and suggests that it may serve a function similar to that of the Drosophila gene emc and mammalian Ids during development.


Subject(s)
Gene Expression Regulation, Developmental , Repressor Proteins , Transcription Factors/genetics , Zebrafish/embryology , Amino Acid Sequence , Animals , Base Sequence , Cloning, Molecular , Drosophila , Helix-Loop-Helix Motifs/genetics , Inhibitor of Differentiation Protein 1 , Mice , Molecular Sequence Data , Sequence Analysis , Sequence Homology, Amino Acid
14.
Proc Natl Acad Sci U S A ; 94(12): 6250-4, 1997 Jun 10.
Article in English | MEDLINE | ID: mdl-9177203

ABSTRACT

The Enhancer of split [E(spl)] gene complex of Drosophila comprises seven related genes encoding a special type of basic helix-loop-helix proteins, the function of which is to suppress the neural developmental fate. One of these proteins is E(spl) itself. To gain insight into the structural requirements for E(spl) function, we have expressed a large number of deletion variants in transgenic flies. Three protein domains were identified as essential for suppression of bristle development: the carboxyl-terminal tetrapeptide WRPW, the region comprising the putative helix III and helix IV, and the region between helix IV and the WRPW motif. Lack of the basic helix-loop-helix domain, helix III or IV, only partially inhibits the suppressor activity of the protein. Truncated variants that lack all the regions carboxyl-terminal to helix IV elicit the development of additional neural progenitors, and thus act as dominant-negative variants. All these results suggest that E(spl) suppresses neural development by direct interaction with other proteins, such as groucho and the proneural proteins.


Subject(s)
DNA-Binding Proteins/biosynthesis , Drosophila Proteins , Drosophila/growth & development , Gene Expression Regulation, Developmental , Insect Proteins/biosynthesis , Repressor Proteins , Animals , Animals, Genetically Modified , Basic Helix-Loop-Helix Transcription Factors , DNA-Binding Proteins/genetics , Drosophila/embryology , Drosophila/genetics , Genes, Insect , Genetic Variation , Helix-Loop-Helix Motifs , Insect Proteins/genetics , Mutagenesis, Site-Directed , Nervous System/embryology , Nervous System/growth & development , Oligodeoxyribonucleotides , Sequence Deletion
15.
Mech Dev ; 63(2): 159-71, 1997 May.
Article in English | MEDLINE | ID: mdl-9203139

ABSTRACT

We describe here the isolation and characterization of a zebrafish Delta homologue (delta D). A PCR fragment was used to obtain overlapping cDNA clones encoding a protein of 717 amino acids with all characteristic features of proteins of this family, a signal peptide, a transmembrane domain, and an extracellular region comprising the DSL domain and eight EGF-like repeats. The gene is transcribed in a complex pattern in the developing nervous system as well as in the hypoblast. Overexpression of this gene following mRNA injections leads to a reduction in the number of islet-I positive cells, which are assumed to be primary neurons, and to various defects in the adaxial mesoderm, as well as in the somites and myotomes. This suggests that delta D, and the Notch signalling pathway are involved in the differentiation of primary neurons within the neural plate, as well as in somite development.


Subject(s)
Central Nervous System/embryology , Gene Expression Regulation, Developmental , Membrane Proteins/genetics , Neurons/cytology , Somites/physiology , Zebrafish/genetics , Amino Acid Sequence , Animals , Brain Stem/metabolism , Cell Differentiation/genetics , Central Nervous System/metabolism , Cloning, Molecular , Drosophila/genetics , In Situ Hybridization , Intracellular Signaling Peptides and Proteins , Membrane Proteins/biosynthesis , Membrane Proteins/physiology , Molecular Sequence Data , Sequence Homology, Amino Acid , Zebrafish/embryology
16.
Mech Dev ; 63(1): 75-87, 1997 Apr.
Article in English | MEDLINE | ID: mdl-9178258

ABSTRACT

Classical genetics indicates that the achaete-scute gene complex (AS-C) of Drosophila promotes development of neural progenitor cells. To further analyze the function of proneural genes, we have studied the effects of Gal4-mediated expression of lethal of scute, a member of the AS-C, during embryogenesis. Expression of lethal of scute forces progenitor cells of larval internal sensory organs, which are normally committed to this fate independently of the activity of the AS-C, to take on features of external sensory organs. Supernumerary neural cells can be induced ectopically only if daughterless is overexpressed, either alone or together with lethal of scute: cells of the amnioserosa and the hindgut then express neuronal markers. Furthermore, cells of the proctodeal anlage, which normally lack neural competence, acquire the ability to develop as neuroblasts following transplantation into the neuroectoderm. We show here that activated Notch prevents the cells of the neuroectoderm from forming extra neural tissue when they express an excess of proneural proteins. Under the present conditions, lateral inhibition is thus dominant over the activity of proneural genes.


Subject(s)
DNA-Binding Proteins/biosynthesis , Drosophila Proteins , Drosophila melanogaster/embryology , Gene Expression Regulation, Developmental , Neurons/physiology , Nuclear Proteins/biosynthesis , Saccharomyces cerevisiae Proteins , Transcription Factors/biosynthesis , Animals , Basic Helix-Loop-Helix Transcription Factors , Biomarkers , Chromosome Mapping , Drosophila melanogaster/genetics , Embryonic Induction , Fungal Proteins/biosynthesis , Genes, Insect , Helix-Loop-Helix Motifs , Insect Hormones/biosynthesis , Nervous System/embryology , Recombinant Fusion Proteins/biosynthesis
18.
Curr Biol ; 7(11): R726-8, 1997 Nov 01.
Article in English | MEDLINE | ID: mdl-9382791

ABSTRACT

The cytoplasmic determinants Numb and Prospero are distributed asymmetrically into the daughter cells of Drosophila neuroblasts. The proteins encoded by the genes inscuteable, staufen and miranda are involved in the localisation of Prospero.


Subject(s)
Drosophila Proteins , Drosophila/embryology , Neurons/cytology , Neurons/physiology , Transcription Factors , Animals , Cell Division/genetics , Juvenile Hormones/genetics , Nerve Tissue Proteins/genetics , Nuclear Proteins/genetics
19.
Dev Genes Evol ; 207(3): 156-166, 1997 Aug.
Article in English | MEDLINE | ID: mdl-27747413

ABSTRACT

GROUCHO is a Drosophila nuclear protein with structural similarity to the transcriptional repressor TUP1. Drosophila GROUCHO forms complexes with bHLH proteins of the HAIRY-ENHANCER OF SPLIT [-E(SPL)] family, that then act as repressors, for example downstream of the NOTCH signalling pathway. We describe the isolation and sequence of two zebrafish GROUCHO homologues and the pattern of transcript distribution during embryogenesis. Both GRO1 and GRO2 exhibit all sequence features characteristic of the GROUCHO family and, with 79% sequence similarity at the DNA level, can be considered as orthologues of the human GROUCHO homologue TLE3. RNA in situ hybridization shows a distinct pattern of transcript distribution for both genes during embryogenesis suggestive of their participation in neurogenesis and somitogenesis.

20.
Development ; 122(7): 2071-8, 1996 Jul.
Article in English | MEDLINE | ID: mdl-8681788

ABSTRACT

her1 is a zebrafish cDNA encoding a bHLH protein with all features characteristic of members of the Drosophila HAIRY-E(SPL) family. During late gastrulation stages, her1 is expressed in the epibolic margin and in two distinct transverse bands of hypoblastic cells behind the epibolic front. After completion of epiboly, this pattern persists essentially unchanged through postgastrulation stages; the marginal domain is incorporated in the tail bud and, depending on the time point, either two or three paired bands of expressing cells are present within the paraxial presomitic mesoderm separated by regions devoid of transcripts. Labelling of cells within the her1 expression domains with fluorescein-dextran shows that the cells in the epibolic margin and the tail bud are not allocated to particular somites. However, allocation of cells to somites occurs between the marginal expression domain and the first expression band, anterior to it. Moreover, the her1 bands, and the intervening non-expressing zones, each represents the primordium of a somite. This expression pattern is highly reminiscent of that of Drosophila pair-rule genes. A possible participation of her1 in functions related to somite formation is discussed.


Subject(s)
Gene Expression Regulation, Developmental/genetics , Homeodomain Proteins/genetics , Insect Hormones/genetics , Insect Proteins , Repressor Proteins , Transcription Factors/genetics , Zebrafish/genetics , Animals , Basic Helix-Loop-Helix Transcription Factors , DNA, Complementary/analysis , Gastrula/cytology , In Situ Hybridization , Mesoderm/cytology , Mesoderm/physiology , Morphogenesis/genetics , Transcription, Genetic , Zebrafish/embryology
SELECTION OF CITATIONS
SEARCH DETAIL
...