Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Development ; 128(17): 3371-9, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11546753

ABSTRACT

In both Drosophila and vertebrates, spatially restricted expression of HOX genes is controlled by the Polycomb group (PcG) repressors. Here we characterize a novel Drosophila PcG gene, Suppressor of zeste 12 (Su(z)12). Su(z)12 mutants exhibit very strong homeotic transformations and Su(z)12 function is required throughout development to maintain the repressed state of HOX genes. Unlike most other PcG mutations, Su(z)12 mutations are strong suppressors of position-effect variegation (PEV), suggesting that Su(z)12 also functions in heterochromatin-mediated repression. Furthermore, Su(z)12 function is required for germ cell development. The Su(z)12 protein is highly conserved in vertebrates and is related to the Arabidopsis proteins EMF2, FIS2 and VRN2. Notably, EMF2 is a repressor of floral homeotic genes. These results suggest that at least some of the regulatory machinery that controls homeotic gene expression is conserved between animals and plants.


Subject(s)
Conserved Sequence , Drosophila Proteins , Drosophila melanogaster/genetics , Genes, Insect , Insect Proteins/genetics , Amino Acid Sequence , Animals , Arabidopsis/genetics , Drosophila melanogaster/embryology , Female , Gene Expression , Genes, Homeobox , Histone-Lysine N-Methyltransferase , Humans , Insect Proteins/physiology , Male , Molecular Sequence Data , Mutagenesis , Polycomb Repressive Complex 1 , Polycomb Repressive Complex 2 , Repressor Proteins , Sequence Homology, Amino Acid , Vertebrates/genetics
2.
Development ; 128(6): 993-1004, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11222153

ABSTRACT

Early in Drosophila embryogenesis, transcriptional repressors encoded by Gap genes prevent the expression of particular combinations of Hox genes in each segment. During subsequent development, those Hox genes that were initially repressed in each segment remain off in all the descendent cells, even though the Gap repressors are no longer present. This phenomenon of heritable silencing depends on proteins of the Polycomb Group (PcG) and on cis-acting Polycomb response elements (PREs) in the Hox gene loci. We have removed individual PcG proteins from proliferating cells and then resupplied these proteins after a few or several cell generations. We show that most PcG proteins are required throughout development: when these proteins are removed, Hox genes become derepressed. However, we find that resupply of at least some PcG proteins can cause re-repression of Hox genes, provided that it occurs within a few cell generations of the loss of repression. These results suggest a functional distinction between transcriptional repression and heritable silencing: in at least some contexts, Hox genes can retain the capacity to be heritably silenced, despite being transcribed and replicated. We propose that silenced Hox genes bear a heritable, molecular mark that targets them for transcriptional repression. Some PcG proteins may be required to define and propagate this mark; others may function to repress the transcription of Hox genes that bear the mark.


Subject(s)
Drosophila Proteins , Drosophila melanogaster/embryology , Drosophila melanogaster/genetics , Gene Silencing , Genes, Homeobox , Genes, Insect , Heat-Shock Proteins/genetics , Insect Proteins/metabolism , Repressor Proteins/metabolism , Animals , Animals, Genetically Modified , Binding Sites , Gene Expression Regulation, Developmental , Morphogenesis , Polycomb Repressive Complex 1 , Wings, Animal/embryology , X Chromosome
3.
Science ; 282(5395): 1897-900, 1998 Dec 04.
Article in English | MEDLINE | ID: mdl-9836641

ABSTRACT

Early in Drosophila embryogenesis, gap gene products directly repress transcription of homeotic (HOX) genes and thereby delimit HOX expression domains. Subsequently, Polycomb-group proteins maintain this repression. Currently, there is no known molecular link between gap and Polycomb-group proteins. Here, dMi-2 is identified as a protein that binds to a domain in the gap protein Hunchback that is specifically required for the repression of HOX genes. Genetic analyses show that dMi-2 participates in both Hunchback and Polycomb repression in vivo. Hence, recruitment of dMi-2 may serve as a link between repression of HOX genes by Hunchback and Polycomb proteins.


Subject(s)
Adenosine Triphosphatases , Autoantigens/genetics , Autoantigens/metabolism , Carrier Proteins/genetics , Carrier Proteins/metabolism , DNA-Binding Proteins/metabolism , Drosophila Proteins , Gene Expression Regulation, Developmental , Genes, Homeobox , Insect Proteins/metabolism , Transcription Factors/metabolism , Animals , Autoantigens/chemistry , Carrier Proteins/chemistry , DNA-Binding Proteins/genetics , Drosophila/embryology , Drosophila/genetics , Embryo, Nonmammalian/metabolism , Gene Dosage , Genes, Insect , Genetic Complementation Test , Germ Cells/metabolism , Heterozygote , Homeodomain Proteins/genetics , In Situ Hybridization , Insect Proteins/genetics , Mutation , Polycomb Repressive Complex 1 , Recombinant Fusion Proteins
4.
Development ; 123: 1-36, 1996 Dec.
Article in English | MEDLINE | ID: mdl-9007226

ABSTRACT

In a large-scale screen, we isolated mutants displaying a specific visible phenotype in embryos or early larvae of the zebrafish, Danio rerio. Males were mutagenized with ethylnitrosourea (ENU) and F2 families of single pair matings between sibling F1 fish, heterozygous for a mutagenized genome, were raised. Egg lays were obtained from several crosses between F2 siblings, resulting in scoring of 3857 mutagenized genomes. F3 progeny were scored at the second, third and sixth day of development, using a stereomicroscope. In a subsequent screen, fixed embryos were analyzed for correct retinotectal projection. A total of 4264 mutants were identified. Two thirds of the mutants displaying rather general abnormalities were eventually discarded. We kept and characterized 1163 mutants. In complementation crosses performed between mutants with similar phenotypes, 894 mutants have been assigned to 372 genes. The average allele frequency is 2.4. We identified genes involved in early development, notochord, brain, spinal cord, somites, muscles, heart, circulation, blood, skin, fin, eye, otic vesicle, jaw and branchial arches, pigment pattern, pigment formation, gut, liver, motility and touch response. Our collection contains alleles of almost all previously described zebrafish mutants. From the allele frequencies and other considerations we estimate that the 372 genes defined by the mutants probably represent more than half of all genes that could have been discovered using the criteria of our screen. Here we give an overview of the spectrum of mutant phenotypes obtained, and discuss the limits and the potentials of a genetic saturation screen in the zebrafish.


Subject(s)
Genes , Zebrafish/embryology , Zebrafish/genetics , Animals , Crosses, Genetic , Embryonic Development , Gene Expression Regulation, Developmental , Genetic Complementation Test , Male , Mutagenesis , Phenotype , Zebrafish/growth & development
5.
Development ; 123: 129-42, 1996 Dec.
Article in English | MEDLINE | ID: mdl-9007235

ABSTRACT

Tissues of the dorsal midline of vertebrate embryos, such as notochord and floor plate, have been implicated in inductive interactions that pattern the neural tube and somites. In our screen for embryonic visible mutations in the zebrafish we found 113 mutations in more than 27 genes with altered body shape, often with additional defects in CNS development. We concentrated on a subgroup of mutations in ten genes (the midline-group) that cause defective development of the floor plate. By using floor plate markers, such as the signaling molecule sonic hedgehog, we show that the schmalspur (sur) gene is needed for early floor plate development, similar to one-eyed-pinhead (oep) and the previously described cyclops (cyc) gene. In contrast to oep and cyc, sur embryos show deletions of ventral CNS tissue restricted to the mid- and hindbrain, whereas the forebrain appears largely unaffected. In the underlying mesendodermal tissue of the head, sur is needed only for development of the posterior prechordal plate, whereas oep and cyc are required for both anterior and posterior prechordal plate development. Our analysis of sur mutants suggests that defects within the posterior prechordal plate may cause aberrant development of ventral CNS structures in the mid- and hindbrain. Later development of the floor plate is affected in mutant chameleon, you-too, sonic-you, iguana, detour, schmalhans and monorail embryos; these mutants often show additional defects in tissues that are known to depend on signals from notochord and floor plate. For example, sur, con and yot mutants show reduction of motor neurons; median deletions of brain tissue are seen in sur, con and yot embryos; and cyc, con, yot, igu and dtr mutants often show no or abnormal formation of the optic chiasm. We also find fusions of the ventral neurocranium for all midline mutants tested, which may reveal a hitherto unrecognized function of the midline in influencing differentiation of neural crest cells at their destination. As a working hypothesis, we propose that midline-group genes may act to maintain proper structure and inductive function of zebrafish midline tissues.


Subject(s)
Body Patterning/genetics , Gene Expression Regulation, Developmental , Mutation , Zebrafish/anatomy & histology , Zebrafish/embryology , Animals , Axons/physiology , Brain/embryology , Brain/pathology , Embryo, Nonmammalian/anatomy & histology , Embryonic Development , Genetic Complementation Test , Mesoderm/pathology , Motor Neurons/pathology , Nervous System/embryology , Zebrafish/genetics
6.
Development ; 123: 179-90, 1996 Dec.
Article in English | MEDLINE | ID: mdl-9007239

ABSTRACT

Mutations in two genes affect the formation of the boundary between midbrain and hindbrain (MHB): no isthmus (noi) and acerebellar (ace). noi mutant embryos lack the MHB constriction, the cerebellum and optic tectum, as well as the pronephric duct. Analysis of noi mutant embryos with neuron-specific antibodies shows that the MHB region and the dorsal and ventral midbrain are absent or abnormal, but that the rostral hindbrain is unaffected with the exception of the cerebellum. Using markers that are expressed during its formation (eng, wnt1 and pax-b), we find that the MHB region is already misspecified in noi mutant embryos during late gastrulation. The tectum is initially present and later degenerates. The defect in ace mutant embryos is more restricted: MHB and cerebellum are absent, but a tectum is formed. Molecular organisation of the tectum and tegmentum is disturbed, however, since eng, wnt1 and pax-b marker gene expression is not maintained. We propose that noi and ace are required for development of the MHB region and of the adjacent mid- and hindbrain, which are thought to be patterned by the MHB region. Presence of pax-b RNA, and absence of pax-b protein, together with the observation of genetic linkage and the occurrence of a point mutation, show that noi mutations are located in the pax-b gene. pax-b is a vertebrate orthologue of the Drosophila gene paired, which is involved in a pathway of cellular interactions at the posterior compartment boundary in Drosophila. Our results confirm and extend a previous report, and show that at least one member of this conserved signalling pathway is required for formation of the boundary between midbrain and hindbrain in the zebrafish.


Subject(s)
Mesencephalon/embryology , Mutation , Rhombencephalon/embryology , Zebrafish/embryology , Zebrafish/genetics , Animals , Cell Death/genetics , Central Nervous System/embryology , Embryo, Nonmammalian/cytology , Gene Deletion , Genes , Genetic Linkage , Genetic Markers , Phenotype , Superior Colliculi/cytology , Superior Colliculi/embryology
7.
Development ; 123: 191-203, 1996 Dec.
Article in English | MEDLINE | ID: mdl-9007240

ABSTRACT

We identified four zebrafish mutants with defects in forebrain induction and patterning during embryogenesis. The four mutants define three genes: masterblind (mbl), silberblick (slb), and knollnase (kas). In mbl embryos, the anterior forebrain acquires posterior forebrain characteristics: anterior structures such as the eyes, olfactory placodes and the telencephalon are missing, whereas the epiphysis located in the posterior forebrain is expanded. In slb embryos, the extension of the embryonic axis is initially delayed and eventually followed by a partial fusion of the eyes. Finally, in kas embryos, separation of the telencephalic primordia is incomplete and dorsal midline cells fail to form a differentiated roof plate. Analysis of the mutant phenotypes indicates that we have identified genes essential for the specification of the anterior forebrain (mbl), positioning of the eyes (slb) and differentiation of the roof plate (kas). In an appendix to this study we list mutants showing alterations in the size of the eyes and abnormal differentiation of the lenses.


Subject(s)
Genes , Prosencephalon/embryology , Zebrafish/embryology , Zebrafish/genetics , Animals , Body Patterning/genetics , Cerebral Ventricles/embryology , Ectoderm/cytology , Ectoderm/physiology , Gastrula/physiology , Genetic Linkage , Mice , Mutagenesis , Nervous System/embryology , Phenotype , Telencephalon/embryology
8.
Development ; 123: 205-16, 1996 Dec.
Article in English | MEDLINE | ID: mdl-9007241

ABSTRACT

In a screen for embryonic mutants in the zebrafish a large number of mutants were isolated with abnormal brain morphology. We describe here 26 mutants in 13 complementation groups that show abnormal development of large regions of the brain. Early neurogenesis is affected in white tail (wit). During segmentation stages, homozygous wit embryos display an irregularly formed neural keel, particularly in the hindbrain. Using a variety of molecular markers, a severe increase in the number of various early differentiating neurons can be demonstrated. In contrast, late differentiating neurons, radial glial cells and some nonneural cell types, such as the neural crest-derived melanoblasts, are much reduced. Somitogenesis appears delayed. In addition, very reduced numbers of melanophores are present posterior to the mid-trunk. The wit phenotype is reminiscent of neurogenic mutants in Drosophila, such as Notch or Delta. In mutant parachute (pac) embryos the general organization of the hindbrain is disturbed and many rounded cells accumulate loosely in the hindbrain and midbrain ventricles. Mutants in a group of 6 genes, snakehead(snk), natter (nat), otter (ott), fullbrain (ful), viper (vip) and white snake (wis) develop collapsed brain ventricles, before showing signs of general degeneration. atlantis (atl), big head (bid), wicked brain (win), scabland (sbd) and eisspalte (ele) mutants have different malformation of the brain folds. Some of them have transient phenotypes, and mutant individuals may grow up to adults.


Subject(s)
Brain/embryology , Mutagenesis , Zebrafish/embryology , Zebrafish/genetics , Animals , Brain/pathology , Cell Differentiation/genetics , Cerebral Ventricles/embryology , Hyperplasia , Muscle, Skeletal/embryology , Neural Crest/cytology , Neural Crest/embryology , Neuroglia/cytology , Neurons/cytology , Phenotype , Somites/physiology
9.
Development ; 123: 229-39, 1996 Dec.
Article in English | MEDLINE | ID: mdl-9007243

ABSTRACT

Forty zebrafish mutants with localized or general neural degeneration are described. The onset and duration of degeneration and the distribution of ectopically dying cells are specific characteristics of each mutant. Mutants are classified into four groups by these parameters. Class I: late focal neural degeneration mutants. These 18 mutants have restricted cell death mainly in the tectum and the dorsal hindbrain after 36 hours. The degeneration does not spread and disappears at later stages of development. Class II: early focal neural degeneration mutants. Ten mutants in this class exhibit transient restricted degeneration affecting mainly the diencephalon, the hindbrain and the spinal cord at 20 hours. The midbrain is less affected. The degeneration shifts to the dorsal diencephalon and the tectum at 36 hours. Class III: late spreading neural degeneration mutants. The 8 mutants in this class display a degeneration that is first seen in the tectum and subsequently spreads throughout the nervous system from 36 hours on. Class IV: early general neural degeneration mutants. This class of four mutants already shows overall cell degeneration in the nervous system at the 15-somite stage. Three of the class I mutants show a change in the pattern of gene expression in the anlage of a brain structure prior to the onset of degeneration. These results suggest that focal cell death may be a useful clue for the detection of early patterning defects of the vertebrate nervous system in regions devoid of visible landmarks.


Subject(s)
Mutation , Nerve Degeneration/genetics , Zebrafish/embryology , Zebrafish/genetics , Animals , Apoptosis/genetics , Body Patterning/genetics , Genetic Markers , In Situ Hybridization , Phenotype
10.
Development ; 123: 329-44, 1996 Dec.
Article in English | MEDLINE | ID: mdl-9007253

ABSTRACT

Jaws and branchial arches together are a basic, segmented feature of the vertebrate head. Seven arches develop in the zebrafish embryo (Danio rerio), derived largely from neural crest cells that form the cartilaginous skeleton. In this and the following paper we describe the phenotypes of 109 arch mutants, focusing here on three classes that affect the posterior pharyngeal arches, including the hyoid and five gill-bearing arches. In lockjaw, the hyoid arch is strongly reduced and subsets of branchial arches do not develop. Mutants of a large second class, designated the flathead group, lack several adjacent branchial arches and their associated cartilages. Five alleles at the flathead locus all lead to larvae that lack arches 4-6. Among 34 other flathead group members complementation tests are incomplete, but at least six unique phenotypes can be distinguished. These all delete continuous stretches of adjacent branchial arches and unpaired cartilages in the ventral midline. Many show cell death in the midbrain, from which some neural crest precursors of the arches originate. lockjaw and a few mutants in the flathead group, including pistachio, affect both jaw cartilage and pigmentation, reflecting essential functions of these genes in at least two neural crest lineages. Mutants of a third class, including boxer, dackel and pincher, affect pectoral fins and axonal trajectories in the brain, as well as the arches. Their skeletal phenotypes suggest that they disrupt cartilage morphogenesis in all arches. Our results suggest that there are sets of genes that: (1) specify neural crest cells in groups of adjacent head segments, and (2) function in common genetic pathways in a variety of tissues including the brain, pectoral fins and pigment cells as well as pharyngeal arches.


Subject(s)
Branchial Region/embryology , Jaw/embryology , Mutation , Zebrafish/embryology , Zebrafish/genetics , Animals , Branchial Region/abnormalities , Extremities/embryology , Facial Bones/embryology , Hyoid Bone/embryology , Limb Deformities, Congenital , Mesencephalon/abnormalities , Mesencephalon/embryology , Mouth/embryology , Necrosis , Pharynx/abnormalities , Pharynx/embryology , Phenotype , Pigmentation/genetics , Skull/embryology , Superior Colliculi/abnormalities , Superior Colliculi/embryology
11.
Development ; 123: 345-56, 1996 Dec.
Article in English | MEDLINE | ID: mdl-9007254

ABSTRACT

In a large scale screen for mutants that affect the early development of the zebrafish, 109 mutants were found that cause defects in the formation of the jaw and the more posterior pharyngeal arches. Here we present the phenotypic description and results of the complementation analysis of mutants belonging to two major classes: (1) mutants with defects in the mandibular and hyoid arches and (2) mutants with defects in cartilage differentiation and growth in all arches. Mutations in four of the genes identified during the screen show specific defects in the first two arches and leave the more posterior pharyngeal arches largely unaffected (schmerle, sucker, hoover and sturgeon). In these mutants ventral components of the mandibular and hyoid arches are reduced (Meckel's cartilage and ceratohyal cartilage) whereas dorsal structures (palatoquadrate and hyosymplectic cartilages) are of normal size or enlarged. Thus, mutations in single genes cause defects in the formation of first and second arch structures but also differentially affect development of the dorsal and ventral structures within one arch. In 27 mutants that define at least 8 genes, the differentiation of cartilage and growth is affected. In hammerhead mutants particularly the mesodermally derived cartilages are reduced, whereas jellyfish mutant larvae are characterized by a severe reduction of all cartilaginous elements, leaving only two pieces in the position of the ceratohyal cartilages. In all other mutant larvae all skeletal elements are present, but consist of smaller and disorganized chondrocytes. These mutants also exhibit shortened heads and reduced pectoral fins. In homozygous knorrig embryos, tumor-like outgrowths of chondrocytes occur along the edges of all cartilaginous elements. The mutants presented here may be valuable tools for elucidating the genetic mechanisms that underlie the development of the mandibular and the hyoid arches, as well as the process of cartilage differentiation.


Subject(s)
Branchial Region/embryology , Cartilage/embryology , Jaw/embryology , Mutation , Zebrafish/embryology , Zebrafish/genetics , Animals , Branchial Region/abnormalities , Cartilage/abnormalities , Cartilage/pathology , Cell Division/genetics , Extracellular Matrix/pathology , Head and Neck Neoplasms/embryology , Head and Neck Neoplasms/genetics , Head and Neck Neoplasms/pathology , Larva , Phenotype , Skull/embryology
12.
Development ; 123: 369-89, 1996 Dec.
Article in English | MEDLINE | ID: mdl-9007256

ABSTRACT

Neural crest development involves cell-fate specification, proliferation, patterned cell migration, survival and differentiation. Zebrafish neural crest derivatives include three distinct chromatophores, which are well-suited to genetic analysis of their development. As part of a large-scale mutagenesis screen for embryonic/early larval mutations, we have isolated 285 mutations affecting all aspects of zebrafish larval pigmentation. By complementation analysis, we define 94 genes. We show here that comparison of their phenotypes permits classification of these mutations according to the types of defects they cause, and these suggest which process of neural crest development is probably affected. Mutations in eight genes affect the number of chromatophores: these include strong candidates for genes necessary for the processes of pigment cell specification and proliferation. Mutations in five genes remove part of the wild-type pigment pattern, and suggest a role in larval pigment pattern formation. Mutations in five genes show ectopic chromatophores in distinct sites, and may have implications for chromatophore patterning and proliferation. 76 genes affect pigment or morphology of one or more chromatophore types: these mutations include strong candidates for genes important in various aspects of chromatophore differentiation and survival. In combination with the embryological advantages of zebrafish, these mutations should permit cellular and molecular dissection of many aspects of neural crest development.


Subject(s)
Mutation , Neural Crest/embryology , Pigmentation/genetics , Zebrafish/embryology , Zebrafish/genetics , Adaptation, Physiological/genetics , Animals , Body Patterning/genetics , Cell Count , Cell Differentiation/genetics , Chromatophores/metabolism , Chromatophores/pathology , Chromatophores/physiology , Larva , Melanins/biosynthesis , Melanins/genetics , Melanophores/pathology , Pigments, Biological/genetics
13.
Development ; 120(4): 843-52, 1994 Apr.
Article in English | MEDLINE | ID: mdl-7600961

ABSTRACT

goosecoid is an immediate early gene expressed at the dorsal blastoporal lip of the Xenopus gastrula. Microinjection experiments have suggested a direct role for goosecoid in organizing the dorsoventral axis of the frog embryo. Here we characterize the zebrafish homologue of goosecoid (gsc) and compare its expression to that of Brachyury or no tail (ntl), another immediate early gene required in developing mesoderm. We show that gsc exhibits two independent phases of expression: an early one in cells anterior to the presumptive notochord, but not in cells of the notochord itself, and a later one in neural crest derivatives in the larval head. Zygotic gsc transcripts are detected soon after the midblastula transition, and at the blastula stage form a gradient with a maximum at the dorsal side. Use of gsc as a dorsal marker allowed us to demonstrate that ntl expression is initially activated at the dorsal side of the blastula. At this early stage, gsc and ntl show overlapping domains of expression and are co-expressed in cells at the dorsal midline of the early gastrula. However, gsc- and ntl-expressing cells become separated in the course of gastrulation, with gsc being expressed in the axial hypoblast (prechordal plate) anterior to the ntl-expressing presumptive notochord cells. Studies with mutant embryos suggest that gsc is independent of ntl function in vivo.


Subject(s)
DNA-Binding Proteins/genetics , Fetal Proteins/genetics , Gastrula/physiology , Homeodomain Proteins , Mesoderm/physiology , Repressor Proteins , T-Box Domain Proteins , Transcription Factors , Zebrafish/genetics , Amino Acid Sequence , Animals , Blastocyst/physiology , Blotting, Northern , Chickens , Goosecoid Protein , In Situ Hybridization , Mice , Molecular Sequence Data , Sequence Homology, Amino Acid , Xenopus , Zebrafish/embryology , Zebrafish Proteins , Zygote/physiology
14.
Cell ; 66(1): 51-63, 1991 Jul 12.
Article in English | MEDLINE | ID: mdl-1712672

ABSTRACT

The posterior group gene staufen is required both for the localization of maternal determinants to the posterior pole of the Drosophila egg and for bicoid RNA to localize correctly to the anterior pole. We report the cloning and sequencing of staufen and show that staufen protein is one of the first molecules to localize to the posterior pole of the oocyte, perhaps in association with oskar RNA. Once localized, staufen is found in the polar granules and is required to hold other polar granule components at the posterior pole. By the time the egg is laid, staufen protein is also concentrated at the anterior pole, in the same region as bicoid RNA.


Subject(s)
Drosophila Proteins , Drosophila/genetics , Genes , Insect Hormones/genetics , Oogenesis , Ovum/physiology , RNA-Binding Proteins , RNA/genetics , Amino Acid Sequence , Animals , Base Sequence , Cloning, Molecular , Drosophila/physiology , Female , Gene Expression , Molecular Sequence Data , Mutation , Restriction Mapping
SELECTION OF CITATIONS
SEARCH DETAIL
...