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 ; 77(9): 1101-1113, 2017 09.
Article in English | MEDLINE | ID: mdl-28371371

ABSTRACT

Rett syndrome is a severe neurodevelopmental disorder. It is caused by a mutation in methyl-CpG binding protein 2 (MecP2), a transcriptional regulator that recruits protein complexes involved in histone modification and chromatin remodeling. However, the role of Mecp2 in Rett syndrome remains unclear. In this study, we investigated the function of Mecp2 in neuronal development using zebrafish embryos. Mecp2 expression was detected ubiquitously in the central nervous system and muscles at 28 h postfertilization (hpf). We injected an antisense morpholino oligonucleotide (AMO) to induce Mecp2 knockdown phenotype. In mecp2 morphants (embryos with Mecp2 knockdown by AMO) at 28 and 72 hpf, we found an increase in abnormal axonal branches of caudal primary motor neurons and a decrease in motor activity. In mecp2 morphants at 24 hpf, we observed an increase in the expression of an mecp2 downstream candidate gene, brain derived neurotrophic factor (bdnf). In mecp2 morphants at 72 hpf, the presynaptic area stained by an anti-SV2 antibody was increased at the neuromuscular junction (NMJ). Interestingly, the size of SV2-positive presynaptic area at the NMJ was also increased following bdnf mRNA injection, while it was normalized in a double knockdown of mecp2 and bdnf. These results imply that Mecp2 is an important functional regulator of bdnf gene expression during neural circuit formation in zebrafish embryo. © 2017 Wiley Periodicals, Inc. Develop Neurobiol 77: 1101-1113, 2017.


Subject(s)
Gene Expression Regulation, Developmental/physiology , Methyl-CpG-Binding Protein 2/metabolism , Motor Neurons/cytology , Neuromuscular Junction/metabolism , Neuronal Outgrowth/physiology , Age Factors , Animals , Brain-Derived Neurotrophic Factor/metabolism , Cell Differentiation , Embryo, Nonmammalian , Gene Expression Regulation, Developmental/drug effects , Larva , Methyl-CpG-Binding Protein 2/genetics , Motor Activity/drug effects , Motor Activity/physiology , Motor Neurons/drug effects , Neuromuscular Junction/drug effects , Neuronal Outgrowth/drug effects , Oligonucleotides, Antisense/pharmacology , Physical Stimulation , Presynaptic Terminals/drug effects , Presynaptic Terminals/physiology , RNA, Messenger/metabolism , RNA, Messenger/pharmacology , Tubulin/metabolism , Zebrafish
2.
Dev Neurobiol ; 77(4): 474-482, 2017 04.
Article in English | MEDLINE | ID: mdl-27480480

ABSTRACT

Adult neurogenesis attracts broad attention as a possible cure for neurological disorders. However, its regulatory mechanism is still unclear. Therefore, they have been studying the cell proliferation mechanisms of neural stem cells (NSCs) using zebrafish, which have high regenerative potential in the adult brain. The presence of neuroepithelial-type NSCs in the optic tectum of adult zebrafish has been previously reported. In the present study, it was first confirmed that NSCs in the optic tectum decrease or increase in proportion to projection of the optic nerves from the retina. At 4 days after optic nerve crush (ONC), BrdU-positive cells decreased in the optic tectum's operation side. In contrast, at 3 weeks after ONC, BrdU-positive cells increased in the optic tectum's operation side. To study the regulatory mechanisms, they focused on the BDNF/TrkB system as a regulatory factor in the ONC model. It was found that bdnf was mainly expressed in the periventricular gray zone (PGZ) of the optic tectum by using in situ hybridization. Interestingly, expression level of bdnf significantly decreased in the optic tectum at 4 days after ONC, and its expression level tended to increase at 3 weeks after ONC. They conducted rescue experiments using a TrkB agonist and confirmed that decrease of NSC proliferation in the optic tectum by ONC was rescued by TrkB signal activation, suggesting stimuli-dependent regulation of NSC proliferation in the optic tectum of adult zebrafish. © 2016 Wiley Periodicals, Inc. Develop Neurobiol 77: 419-437, 2017.


Subject(s)
Brain-Derived Neurotrophic Factor/metabolism , Cell Proliferation/physiology , Neural Stem Cells/physiology , Neurogenesis/physiology , Optic Nerve Injuries , Receptor, trkB/metabolism , Superior Colliculi/physiology , Zebrafish Proteins/metabolism , Animals , Disease Models, Animal , Neural Stem Cells/cytology , Receptor, trkB/agonists , Superior Colliculi/cytology , Zebrafish
3.
Genesis ; 53(7): 431-9, 2015 Jul.
Article in English | MEDLINE | ID: mdl-26045148

ABSTRACT

The zebrafish brain can continue to produce new neurons in widespread neurogenic brain regions throughout life. In contrast, neurogenesis in the adult mammalian brain is restricted to the subventricular zone (SVZ) and dentate gyrus (DG). In neurogenic regions in the adult brain, radial glial cells (RGCs) are considered to function as neural stem cells (NSCs). We generated a Tg(gfap:Gal4FF) transgenic zebrafish line, which enabled us to express specific genes in RGCs. To study the function of RGCs in neurogenesis in the adult zebrafish brain, we also generated a Tg(gfap: Gal4FF; UAS:nfsB-mcherry) transgenic zebrafish line, which allowed us to induce cell death exclusively within RGCs upon addition of metronidazole (Mtz) to the media. RGCs expressing nitroreductase were specifically ablated by the Mtz treatment, decreasing the number of proliferative RGCs. Using the Tg(gfap:Gal4FF; UAS:nfsB-mcherry) transgenic zebrafish line, we found that RGCs were specifically ablated in the adult zebrafish telencephalon. The Tg(gfap:Gal4FF) line could be useful to study the function of RGCs.


Subject(s)
Brain/cytology , Ependymoglial Cells/cytology , Ablation Techniques/methods , Animals , Animals, Genetically Modified , Brain/metabolism , Dentate Gyrus/cytology , Dentate Gyrus/metabolism , Ependymoglial Cells/metabolism , Glial Fibrillary Acidic Protein/biosynthesis , Glial Fibrillary Acidic Protein/genetics , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Neurogenesis/physiology , Zebrafish
4.
Dev Dyn ; 243(11): 1401-15, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25091230

ABSTRACT

BACKGROUND: Valproic acid (VPA) has been used to treat epilepsy and bipolar disorder. Several reports have demonstrated that VPA functions as a histone deacetylase (HDAC) inhibitor. While VPA is known to cause teratogenic changes in the embryonic zebrafish brain, its effects on neural stem cells (NSCs) in both the embryonic and adult zebrafish are not well understood. RESULTS: In this study, we observed a proliferative effect of VPA on NSCs in the embryonic hindbrain. In contrast, VPA reduced cell proliferation in the adult zebrafish optic tectum. Treatment with HDAC inhibitors showed a similar inhibitory effect on cell proliferation in the adult zebrafish optic tectum, suggesting that VPA reduces cell proliferation through HDAC inhibition. Cell cycle progression was also suppressed in the optic tectum of the adult zebrafish brain because of HDAC inhibition. Recent studies have demonstrated that HDAC inhibits the Notch signaling pathway; hence, adult zebrafish were treated with a Notch inhibitor. This increased the number of proliferating cells in the adult zebrafish optic tectum with down-regulated expression of her4, a target of Notch signaling. CONCLUSIONS: These results suggest that VPA inhibits HDAC activity and upregulates Notch signaling to reduce cell proliferation in the optic tectum of adult zebrafish.


Subject(s)
Cell Proliferation/drug effects , Histone Deacetylase Inhibitors/pharmacology , Neural Stem Cells/drug effects , Superior Colliculi/cytology , Valproic Acid/pharmacology , Zebrafish/physiology , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Blotting, Western , Bromodeoxyuridine , Cell Cycle/drug effects , Cell Cycle/physiology , Cell Proliferation/physiology , DNA Primers/genetics , Gene Expression Regulation/drug effects , Immunohistochemistry , In Situ Hybridization, Fluorescence , In Situ Nick-End Labeling , Real-Time Polymerase Chain Reaction , Receptors, Notch/metabolism , Signal Transduction/drug effects , Superior Colliculi/drug effects , Zebrafish/metabolism , Zebrafish Proteins/metabolism
5.
Dev Neurobiol ; 73(12): 911-20, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23929741

ABSTRACT

Dpysls (CRMPs) that were initially identified as mediator proteins of Semaphorin3a (Sema3a) signaling are involved in neuronal polarity and axon elongation in cultured neurons. Previous studies have shown that knockdown of neuropilin1a, one of the sema3a receptors, exhibited ectopic primary motor neurons (PMNs) outside of the spinal cord in zebrafish. However, downstream molecules of sema3a signaling involved in the positioning of motor neurons are largely unknown. Here, we addressed the role of Dpysl2 (CRMP2) and Dpysl3 (CRMP4) in the positioning of PMNs in the zebrafish spinal cord. We found that the knockdown of dpysls by antisense morpholino oligonucleotides (AMO) causes abnormal positioning of caudal primary (CaP) motor neurons outside the spinal cord. The knockdown of cdk5 and dyrk2 by AMO also caused similar phenotype in the positioning of CaP motor neurons, and this phenotype was rescued by co-injection of phosphorylation-mimic type dpysl2 mRNA. These results suggest that the phosphorylation of Dpysl2 and Dpysl3 by Cdk5 and Dyrk2 is required for correct positioning of CaP motor neurons in the zebrafish spinal cord.


Subject(s)
Gene Expression Regulation, Developmental/genetics , Motor Neurons/metabolism , Nerve Tissue Proteins/metabolism , Spinal Cord/metabolism , Zebrafish Proteins/metabolism , Zebrafish/metabolism , Animals , Cyclin-Dependent Kinase 5/genetics , Cyclin-Dependent Kinase 5/metabolism , Gene Knockdown Techniques/methods , Nerve Tissue Proteins/genetics , Phosphorylation/physiology , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Protein-Tyrosine Kinases/genetics , Protein-Tyrosine Kinases/metabolism , Semaphorin-3A/genetics , Semaphorin-3A/metabolism , Signal Transduction/genetics , Signal Transduction/physiology , Zebrafish/genetics , Zebrafish Proteins/genetics
6.
Dev Dyn ; 242(2): 189-200, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23203913

ABSTRACT

BACKGROUND: Sphingolipids represent a major class of lipids which both serve as structural components of membranes and as bioactive molecules involved in lipid signaling. Ceramide synthases (cers) reside in the center of sphingolipid metabolism by producing ceramide through de novo synthesis or degradative pathways. While the six mammalian cers family members have been extensively studied in cell culture and in adult tissues, a systematic analysis of cers expression and function during embryogenesis is still lacking. RESULTS: Using bioinformatic and phylogenetic analysis, we identified nine highly conserved homologs of the vertebrate cers gene family in the zebrafish genome. A systematic expression analysis throughout five developmental stages indicates that, whereas until 48 hours post fertilization most zebrafish cers homologs are expressed in distinct patterns, e.g., in the intermediate cell mass and the pronephric duct, they show a highly overlapping expression during later stages of embryonic development, mostprominently in the developing brain. CONCLUSIONS: In this study, the expression of the cers gene homologs is comprehensively analyzed for the first time during vertebrate embryogenesis. Our data indicate that each embryonic tissue has a unique profile of cers expression during zebrafish embryogenesis suggesting tissue-specific profiles of ceramides and their derivatives.


Subject(s)
Gene Expression Regulation, Developmental/physiology , Gene Expression Regulation, Enzymologic/physiology , Multigene Family/genetics , Oxidoreductases/genetics , Oxidoreductases/metabolism , Zebrafish/embryology , Zebrafish/genetics , Animals , Brain/metabolism , Computational Biology , Gene Expression Regulation, Developmental/genetics , Gene Expression Regulation, Enzymologic/genetics , In Situ Hybridization , Likelihood Functions , Models, Genetic , Organ Specificity/genetics , Phylogeny , Zebrafish/metabolism
7.
Dev Biol ; 370(2): 223-36, 2012 Oct 15.
Article in English | MEDLINE | ID: mdl-22898304

ABSTRACT

Dpysl2 (CRMP2) and Dpysl3 (CRMP4) are involved in neuronal polarity and axon elongation in cultured neurons. These proteins are expressed in various regions of the developing nervous system, but their roles in vivo are largely unknown. In dpysl2 and dpysl3 double morphants, Rohon-Beard (RB) primary sensory neurons that were originally located bilaterally along the midline shifted their position to a more medial location in the dorsal-most part of spinal cord. A similar phenotype was observed in the cdk5 and dyrk2 double morphants. Dpysl2 and Dpysl3 phosphorylation mimics recovered this phenotype. Cell transplantation analysis demonstrated that this ectopic RB cell positioning was non-cell autonomous and correlated with the abnormal position of neural crest cells (NCCs), which also occupied the dorsal-most part of the spinal cord during the neural rod formation stage. The cell position of other interneuron and motor neurons within the central nervous system was normal in these morphants. These results suggest that the phosphorylation of Dpysl2 and Dpysl3 by Cdk5 and DYRK2 is required for the proper positioning of RB neurons and NCCs during neurulation in zebrafish embryos.


Subject(s)
Cyclin-Dependent Kinase 5/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Nerve Tissue Proteins/metabolism , Neural Crest/metabolism , Neurulation , Protein Serine-Threonine Kinases/metabolism , Protein-Tyrosine Kinases/metabolism , Zebrafish Proteins/metabolism , Zebrafish/embryology , Animals , Neural Crest/cytology , Neurons/metabolism , Phosphorylation , Zebrafish/metabolism , Dyrk Kinases
8.
Neuron ; 69(2): 215-30, 2011 Jan 27.
Article in English | MEDLINE | ID: mdl-21262462

ABSTRACT

How the mitosis of neuroepithelial stem cells is restricted to the apical ventricular area remains unclear. In zebrafish, the mosaic eyes(rw306) (moe/epb41l5(rw306)) mutation disrupts the interaction between the putative adaptor protein Moe and the apicobasal polarity regulator Crumbs (Crb), and impairs the maintenance of neuroepithelial apicobasal polarity. While Crb interacts directly with Notch and inhibits its activity, Moe reverses this inhibition. In the moe(rw306) hindbrain, Notch activity is significantly reduced, and the number of cells that proliferate basally away from the apical area is increased. Surprisingly, activation of Notch in the moe(rw306) mutant rescues not only the basally localized proliferation but also the aberrant neuroepithelial apicobasal polarity. We present evidence that the Crb⋅Moe complex and Notch play key roles in a positive feedback loop to maintain the apicobasal polarity and the apical-high basal-low gradient of Notch activity in neuroepithelial cells, both of which are essential for their apically restricted mitosis.


Subject(s)
Cell Polarity/physiology , Eye Proteins/metabolism , Mitosis/physiology , Nerve Tissue Proteins/metabolism , Neuroepithelial Cells , Receptors, Notch/metabolism , Zebrafish Proteins/metabolism , Animals , Animals, Genetically Modified , Cell Movement/physiology , Embryonic Development/physiology , Eye Proteins/genetics , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Motor Neurons/cytology , Motor Neurons/physiology , Mutation , Nerve Tissue Proteins/genetics , Neuroepithelial Cells/cytology , Neuroepithelial Cells/physiology , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Notch/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Stem Cells/cytology , Stem Cells/physiology , Zebrafish/anatomy & histology , Zebrafish/physiology , Zebrafish Proteins/genetics
9.
Dev Dyn ; 240(1): 9-22, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21108318

ABSTRACT

We isolated a novel zebrafish mutant, lullaby (llb), and showed that the llb locus encodes the zebrafish orthologue of isl1. Rohon-Beard (RB) primary sensory neurons are multipolar neurons that extend their central axons longitudinally within the spinal cord and also extend their peripheral axons under the skin. In llb embryos, the outgrowth of the peripheral axons of RB neurons was selectively impaired, which correlated with down-regulation of the expression of dihydropyrimidinase-like 3 (dpysl3, also known as collapsin response mediator protein 4, crmp4). Antisense morpholino oligonucleotide (AMO)-mediated knockdown of dpysl3 inhibited the outgrowth of the peripheral axons of RB neurons, and semaphorin 3d (sema3d) AMO enhanced this effect. These data indicate that Dpysl3 is cooperating with Sema3d in the peripheral axon outgrowth, and Isl1 is required for the selective outgrowth of the peripheral axons of RB neurons by maintaining the expression of dpysl3.


Subject(s)
Axons/physiology , Homeodomain Proteins/physiology , Nerve Tissue Proteins/physiology , Sensory Receptor Cells/physiology , Zebrafish Proteins/physiology , Amino Acid Sequence , Animals , Animals, Genetically Modified , Axons/metabolism , Base Sequence , Embryo, Nonmammalian , Gene Expression Regulation, Developmental , Gene Knockdown Techniques , Genetic Loci , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , LIM-Homeodomain Proteins , Models, Biological , Molecular Sequence Data , Motor Neurons/metabolism , Motor Neurons/physiology , Mutation/physiology , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Sensory Receptor Cells/metabolism , Sequence Homology , Spinal Nerves/abnormalities , Spinal Nerves/embryology , Spinal Nerves/metabolism , Transcription Factors , Zebrafish/embryology , Zebrafish/genetics , Zebrafish/physiology , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism
10.
Dev Biol ; 342(1): 26-38, 2010 Jun 01.
Article in English | MEDLINE | ID: mdl-20346355

ABSTRACT

In the adult teleost brain, proliferating cells are observed in a broad area, while these cells have a restricted distribution in adult mammalian brains. In the adult teleost optic tectum, most of the proliferating cells are distributed in the caudal margin of the periventricular gray zone (PGZ). We found that the PGZ is largely divided into 3 regions: 1 mitotic region and 2 post-mitotic regions-the superficial and deep layers. These regions are distinguished by the differential expression of several marker genes: pcna, sox2, msi1, elavl3, gfap, fabp7a, and s100beta. Using transgenic zebrafish Tg (gfap:GFP), we found that the deep layer cells specifically express gfap:GFP and have a radial glial morphology. We noted that bromodeoxyuridine (BrdU)-positive cells in the mitotic region did not exhibit glial properties, but maintained neuroepithelial characteristics. Pulse chase experiments with BrdU-positive cells revealed the presence of self-renewing stem cells within the mitotic region. BrdU-positive cells differentiate into glutamatergic or GABAergic neurons and oligodendrocytes in the superficial layer and into radial glial cells in the deep layer. These results demonstrate that the proliferating cells in the PGZ contribute to neuronal and glial lineages to maintain the structure of the optic tectum in adult zebrafish.


Subject(s)
Brain/metabolism , Neuroglia/metabolism , Neurons/metabolism , Stem Cells/metabolism , Superior Colliculi/metabolism , Adult , Animals , Animals, Genetically Modified , Brain/cytology , Bromodeoxyuridine/metabolism , Cell Differentiation/genetics , Cell Differentiation/physiology , Glial Fibrillary Acidic Protein/genetics , Glial Fibrillary Acidic Protein/metabolism , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Multipotent Stem Cells/metabolism , Neuroglia/cytology , Neurons/cytology , Stem Cells/cytology , Stem Cells/physiology , Zebrafish/genetics , Zebrafish/metabolism
11.
Development ; 136(10): 1653-63, 2009 May.
Article in English | MEDLINE | ID: mdl-19369395

ABSTRACT

The molecular mechanisms by which neurons migrate and accumulate to form the neural layers and nuclei remain unclear. The formation of vagus motor nuclei in zebrafish embryos is an ideal model system in which to address this issue because of the transparency of the embryos and the availability of established genetic and molecular biological techniques. To determine the genes required for the formation of the vagus motor nuclei, we performed N-ethyl-N-nitrosourea-based mutant screening using a zebrafish line that expresses green fluorescent protein in the motor neurons. In wild-type embryos, the vagus motor neuron progenitors are born in the ventral ventricular zone, then migrate tangentially in the dorsolateral direction, forming the nuclei. However, in towhead (twd(rw685)) mutant embryos, the vagus motor neuron progenitors stray medially away from the normal migratory pathway and fail to stop in the right location. The twd(rw685) mutant has a defect in the GDP-mannose 4,6 dehydratase (gmds) gene, which encodes a key enzyme in the fucosylation pathway. Levels of fucosylated glycans were markedly and specifically reduced in twd(rw685) mutant embryos. Cell transplantation analysis revealed that GMDS is not essential in the vagus motor neuron progenitors for correct formation of the vagus motor nuclei, but is required in the neuroepithelial cells that surround the progenitors. Together, these findings suggest that fucosylated glycans expressed in neuroepithelial cells are required to guide the migration of vagus motor neuron progenitors.


Subject(s)
Motor Neurons/physiology , Neuroepithelial Cells/physiology , Polysaccharides/physiology , Rhombencephalon/embryology , Stem Cells/physiology , Vagus Nerve/physiology , Zebrafish/physiology , Amino Acid Sequence , Animals , Animals, Genetically Modified , Body Patterning , Cell Movement , Embryo, Nonmammalian/cytology , Embryo, Nonmammalian/physiology , Green Fluorescent Proteins/biosynthesis , Green Fluorescent Proteins/genetics , Hydro-Lyases/genetics , Molecular Sequence Data , Mutation , Rhombencephalon/cytology , Vagus Nerve/cytology , Vagus Nerve/embryology , Zebrafish/embryology
12.
Mech Dev ; 125(11-12): 932-46, 2008.
Article in English | MEDLINE | ID: mdl-18824093

ABSTRACT

In mammals, the blockade of the phototransduction cascade causes loss of vision and, in some cases, degeneration of photoreceptors. However, the molecular mechanisms that link phototransduction with photoreceptor degeneration remain to be elucidated. Here, we report that a mutation in the gene encoding a central effector of the phototransduction cascade, cGMP phosphodiesterase 6alpha'-subunit (PDE6alpha'), affects not only the vision but also the survival of cone photoreceptors in zebrafish. We isolated a zebrafish mutant, called eclipse (els), which shows no visual behavior such as optokinetic response (OKR). The cloning of the els mutant gene revealed that a missense mutation occurred in the pde6alpha' gene, resulting in a change in a conserved amino acid. The PDE6 expressed in rod photoreceptors is a heterotetramer comprising two closely related similar hydrolytic alpha and beta subunits and two identical inhibitory gamma subunits, while the PDE6 expressed in cone photoreceptors consists of two homodimers of alpha' subunits, each with gamma subunits. The els mutant displays no visual response to bright light, where cones are active, but shows relatively normal OKR to dim light, where only rods function, suggesting that only the cone-specific phototransduction pathway is disrupted in the els mutant. Furthermore, in the els mutant, cones are selectively eliminated but rods are retained at the adult stage, suggesting that cones undergo a progressive degeneration in the els mutant retinas. Taken together, these data suggest that PDE6alpha' activity is important for the survival of cones in zebrafish.


Subject(s)
Cyclic Nucleotide Phosphodiesterases, Type 6/genetics , Gene Expression Regulation , Mutation, Missense , Retinal Cone Photoreceptor Cells/pathology , Retinal Degeneration/genetics , Alleles , Amino Acid Sequence , Animals , Cyclic Nucleotide Phosphodiesterases, Type 6/physiology , Humans , Light Signal Transduction , Molecular Sequence Data , Retinal Cone Photoreceptor Cells/physiology , Retinal Rod Photoreceptor Cells/pathology , Retinal Rod Photoreceptor Cells/physiology , Sequence Homology, Amino Acid , Vision, Ocular , Zebrafish
13.
Development ; 134(18): 3259-69, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17699608

ABSTRACT

In zebrafish embryos, the axons of the posterior trigeminal (Vp) and facial (VII) motoneurons project stereotypically to a small number of target muscles derived from the first and second branchial arches (BA1, BA2). Use of the Islet1 (Isl1)-GFP transgenic line enabled precise real-time observations of the growth cone behaviour of the Vp and VII motoneurons within BA1 and BA2. Screening for N-ethyl-N-nitrosourea-induced mutants identified seven distinct mutations affecting different steps in the axonal pathfinding of these motoneurons. The class 1 mutations caused severe defasciculation and abnormal pathfinding in both Vp and VII motor axons before they reached their target muscles in BA1. The class 2 mutations caused impaired axonal outgrowth of the Vp motoneurons at the BA1-BA2 boundary. The class 3 mutation caused impaired axonal outgrowth of the Vp motoneurons within the target muscles derived from BA1 and BA2. The class 4 mutation caused retraction of the Vp motor axons in BA1 and abnormal invasion of the VII motor axons in BA1 beyond the BA1-BA2 boundary. Time-lapse observations of the class 1 mutant, vermicelli (vmc), which has a defect in the plexin A3 (plxna3) gene, revealed that Plxna3 acts with its ligand Sema3a1 for fasciculation and correct target selection of the Vp and VII motor axons after separation from the common pathways shared with the sensory axons in BA1 and BA2, and for the proper exit and outgrowth of the axons of the primary motoneurons from the spinal cord.


Subject(s)
Axons/physiology , Embryonic Development/genetics , Facial Nerve/embryology , Receptors, Cell Surface/physiology , Trigeminal Nerve/embryology , Zebrafish Proteins/physiology , Zebrafish/embryology , Animals , Motor Neurons/physiology , Mutation , Nerve Growth Factors , Receptors, Cell Surface/genetics , Semaphorins/physiology , Zebrafish/genetics , Zebrafish Proteins/genetics
14.
Development ; 133(23): 4749-59, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17079269

ABSTRACT

Migration of neurons from their birthplace to their final target area is a crucial step in brain development. Here, we show that expression of the off-limits/frizzled3a (olt/fz3a) and off-road/celsr2 (ord/celsr2) genes in neuroepithelial cells maintains the facial (nVII) motor neurons near the pial surface during their caudal migration in the zebrafish hindbrain. In the absence of olt/fz3a expression in the neuroepithelium, nVII motor neurons extended aberrant radial processes towards the ventricular surface and mismigrated radially to the dorsomedial part of the hindbrain. Our findings reveal a novel role for these genes, distinctive from their already known functions, in the regulation of the planar cell polarity (i.e. preventing integration of differentiated neurons into the neuroepithelial layer). This contrasts markedly with their reported role in reintegration of neuroepithelial daughter cells into the neuroepithelial layer after cell division.


Subject(s)
Cadherins/genetics , Frizzled Receptors/genetics , Rhombencephalon/embryology , Zebrafish Proteins/genetics , Zebrafish/embryology , Zebrafish/genetics , Animals , Animals, Genetically Modified , Cadherins/metabolism , Cell Movement/genetics , Cell Movement/physiology , Frizzled Receptors/metabolism , Gene Expression Regulation, Developmental , Motor Neurons/cytology , Motor Neurons/metabolism , Mutation , Neuroepithelial Cells/cytology , Neuroepithelial Cells/metabolism , Rhombencephalon/metabolism , Zebrafish/metabolism , Zebrafish Proteins/metabolism
15.
Development ; 132(10): 2273-85, 2005 May.
Article in English | MEDLINE | ID: mdl-15829519

ABSTRACT

In the developing vertebrate hindbrain, the characteristic trajectory of the facial (nVII) motor nerve is generated by caudal migration of the nVII motor neurons. The nVII motor neurons originate in rhombomere (r) 4, and migrate caudally into r6 to form the facial motor nucleus. In this study, using a transgenic zebrafish line that expresses green fluorescent protein (GFP) in the cranial motor neurons, we isolated two novel mutants, designated landlocked (llk) and off-road (ord), which both show highly specific defects in the caudal migration of the nVII motor neurons. We show that the landlocked locus contains the gene scribble1 (scrb1), and that its zygotic expression is required for migration of the nVII motor neurons mainly in a non cell-autonomous manner. Taking advantage of the viability of the llk mutant embryos, we found that maternal expression of scrb1 is required for convergent extension (CE) movements during gastrulation. Furthermore, we show a genetic interaction between scrb1 and trilobite(tri)/strabismus(stbm) in CE. The dual roles of the scrb1 gene in both neuronal migration and CE provide a novel insight into the underlying mechanisms of cell movement in vertebrate development.


Subject(s)
Cell Movement/physiology , Facial Nerve/embryology , Gene Expression Regulation, Developmental , Membrane Proteins/genetics , Membrane Proteins/metabolism , Motor Neurons/cytology , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism , Zebrafish/embryology , Animals , Base Sequence , Cell Movement/genetics , Chromosome Mapping , Crosses, Genetic , DNA Primers , Gastrula/metabolism , Green Fluorescent Proteins/metabolism , Immunohistochemistry , In Situ Hybridization , Molecular Sequence Data , Mutation/genetics , Oligonucleotides, Antisense , Reverse Transcriptase Polymerase Chain Reaction , Rhombencephalon/embryology , Sequence Analysis, DNA
16.
Curr Biol ; 15(5): 480-8, 2005 Mar 08.
Article in English | MEDLINE | ID: mdl-15753045

ABSTRACT

The Hedgehog (Hh) signal plays a pivotal role in induction of ventral neuronal and muscle cell types around the midline during vertebrate development [1]. We report that the gene disrupted in zebrafish you mutants, in which Hh signaling is impaired, encodes the secreted matrix protein Scube2. Consistently, epistasis analyses suggested that Scube2 functions upstream of Hh ligands or through a parallel pathway. In addition, overexpression analyses suggested that Scube2 is an essential, but a permissive, mediator of Hh signaling in zebrafish embryos. Surprisingly, the you gene is expressed in the dorsal neural tube, raising the possibility that Scube2 could indirectly act via a long-range regulator of Hh signaling. The dorsal Bmps have a long-range and opposing influence on Hh signaling [2-5]. We show that neural plate patterning is affected in you mutants in a way that is consistent with the aberrant long-range action of a Bmp-dependent signal. We further show that Bmp activity can be attenuated by the coexpression of Scube2. Our data support the idea that Scube2 can modulate the long-range action of Bmp-dependent signaling in the neural tube and somites.


Subject(s)
Body Patterning/physiology , Central Nervous System/embryology , Extracellular Matrix Proteins/metabolism , Phenotype , Signal Transduction/physiology , Trans-Activators/metabolism , Zebrafish Proteins/metabolism , Zebrafish/metabolism , Amino Acid Sequence , Animals , Base Sequence , Blotting, Western , Bone Morphogenetic Proteins/metabolism , Chromosome Mapping , DNA Primers , Epistasis, Genetic , Extracellular Matrix Proteins/genetics , Gene Components , Genotype , Hedgehog Proteins , In Situ Hybridization , Molecular Sequence Data , Mutation/genetics , Sequence Alignment , Sequence Analysis, DNA , Zebrafish/embryology , Zebrafish Proteins/genetics
17.
Development ; 130(11): 2479-94, 2003 Jun.
Article in English | MEDLINE | ID: mdl-12702661

ABSTRACT

The complex, yet highly ordered and predictable, structure of the neural retina is one of the most conserved features of the vertebrate central nervous system. In all vertebrate classes, retinal neurons are organized into laminae with each neuronal class adopting specific morphologies and patterns of connectivity. Using genetic analyses in zebrafish, we demonstrate that N-cadherin (Ncad) has several distinct and crucial functions during the establishment of retinal organization. Although the location of cell division is disorganized in embryos with reduced or no Ncad function, different classes of retinal neurons are generated. However, these neurons fail to organize into correct laminae, most probably owing to compromised adhesion between retinal cells. In addition, amacrine cells exhibit exuberant and misdirected outgrowth of neurites that contributes to severe disorganization of the inner plexiform layer. Retinal ganglion cells also exhibit defects in process outgrowth, with axons exhibiting fasciculation defects and adopting incorrect ipsilateral trajectories. At least some of these defects are likely to be due to a failure to maintain compartment boundaries between eye, optic nerve and brain. Although in vitro studies have implicated Fgf receptors in modulating the axon outgrowth promoting properties of Ncad, most aspects of the Ncad mutant phenotype are not phenocopied by treatments that block Fgf receptor function.


Subject(s)
Cadherins/metabolism , Prosencephalon/embryology , Retina/embryology , Zebrafish Proteins/metabolism , Zebrafish/embryology , Zebrafish/metabolism , Adherens Junctions/metabolism , Alleles , Animals , Base Sequence , Body Patterning , Cadherins/genetics , Cell Adhesion , Cell Division , DNA, Complementary/genetics , Mutation , Neurons/cytology , Prosencephalon/metabolism , Receptors, Fibroblast Growth Factor/metabolism , Retina/cytology , Retina/metabolism , Retinal Ganglion Cells/cytology , Retinal Ganglion Cells/metabolism , Signal Transduction , Zebrafish/genetics , Zebrafish Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...