Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Curr Biol ; 33(11): R440-R442, 2023 06 05.
Article in English | MEDLINE | ID: mdl-37279664

ABSTRACT

Social conflict between conspecifics results in the establishment of a hierarchy composed of a winner and loser. A recent study elucidates the molecular mechanism that may underlie the behavioral switch between winner and loser states.


Subject(s)
Aggression , Social Dominance , Animals , Behavior, Animal
2.
Sci Rep ; 13(1): 4651, 2023 03 21.
Article in English | MEDLINE | ID: mdl-36944644

ABSTRACT

Since the discovery of olfactory receptor (OR) genes, their expression in non-olfactory tissues have been reported in rodents and humans. For example, mouse OR23 (mOR23) is expressed in sperm and muscle cells and has been proposed to play a role in chemotaxis and muscle migration, respectively. In addition, mouse mesencephalic dopaminergic neurons express various ORs, which respond to corresponding ligands. As the OR genes comprise the largest multigene family of G protein-coupled receptors in vertebrates (over 400 genes in human and 1000 in rodents), it has been difficult to categorize the extent of their diverse expression in non-olfactory tissues making it challenging to ascertain their function. The zebrafish genome contains significantly fewer OR genes at around 140 genes, and their expression pattern can be easily analyzed by carrying out whole mount in situ hybridization (ISH) assay in larvae. In this study, we found that 31 out of 36 OR genes, including or104-2, or108-1, or111-1, or125-4, or128-1, or128-5, 133-4, or133-7, or137-3 are expressed in various tissues, including the trunk, pharynx, pancreas and brain in the larvae. In addition, some OR genes are expressed in distinct brain regions such as the hypothalamus and the habenula in a dynamic temporal pattern between larvae, juvenile and adult zebrafish. We further confirmed that OR genes are expressed in non-olfactory tissues by RT-PCR in larvae and adults. These results indicate tight regulation of OR gene expression in the brain in a spatial and temporal manner and that the expression of OR genes in non-olfactory tissues are conserved in vertebrates. This study provides a framework to start investigating the function of ORs in the zebrafish brain.


Subject(s)
Olfactory Receptor Neurons , Receptors, Odorant , Animals , Male , Mesencephalon/metabolism , Olfactory Receptor Neurons/metabolism , Receptors, Odorant/genetics , Receptors, Odorant/metabolism , Semen/metabolism , Spermatozoa/metabolism , Zebrafish/genetics , Zebrafish/metabolism
4.
Curr Biol ; 31(20): 4584-4595.e4, 2021 10 25.
Article in English | MEDLINE | ID: mdl-34478646

ABSTRACT

In the developing central nervous system, electrical signaling is thought to rely exclusively on differentiating neurons as they acquire the ability to generate and propagate action potentials. Accordingly, neuroepithelial progenitors (NEPs), which give rise to all neurons and glial cells during development, have been reported to remain electrically passive. Here, we investigated the physiological properties of NEPs at the onset of spontaneous neural activity (SNA) initiating motor behavior in mouse embryonic spinal cord. Using patch-clamp recordings, we discovered that spinal NEPs exhibit spontaneous membrane depolarizations during episodes of SNA. These rhythmic depolarizations exhibited a ventral-to-dorsal gradient with the highest amplitude located in the floor plate, the ventral-most part of the neuroepithelium. Paired recordings revealed that NEPs are coupled via gap junctions and form an electrical syncytium. Although other NEPs were electrically passive, we discovered that floor-plate NEPs generated large Na+/Ca2+ action potentials. Unlike in neurons, floor-plate action potentials relied primarily on the activation of voltage-gated T-type calcium channels (TTCCs). In situ hybridization showed that all 3 known subtypes of TTCCs are predominantly expressed in the floor plate. During SNA, we found that acetylcholine released by motoneurons rhythmically triggers floor-plate action potentials by acting through nicotinic acetylcholine receptors. Finally, by expressing the genetically encoded calcium indicator GCaMP6f in the floor plate, we demonstrated that neuroepithelial action potentials are associated with calcium waves and propagate along the entire length of the spinal cord. Our work reveals a novel physiological mechanism to generate and propagate electrical signals across a neural structure independently from neurons.


Subject(s)
Motor Neurons , Spinal Cord , Action Potentials/physiology , Animals , Calcium Channels , Gap Junctions , Mice , Motor Neurons/physiology , Spinal Cord/physiology
5.
Curr Biol ; 31(21): 4762-4772.e5, 2021 11 08.
Article in English | MEDLINE | ID: mdl-34529937

ABSTRACT

Survival of animals is dependent on the correct selection of an appropriate behavioral response to competing external stimuli. Theoretical models have been proposed and underlying mechanisms are emerging to explain how one circuit is selected among competing neural circuits. The evolutionarily conserved forebrain to midbrain habenulo-interpeduncular nucleus (Hb-IPN) pathway consists of cholinergic and non-cholinergic neurons, which mediate different aversive behaviors. Simultaneous calcium imaging of neuronal cell bodies and of the population dynamics of their axon terminals reveals that signals in the cell bodies are not reflective of terminal activity. We find that axon terminals of cholinergic and non-cholinergic habenular neurons exhibit stereotypic patterns of spontaneous activity that are negatively correlated and localize to discrete subregions of the target IPN. Patch-clamp recordings show that calcium bursts in cholinergic terminals at the ventral IPN trigger excitatory currents in IPN neurons, which precede inhibition of non-cholinergic terminals at the adjacent dorsal IPN. Inhibition is mediated through presynaptic GABAB receptors activated in non-cholinergic habenular neurons upon GABA release from the target IPN. Together, the results reveal a hardwired mode of competition at the terminals of two excitatory neuronal populations, providing a physiological framework to explore the relationship between different aversive responses.


Subject(s)
Habenula , Presynaptic Terminals , Animals , Calcium/metabolism , Cholinergic Agents/metabolism , Habenula/physiology , Presynaptic Terminals/metabolism , gamma-Aminobutyric Acid/metabolism
6.
J Biol Phys ; 46(4): 371-394, 2020 12.
Article in English | MEDLINE | ID: mdl-33237338

ABSTRACT

Reliability engineering concerned with failure of technical inanimate systems usually uses the vocabulary and notions of human mortality, e.g., infant mortality vs. senescence mortality. Yet, few data are available to support such a parallel description. Here, we focus on early-stage (infant) mortality for two inanimate systems, incandescent light bulbs and soap films, and show the parallel description is clearly valid. Theoretical considerations of the thermo-electrical properties of electrical conductors allow us to link bulb failure to inherent mechanical defects. We then demonstrate the converse, that is, knowing the failure rate for an ensemble of light bulbs, it is possible to deduce the distribution of defects in wire thickness in the ensemble. Using measurements of lifetimes for soap films, we show how this methodology links failure rate to geometry of the system; in the case presented, this is the length of the tube containing the films. In a similar manner, for a third example, the time-dependent death rate due to congenital aortic valve stenosis is related to the distribution of degrees of severity of this condition, as a function of time. The results not only validate clearly the parallel description noted above, but also point firmly to application of the methodology to humans, with the consequent ability to gain more insight into the role of abnormalities in infant mortality.


Subject(s)
Infant Mortality , Models, Theoretical , Humans , Infant , Mechanical Phenomena , Temperature
7.
Sci Rep ; 10(1): 15338, 2020 09 18.
Article in English | MEDLINE | ID: mdl-32948826

ABSTRACT

While the role of cholinergic neurotransmission from motoneurons is well established during neuromuscular development, whether it regulates central nervous system development in the spinal cord is unclear. Zebrafish presents a powerful model to investigate how the cholinergic system is set up and evolves during neural circuit formation. In this study, we carried out a detailed spatiotemporal analysis of the cholinergic system in embryonic and larval zebrafish. In 1-day-old embryos, we show that spinal motoneurons express presynaptic cholinergic genes including choline acetyltransferase (chata), vesicular acetylcholine transporters (vachta, vachtb), high-affinity choline transporter (hacta) and acetylcholinesterase (ache), while nicotinic acetylcholine receptor (nAChR) subunits are mainly expressed in interneurons. However, in 3-day-old embryos, we found an unexpected decrease in presynaptic cholinergic transcript expression in a rostral to caudal gradient in the spinal cord, which continued during development. On the contrary, nAChR subunits remained highly expressed throughout the spinal cord. We found that protein and enzymatic activities of presynaptic cholinergic genes were also reduced in the rostral spinal cord. Our work demonstrating that cholinergic genes are initially expressed in the embryonic spinal cord, which is dynamically downregulated during development suggests that cholinergic signaling may play a pivotal role during the formation of intra-spinal locomotor circuit.


Subject(s)
Central Nervous System/embryology , Gene Expression Regulation, Developmental , Spinal Cord/embryology , Zebrafish Proteins/metabolism , Zebrafish/embryology , Animals , Animals, Genetically Modified , Central Nervous System/metabolism , Choline O-Acetyltransferase/genetics , Choline O-Acetyltransferase/metabolism , Embryo, Nonmammalian , Larva/metabolism , Motor Neurons/metabolism , Neurons/physiology , Neurotransmitter Agents/metabolism , Spinal Cord/metabolism , Vesicular Acetylcholine Transport Proteins/genetics , Vesicular Acetylcholine Transport Proteins/metabolism , Zebrafish/genetics , Zebrafish Proteins/genetics
8.
Cell Rep ; 29(10): 3173-3186.e7, 2019 12 03.
Article in English | MEDLINE | ID: mdl-31801081

ABSTRACT

Sox17, a SoxF family member transiently upregulated during postnatal oligodendrocyte (OL) development, promotes OL cell differentiation, but its function in white matter development and pathology in vivo is unknown. Our analysis of oligodendroglial- and OL-progenitor-cell-targeted ablation in vivo using a floxed Sox17 mouse establishes a dependence of postnatal oligodendrogenesis on Sox17 and reveals Notch signaling as a mediator of Sox17 function. Following Sox17 ablation, reduced numbers of Olig2-expressing cells and mature OLs led to developmental hypomyelination and motor dysfunction. After demyelination, Sox17 deficiency inhibited OL regeneration. OL decline was unexpectedly preceded by transiently increased differentiation and a reduction of OL progenitor cells. Evidence of a dual role for Sox17 in progenitor cell expansion by Notch and differentiation involving TCF7L2 expression were found. A program of progenitor expansion and differentiation promoted by Sox17 through Notch thus contributes to OL production and determines the outcome of white matter repair.


Subject(s)
Cell Differentiation/genetics , Cell Proliferation/genetics , HMGB Proteins/genetics , Oligodendrocyte Precursor Cells/physiology , SOXF Transcription Factors/genetics , Animals , Cell Cycle/genetics , Cells, Cultured , Demyelinating Diseases/metabolism , Female , Gene Expression Regulation, Developmental/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Oligodendrocyte Transcription Factor 2/genetics , Rats , Rats, Sprague-Dawley , Signal Transduction/genetics
9.
Curr Biol ; 27(14): 2154-2162.e3, 2017 Jul 24.
Article in English | MEDLINE | ID: mdl-28712566

ABSTRACT

Fear responses are defensive states that ensure survival of an organism in the presence of a threat. Perception of an aversive cue causes changes in behavior and physiology, such as freezing and elevated cortisol, followed by a return to the baseline state when the threat is evaded [1]. Neural systems that elicit fear behaviors include the amygdala, hippocampus, and medial prefrontal cortex. However, aside from a few examples, little is known about brain regions that promote recovery from an aversive event [2]. Previous studies had implicated the dorsal habenular nuclei in regulating fear responses and boldness in zebrafish [3-7]. We now show, through perturbation of its inherent left-right (L-R) asymmetry at larval stages, that the dorsal habenulo-interpeduncular (dHb-IPN) pathway expedites the return of locomotor activity following an unexpected negative stimulus, electric shock. Severing habenular efferents to the IPN, or only those from the left dHb, prolongs the freezing behavior that follows shock. Individuals with a symmetric, right-isomerized dHb also exhibit increased freezing. In contrast, larvae that have a symmetric, left-isomerized dHb, or in which just the left dHb-IPN projection is optogenetically activated, rapidly resume swimming post shock. In vivo calcium imaging reveals a neuronal subset, predominantly in the left dHb, whose activation is correlated with resumption of swimming. The results demonstrate functional specialization of the left dHb-IPN pathway in attenuating the response to fear.


Subject(s)
Fear/physiology , Habenula/physiology , Zebrafish/physiology , Animals , Animals, Genetically Modified/physiology
10.
Neural Dev ; 11: 1, 2016 Jan 18.
Article in English | MEDLINE | ID: mdl-26782621

ABSTRACT

BACKGROUND: Shaping of the neural tube, the precursor of the brain and spinal cord, involves narrowing and elongation of the neural tissue, concomitantly with other morphogenetic changes that contribue to this process. In zebrafish, medial displacement of neural cells (neural convergence or NC), which drives the infolding and narrowing of the neural ectoderm, is mediated by polarized migration and cell elongation towards the dorsal midline. Failure to undergo proper NC results in severe neural tube defects, yet the molecular underpinnings of this process remain poorly understood. RESULTS: We investigated here the role of the microtubule (MT) cytoskeleton in mediating NC in zebrafish embryos using the MT destabilizing and hyperstabilizing drugs nocodazole and paclitaxel respectively. We found that MTs undergo major changes in organization and stability during neurulation and are required for the timely completion of NC by promoting cell elongation and polarity. We next examined the role of Microtubule-associated protein 1B (Map1b), previously shown to promote MT dynamicity in axons. map1b is expressed earlier than previously reported, in the developing neural tube and underlying mesoderm. Loss of Map1b function using morpholinos (MOs) or δMap1b (encoding a truncated Map1b protein product) resulted in delayed NC and duplication of the neural tube, a defect associated with impaired NC. We observed a loss of stable MTs in these embryos that is likely to contribute to the NC defect. Lastly, we found that Map1b mediates cell elongation in a cell autonomous manner and polarized protrusive activity, two cell behaviors that underlie NC and are MT-dependent. CONCLUSIONS: Together, these data highlight the importance of MTs in the early morphogenetic movements that shape the neural tube and reveal a novel role for the MT regulator Map1b in mediating cell elongation and polarized cell movement in neural progenitor cells.


Subject(s)
Microtubule-Associated Proteins/metabolism , Neural Tube/embryology , Neurulation , Zebrafish Proteins/metabolism , Animals , Cell Movement/drug effects , Cell Polarity/drug effects , Neural Tube/drug effects , Neural Tube/metabolism , Neurulation/drug effects , Nocodazole/administration & dosage , Paclitaxel/administration & dosage , Tubulin Modulators/administration & dosage , Zebrafish
11.
Genesis ; 52(6): 636-55, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24753112

ABSTRACT

The role of the habenular nuclei in modulating fear and reward pathways has sparked a renewed interest in this conserved forebrain region. The bilaterally paired habenular nuclei, each consisting of a medial/dorsal and lateral/ventral nucleus, can be further divided into discrete subdomains whose neuronal populations, precise connectivity, and specific functions are not well understood. An added complexity is that the left and right habenulae show pronounced morphological differences in many non-mammalian species. Notably, the dorsal habenulae of larval zebrafish provide a vertebrate genetic model to probe the development and functional significance of brain asymmetry. Previous reports have described a number of genes that are expressed in the zebrafish habenulae, either in bilaterally symmetric patterns or more extensively on one side of the brain than the other. The goal of our study was to generate a comprehensive map of the zebrafish dorsal habenular nuclei, by delineating the relationship between gene expression domains, comparing the extent of left-right asymmetry at larval and adult stages, and identifying potentially functional subnuclear regions as defined by neurotransmitter phenotype. Although many aspects of habenular organization appear conserved with rodents, the zebrafish habenulae also possess unique properties that may underlie lateralization of their functions.


Subject(s)
Habenula/embryology , Neurotransmitter Agents/metabolism , Zebrafish/embryology , Zebrafish/metabolism , Animals , Animals, Genetically Modified , Gene Expression , Gene Expression Profiling , Gene Expression Regulation, Developmental , Genes, Reporter , Habenula/metabolism , Immunohistochemistry , Neurons/metabolism , Neurotransmitter Agents/genetics , Organ Specificity/genetics , Phenotype , Zebrafish/genetics
12.
Proc Natl Acad Sci U S A ; 110(52): 21171-6, 2013 Dec 24.
Article in English | MEDLINE | ID: mdl-24327734

ABSTRACT

The habenulo-interpeduncular pathway, a highly conserved cholinergic system, has emerged as a valuable model to study left-right asymmetry in the brain. In larval zebrafish, the bilaterally paired dorsal habenular nuclei (dHb) exhibit prominent left-right differences in their organization, gene expression, and connectivity, but their cholinergic nature was unclear. Through the discovery of a duplicated cholinergic gene locus, we now show that choline acetyltransferase and vesicular acetylcholine transporter homologs are preferentially expressed in the right dHb of larval zebrafish. Genes encoding the nicotinic acetylcholine receptor subunits α2 and ß4 are transcribed in the target interpeduncular nucleus (IPN), suggesting that the asymmetrical cholinergic pathway is functional. To confirm this, we activated channelrhodopsin-2 specifically in the larval dHb and performed whole-cell patch-clamp recording of IPN neurons. The response to optogenetic or electrical stimulation of the right dHb consisted of an initial fast glutamatergic excitatory postsynaptic current followed by a slow-rising cholinergic current. In adult zebrafish, the dHb are divided into discrete cholinergic and peptidergic subnuclei that differ in size between the left and right sides of the brain. After exposing adults to nicotine, fos expression was activated in subregions of the IPN enriched for specific nicotinic acetylcholine receptor subunits. Our studies of the newly identified cholinergic gene locus resolve the neurotransmitter identity of the zebrafish habenular nuclei and reveal functional asymmetry in a major cholinergic neuromodulatory pathway of the vertebrate brain.


Subject(s)
Functional Laterality/physiology , Gene Expression Regulation, Developmental/physiology , Habenula/physiology , Models, Animal , Tegmentum Mesencephali/physiology , Acetylcholine/metabolism , Animals , Base Sequence , Choline O-Acetyltransferase/genetics , Choline O-Acetyltransferase/metabolism , DNA Primers/genetics , Electric Stimulation , Habenula/metabolism , In Situ Hybridization , Larva/physiology , Molecular Sequence Data , Neural Pathways/physiology , Optogenetics , Patch-Clamp Techniques , Receptors, Nicotinic/metabolism , Sequence Analysis, RNA , Tegmentum Mesencephali/metabolism , Vesicular Acetylcholine Transport Proteins/metabolism , Zebrafish
13.
J Neurosci ; 31(39): 13921-35, 2011 Sep 28.
Article in English | MEDLINE | ID: mdl-21957254

ABSTRACT

The SRY-box (Sox) transcription factors regulate oligodendrocyte differentiation, but their signaling targets are largely unknown. We have identified a major signal transduction pathway regulated by Sox containing gene 17 (Sox17) in the oligodendrocyte lineage. Microarray analysis in oligodendrocyte progenitor cells (OPCs) after Sox17 attenuation revealed upregulated genes associated with cell cycle control and activation of the Wingless and integration site (Wnt)/ß-catenin pathway. Sox17 knockdown also increases the levels of cyclin D1, Axin2, and activated ß-catenin. In OPCs, the expression pattern of Sox17, cyclin D1, and secreted Frizzled-related protein-1 in the presence of platelet-derived growth factor (PDGF) was coordinately accelerated by addition of thyroid hormone, indicating differentiation-induced regulation of Sox17 targets. In developing white matter, decreased total ß-catenin, activated ß-catenin, and cyclin D1 levels coincided with the peak of Sox17 expression, and immunoprecipitates showed a developmentally regulated interaction among Sox17, T-cell transcription factor 4, and ß-catenin proteins. In OPCs, PDGF stimulated phosphorylation of glycogen synthase 3ß and the Wnt coreceptor LRP6, and enhanced ß-catenin-dependent gene expression. Sox17 overexpression inhibited PDGF-induced TOPFLASH and cyclin D1 promoter activity, and decreased endogenous cyclin D1, activated ß-catenin, as well as total ß-catenin levels. Recombinant Sox17 prevented Wnt3a from repressing myelin protein expression, and inhibition of Sox17-mediated proteasomal degradation of ß-catenin blocked myelin protein induction. These results indicate that Sox17 suppresses cyclin D1 expression and cell proliferation by directly antagonizing ß-catenin, whose activity in OPCs is stimulated not only by Wnt3a, but also by PDGF. Our identification of downstream targets of Sox17 thus defines signaling pathways and molecular mechanisms in OPCs that are regulated by Sox17 during cell cycle exit and the onset of differentiation in oligodendrocyte development.


Subject(s)
HMGB Proteins/physiology , Oligodendroglia/physiology , SOXF Transcription Factors/physiology , Signal Transduction/physiology , Stem Cells/physiology , Wnt Proteins/physiology , beta Catenin/physiology , Animals , Cells, Cultured , Gene Knock-In Techniques , HMGB Proteins/antagonists & inhibitors , HMGB Proteins/genetics , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic , Myelin Proteins/antagonists & inhibitors , Myelin Proteins/biosynthesis , NIH 3T3 Cells , Rats , Rats, Sprague-Dawley , Recombinant Proteins/antagonists & inhibitors , Recombinant Proteins/genetics , Recombinant Proteins/pharmacology , SOXF Transcription Factors/antagonists & inhibitors , SOXF Transcription Factors/genetics , beta Catenin/antagonists & inhibitors
14.
J Vis Exp ; (41)2010 Jul 25.
Article in English | MEDLINE | ID: mdl-20689510

ABSTRACT

Key to understanding the morphogenetic processes that shape the early vertebrate embryo is the ability to image cells at high resolution. In zebrafish embryos, injection of plasmid DNA results in mosaic expression, allowing for the visualization of single cells or small clusters of cells (1) . We describe how injection of plasmid DNA encoding membrane-targeted Green Fluorescent Protein (mGFP) under the control of a ubiquitous promoter can be used for imaging cells undergoing neurulation. Central to this protocol is the methodology for imaging labeled cells at high resolution in sections and also in real time. This protocol entails the injection of mGFP DNA into young zebrafish embryos. Embryos are then processed for vibratome sectioning, antibody labeling and imaging with a confocal microscope. Alternatively, live embryos expressing mGFP can be imaged using time-lapse confocal microscopy. We have previously used this straightforward approach to analyze the cellular behaviors that drive neural tube formation in the hindbrain region of zebrafish embryos (2). The fixed preparations allowed for unprecedented visualization of cell shapes and organization in the neural tube while live imaging complemented this approach enabling a better understanding of the cellular dynamics that take place during neurulation.


Subject(s)
Green Fluorescent Proteins/analysis , Microscopy, Confocal/methods , Rhombencephalon/embryology , Zebrafish/embryology , Animals , DNA/administration & dosage , DNA/genetics , Embryo, Nonmammalian/cytology , Green Fluorescent Proteins/biosynthesis , Green Fluorescent Proteins/genetics , Microinjections/methods , Plasmids/genetics , Rhombencephalon/cytology
15.
Dev Biol ; 341(2): 335-45, 2010 May 15.
Article in English | MEDLINE | ID: mdl-20138861

ABSTRACT

Microtubules are essential regulators of cell polarity, architecture and motility. The organization of the microtubule network is context-specific. In non-polarized cells, microtubules are anchored to the centrosome and form radial arrays. In most epithelial cells, microtubules are noncentrosomal, align along the apico-basal axis and the centrosome templates a cilium. It follows that cells undergoing mesenchyme-to-epithelium transitions must reorganize their microtubule network extensively, yet little is understood about how this process is orchestrated. In particular, the pathways regulating the apical positioning of the centrosome are unknown, a central question given the role of cilia in fluid propulsion, sensation and signaling. In zebrafish, neural progenitors undergo progressive epithelialization during neurulation, and thus provide a convenient in vivo cellular context in which to address this question. We demonstrate here that the microtubule cytoskeleton gradually transitions from a radial to linear organization during neurulation and that microtubules function in conjunction with the polarity protein Pard3 to mediate centrosome positioning. Pard3 depletion results in hydrocephalus, a defect often associated with abnormal cerebrospinal fluid flow that has been linked to cilia defects. These findings thus bring to focus cellular events occurring during neurulation and reveal novel molecular mechanisms implicated in centrosome positioning.


Subject(s)
Carrier Proteins/metabolism , Cell Polarity , Centrosome/metabolism , Microtubules/metabolism , Neurulation , Zebrafish Proteins/metabolism , Zebrafish/embryology , Animals , Axoneme/metabolism , Carrier Proteins/analysis , Cilia/metabolism , Zebrafish Proteins/analysis
16.
Mol Reprod Dev ; 76(10): 954-65, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19653285

ABSTRACT

The central nervous system of vertebrate embryos originates from the neural tube (NT), a simple epithelium surrounding a central lumen. The mechanisms underlying the shaping of the NT, a process otherwise known as neurulation, have been the focus of numerous studies, using a variety of model systems. Yet, it remains unclear to what extent neurulation is conserved across vertebrates. This review provides a comparison between modes of neurulation, with a focus on cellular mechanisms. An emerging concept is that cell behaviors reveal similarities between modes of neurulation that cannot be predicted from morphological comparisons.


Subject(s)
Neurulation/physiology , Animals , Signal Transduction
17.
BMC Dev Biol ; 7: 130, 2007 Nov 28.
Article in English | MEDLINE | ID: mdl-18045497

ABSTRACT

BACKGROUND: The anterior-posterior axis of the vertebrate embryo undergoes a dramatic elongation during early development. Convergence and extension of the mesoderm, occurring during gastrulation, initiates the narrowing and lengthening of the embryo. However the lengthening of the axis continues during post-gastrula stages in the tailbud region, and is thought to involve convergent extension movements as well as other cell behaviors specific to posterior regions. RESULTS: We demonstrate here, using a semi-dominant N-cadherin allele, that members of the classical cadherin subfamily of cell-cell adhesion molecules are required for tailbud elongation in the zebrafish. In vivo imaging of cell behaviors suggests that the extension of posterior axial mesodermal cells is impaired in embryos that carry the semi-dominant N-cadherin allele. This defect most likely results from a general loss of cell-cell adhesion in the tailbud region. Consistent with these observations, N-cadherin is expressed throughout the tailbud during post-gastrulation stages. In addition, we show that N-cadherin interacts synergistically with vang-like 2, a member of the non-canonical Wnt signaling/planar cell polarity pathway, to mediate tail morphogenesis. CONCLUSION: We provide the first evidence here that N-cadherin and other members of the classical cadherin subfamily function in parallel with the planar cell polarity pathway to shape the posterior axis during post-gastrulation stages. These findings further highlight the central role that adhesion molecules play in the cellular rearrangements that drive morphogenesis in vertebrates and identify classical cadherins as major contributors to tail development.


Subject(s)
Body Patterning/genetics , Cadherins/genetics , Embryo, Nonmammalian/embryology , Zebrafish Proteins/genetics , Zebrafish/embryology , Animals , Apoptosis , Cell Adhesion , Cell Polarity , Gene Expression Regulation, Developmental , In Situ Hybridization , Oligonucleotides, Antisense , Protein Biosynthesis , Tail/embryology , Zebrafish/genetics
18.
Development ; 133(19): 3895-905, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16943271

ABSTRACT

Through the direct analysis of cell behaviors, we address the mechanisms underlying anterior neural tube morphogenesis in the zebrafish and the role of the cell adhesion molecule N-cadherin (N-cad) in this process. We demonstrate that although the mode of neurulation differs at the morphological level between amphibians and teleosts, the underlying cellular mechanisms are conserved. Contrary to previous reports, the zebrafish neural plate is a multi-layered structure, composed of deep and superficial cells that converge medially while undergoing radial intercalation, to form a single cell-layered neural tube. Time-lapse recording of individual cell behaviors reveals that cells are polarized along the mediolateral axis and exhibit protrusive activity. In N-cad mutants, both convergence and intercalation are blocked. Moreover, although N-cad-depleted cells are not defective in their ability to form protrusions, they are unable to maintain them stably. Taken together, these studies uncover key cellular mechanisms underlying neural tube morphogenesis in teleosts, and reveal a role for cadherins in promoting the polarized cell behaviors that underlie cellular rearrangements and shape the vertebrate embryo.


Subject(s)
Cadherins/physiology , Nervous System/embryology , Zebrafish/embryology , Animals , Cadherins/genetics , Cell Polarity/genetics , Epithelium/metabolism , Epithelium/ultrastructure , Models, Biological , Morphogenesis/genetics , Mutation , Nervous System/cytology , Neural Tube Defects/genetics , Neuroepithelial Cells/cytology , Neuroepithelial Cells/physiology , Zebrafish/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...