Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Glia ; 67(7): 1401-1411, 2019 07.
Article in English | MEDLINE | ID: mdl-30924555

ABSTRACT

Cell shape is critical for the proper function of every cell in every tissue in the body. This is especially true for the highly morphologically diverse neural and glia cells of the central nervous system. The molecular processes by which these, or indeed any, cells gain their particular cell-specific morphology remain largely unexplored. To identify the genes involved in the morphogenesis of the principal glial cell type in the vertebrate retina, the Müller glia (MG), we used genomic and CRISPR based strategies in zebrafish (Danio rerio). We identified 41 genes involved in various aspects of MG cell morphogenesis and revealed a striking concordance between the sequential steps of anatomical feature addition and the expression of cohorts of functionally related genes that regulate these steps. We noted that the many of the genes preferentially expressed in zebrafish MG showed conservation in glia across species suggesting evolutionarily conserved glial developmental pathways.


Subject(s)
Ependymoglial Cells/physiology , Gene Expression Profiling/methods , Morphogenesis/physiology , Neurogenesis/physiology , Neuroglia/physiology , Transcriptome/physiology , Animals , Animals, Genetically Modified , Cell Differentiation/physiology , Clustered Regularly Interspaced Short Palindromic Repeats/physiology , Zebrafish
2.
Development ; 143(7): 1099-107, 2016 Apr 01.
Article in English | MEDLINE | ID: mdl-26893352

ABSTRACT

Clonal analysis is helping us understand the dynamics of cell replacement in homeostatic adult tissues (Simons and Clevers, 2011). Such an analysis, however, has not yet been achieved for continuously growing adult tissues, but is essential if we wish to understand the architecture of adult organs. The retinas of lower vertebrates grow throughout life from retinal stem cells (RSCs) and retinal progenitor cells (RPCs) at the rim of the retina, called the ciliary marginal zone (CMZ). Here, we show that RSCs reside in a niche at the extreme periphery of the CMZ and divide asymmetrically along a radial (peripheral to central) axis, leaving one daughter in the peripheral RSC niche and the other more central where it becomes an RPC. We also show that RPCs of the CMZ have clonal sizes and compositions that are statistically similar to progenitor cells of the embryonic retina and fit the same stochastic model of proliferation. These results link embryonic and postembryonic cell behaviour, and help to explain the constancy of tissue architecture that has been generated over a lifetime.


Subject(s)
Cell Differentiation/physiology , Retina/cytology , Retina/growth & development , Stem Cells/cytology , Zebrafish/growth & development , Animals , Animals, Genetically Modified , Cell Division , Cell Proliferation , Gene Expression Regulation, Developmental
3.
Development ; 142(15): 2665-77, 2015 Aug 01.
Article in English | MEDLINE | ID: mdl-26116662

ABSTRACT

The mature vertebrate retina is a highly ordered neuronal network of cell bodies and synaptic neuropils arranged in distinct layers. Little, however, is known about the emergence of this spatial arrangement. Here, we investigate how the three main types of retinal inhibitory neuron (RIN)--horizontal cells (HCs), inner nuclear layer amacrine cells (iACs) and displaced amacrine cells (dACs)--reach their specific laminar positions during development. Using in vivo time-lapse imaging of zebrafish retinas, we show that RINs undergo distinct phases of migration. The first phase, common to all RINs, is bipolar migration directed towards the apicobasal centre of the retina. All RINs then transition to a less directionally persistent multipolar phase of migration. Finally, HCs, iACs and dACs each undergo cell type-specific migration. In contrast to current hypotheses, we find that most dACs send processes into the forming inner plexiform layer (IPL) before migrating through it and inverting their polarity. By imaging and quantifying the dynamics of HCs, iACs and dACs from birth to final position, this study thus provides evidence for distinct and new migration patterns during retinal lamination and insights into the initiation of IPL formation.


Subject(s)
Cell Movement/physiology , Neurons/physiology , Retina/embryology , Zebrafish/embryology , Animals , Animals, Genetically Modified , Image Processing, Computer-Assisted , Kinetics , Microscopy, Fluorescence , Neurons/cytology , Time-Lapse Imaging
4.
Development ; 141(9): 1971-80, 2014 May.
Article in English | MEDLINE | ID: mdl-24718991

ABSTRACT

The ability to image cells live and in situ as they proliferate and differentiate has proved to be an invaluable asset to biologists investigating developmental processes. Here, we describe a Spectrum of Fates approach that allows the identification of all the major neuronal subtypes in the zebrafish retina simultaneously. Spectrum of Fates is based on the combinatorial expression of differently coloured fluorescent proteins driven by the promoters of transcription factors that are expressed in overlapping subsets of retinal neurons. Here, we show how a Spectrum of Fates approach can be used to assess various aspects of neural development, such as developmental waves of differentiation, neuropil development, lineage tracing and hierarchies of fates in the developing zebrafish retina.


Subject(s)
Genetic Techniques , Luminescent Proteins/metabolism , Retina/embryology , Zebrafish/embryology , Zebrafish/genetics , Animals , Animals, Genetically Modified , Cell Differentiation , Cell Lineage , Cell Proliferation , Cells, Cultured , Clone Cells , Fluorescence , Retina/cytology
5.
Open Biol ; 3(10): 130065, 2013 Oct 23.
Article in English | MEDLINE | ID: mdl-24153002

ABSTRACT

Autosomal recessive primary microcephaly (MCPH) is a congenital disorder characterized by significantly reduced brain size and mental retardation. Nine genes are currently known to be associated with the condition, all of which encode centrosomal or spindle pole proteins. MCPH is associated with a reduction in proliferation of neural progenitors during fetal development. The cellular mechanisms underlying the proliferation defect, however, are not fully understood. The zebrafish retinal neuroepithelium provides an ideal system to investigate this question. Mutant or morpholino-mediated knockdown of three known MCPH genes (stil, aspm and wdr62) and a fourth centrosomal gene, odf2, which is linked to several MCPH proteins, results in a marked reduction in head and eye size. Imaging studies reveal a dramatic rise in the fraction of proliferating cells in mitosis in all cases, and time-lapse microscopy points to a failure of progression through prometaphase. There was also increased apoptosis in all the MCPH models but this appears to be secondary to the mitotic defect as we frequently saw mitotically arrested cells disappear, and knocking down p53 apoptosis did not rescue the mitotic phenotype, either in whole retinas or clones.


Subject(s)
Metaphase , Retina/embryology , Retina/metabolism , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism , Zebrafish/metabolism , Animals , Apoptosis/genetics , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Line, Transformed , Disease Models, Animal , Embryonic Development , Evolution, Molecular , Eye Abnormalities/embryology , Eye Abnormalities/genetics , Eye Abnormalities/metabolism , Gene Knockdown Techniques , Genes, p53 , Head/abnormalities , Head/embryology , Humans , Microcephaly/genetics , Microcephaly/metabolism , Microcephaly/physiopathology , Mitosis/genetics , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Retina/cytology , Retinal Neurons/cytology , Retinal Neurons/metabolism , Stem Cells/cytology , Time-Lapse Imaging , Zebrafish/embryology , Zebrafish/genetics
6.
Cell Rep ; 3(2): 282-90, 2013 Feb 21.
Article in English | MEDLINE | ID: mdl-23416047

ABSTRACT

How synaptic neuropil is formed within the CNS is poorly understood. The retinal inner plexiform layer (IPL) is positioned between the cell bodies of amacrine cells (ACs) and retinal ganglion cells (RGCs). It consists of bipolar cell (BC) axon terminals that synapse on the dendrites of ACs and RGCs intermingled with projections from Müller glia (MG). We examined whether any of these cellular processes are specifically required for the formation of the IPL. Using genetic and pharmacological strategies, we eliminated RGCs, ACs, and MG individually or in combination. Even in the absence of all of these partner cells, an IPL-like neuropil consisting of only BC axon terminals still forms, complete with presynaptic specializations and sublaminar organization. Previous studies have shown that an IPL can form in the complete absence of BCs; therefore, we conclude that neither presynaptic nor postsynaptic processes are individually essential for the formation of this synaptic neuropil.


Subject(s)
Neuropil/cytology , Retina/ultrastructure , Amacrine Cells/cytology , Amacrine Cells/pathology , Amacrine Cells/ultrastructure , Animals , Animals, Genetically Modified , Embryo, Nonmammalian/metabolism , Microscopy, Fluorescence , Microscopy, Video , Neurons/pathology , Neuropil/pathology , Presynaptic Terminals/pathology , Retina/pathology , Retinal Bipolar Cells/cytology , Retinal Bipolar Cells/pathology , Retinal Bipolar Cells/ultrastructure , Retinal Ganglion Cells/cytology , Retinal Ganglion Cells/pathology , Retinal Ganglion Cells/ultrastructure , Synapses/pathology , Synapses/ultrastructure , Zebrafish/growth & development
7.
Neuron ; 75(5): 786-98, 2012 Sep 06.
Article in English | MEDLINE | ID: mdl-22958820

ABSTRACT

A fundamental question in developmental neuroscience is how a collection of progenitor cells proliferates and differentiates to create a brain of the appropriate size and cellular composition. To address this issue, we devised lineage-tracing assays in developing zebrafish embryos to reconstruct entire retinal lineage progressions in vivo and thereby provide a complete quantitative map of the generation of a vertebrate CNS tissue from individual progenitors. These lineage data are consistent with a simple model in which the retina is derived from a set of equipotent retinal progenitor cells (RPCs) that are subject to stochastic factors controlling lineage progression. Clone formation in mutant embryos reveals that the transcription factor Ath5 acts as a molecular link between fate choice and mode of cell division, giving insight into the elusive molecular mechanisms of histogenesis, the conserved temporal order by which neurons of different types exit the cell cycle.


Subject(s)
Organogenesis/physiology , Retina/embryology , Retinal Neurons/physiology , Stem Cells/physiology , Animals , Animals, Genetically Modified , Cell Lineage/genetics , Clone Cells , Embryo, Nonmammalian/cytology , Embryo, Nonmammalian/embryology , Retina/cytology , Retinal Neurons/cytology , Stem Cells/cytology , Zebrafish
8.
J Neurosci ; 31(7): 2549-62, 2011 Feb 16.
Article in English | MEDLINE | ID: mdl-21325522

ABSTRACT

Multipotent progenitors in the vertebrate retina often generate clonally related mixtures of excitatory and inhibitory neurons. The postmitotically expressed transcription factor, Ptf1a, is essential for all inhibitory fates in the zebrafish retina, including three types of horizontal and 28 types of amacrine cell. Here, we show that specific types of inhibitory neurons arise from the cell-autonomous influence of Ptf1a in the daughters of fate-restricted progenitors, such as Ath5 or Vsx1/2-expressing progenitors, and that in the absence of Ptf1a, cells that would have become these specific inhibitory subtypes revert to the histogenetically appropriate excitatory subtypes of the same lineage. Altered proportions of amacrine subtypes respecified by the misexpression of Ptf1a in the Ath5 lineage suggest that Ath5-expressing progenitors are biased, favoring the generation of some subtypes more than others. Yet the full array of inhibitory cell subtypes in Ath5 mutants implies the existence of Ath5-independent factors involved in inhibitory cell specification. We also show that an extrinsic negative feedback on the expression of Ptf1a provides a control mechanism by which the number of any and all types of inhibitory cells in the retina can be regulated in this lineage-dependent way.


Subject(s)
Cell Differentiation/genetics , Cell Lineage/genetics , Gene Expression Regulation, Developmental/physiology , Neural Inhibition/physiology , Retina/cytology , Amacrine Cells/classification , Amacrine Cells/physiology , Animals , Animals, Genetically Modified , Blastomeres/transplantation , Bromodeoxyuridine/metabolism , Cell Differentiation/drug effects , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Embryo, Nonmammalian , Embryonic Stem Cells/drug effects , Embryonic Stem Cells/physiology , Gene Expression Regulation, Developmental/genetics , Glycine/metabolism , Green Fluorescent Proteins/genetics , Luminescent Proteins/genetics , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neural Inhibition/genetics , Oligonucleotides, Antisense/pharmacology , Retina/metabolism , Stem Cell Transplantation/methods , Transcription Factors/genetics , Transcription Factors/metabolism , Zebrafish , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism , gamma-Aminobutyric Acid/metabolism , Red Fluorescent Protein
9.
Neural Dev ; 5: 1, 2010 Jan 04.
Article in English | MEDLINE | ID: mdl-20047651

ABSTRACT

BACKGROUND: The neural crest is a unique population of cells that arise in the vertebrate ectoderm at the neural plate border after which they migrate extensively throughout the embryo, giving rise to a wide range of derivatives. A number of proteins involved in neural crest development have dynamic expression patterns, and it is becoming clear that ubiquitin-mediated protein degradation is partly responsible for this. RESULTS: Here we demonstrate a novel role for the F-box protein Cdc4/Fbxw7 in neural crest development. Two isoforms of Xenopus laevis Cdc4 were identified, and designated xCdc4alpha and xCdc4beta. These are highly conserved with vertebrate Cdc4 orthologs, and the Xenopus proteins are functionally equivalent in terms of their ability to degrade Cyclin E, an established vertebrate Cdc4 target. Blocking xCdc4 function specifically inhibited neural crest development at an early stage, prior to expression of c-Myc, Snail2 and Snail. CONCLUSIONS: We demonstrate that Cdc4, an ubiquitin E3 ligase subunit previously identified as targeting primarily cell cycle regulators for proteolysis, has additional roles in control of formation of the neural crest. Hence, we identify Cdc4 as a protein with separable but complementary functions in control of cell proliferation and differentiation.


Subject(s)
F-Box Proteins/metabolism , Neural Crest/embryology , Neural Crest/metabolism , Xenopus Proteins/metabolism , Xenopus laevis/embryology , Animals , Blotting, Western , Cell Differentiation/genetics , Cell Proliferation , Cyclin E/metabolism , DNA, Complementary , F-Box Proteins/genetics , F-Box-WD Repeat-Containing Protein 7 , Gene Expression Regulation, Developmental , In Situ Hybridization , Microinjections , Plasmids/genetics , Point Mutation , Polymerase Chain Reaction , Protein Isoforms/metabolism , RNA, Messenger , Sequence Deletion , Ubiquitin/genetics , Xenopus Proteins/genetics , Xenopus laevis/genetics , Xenopus laevis/metabolism
10.
PLoS One ; 2(10): e1004, 2007 Oct 10.
Article in English | MEDLINE | ID: mdl-17925852

ABSTRACT

BACKGROUND: Cell-cell communication is essential in tissue patterning. In early amphibian development, mesoderm is formed in the blastula-stage embryo through inductive interactions in which vegetal cells act on overlying equatorial cells. Members of the TGF-beta family such as activin B, Vg1, derrière and Xenopus nodal-related proteins (Xnrs) are candidate mesoderm inducing factors, with further activity to induce endoderm of the vegetal region. TGF-beta-like ligands, including BMP, are also responsible for patterning of germ layers. In addition, FGF signaling is essential for mesoderm formation whereas FGF signal inhibition has been implicated in endoderm induction. Clearly, several signaling pathways are coordinated to produce an appropriate developmental output; although intracellular crosstalk is known to integrate multiple pathways, relatively little is known about extracellular coordination. METHODOLOGY/PRINCIPAL FINDINGS: Here, we show that Xenopus Tsukushi (X-TSK), a member of the secreted small leucine rich repeat proteoglycan (SLRP) family, is expressed in ectoderm, endoderm, and the organizer during early development. We have previously reported that X-TSK binds to and inhibits BMP signaling in cooperation with chordin. We now demonstrate two novel interactions: X-TSK binds to and inhibits signaling by FGF8b, in addition to binding to and enhancement of Xnr2 signaling. This signal integration by X-TSK at the extracellular level has an important role in germ layer formation and patterning. Vegetally localized X-TSK potentiates endoderm formation through coordination of BMP, FGF and Xnr2 signaling. In contrast, X-TSK inhibition of FGF-MAPK signaling blocks ventrolateral mesoderm formation, while BMP inhibition enhances organizer formation. These actions of X-TSK are reliant upon its expression in endoderm and dorsal mesoderm, with relative exclusion from ventrolateral mesoderm, in a pattern shaped by FGF signals. CONCLUSIONS/SIGNIFICANCE: Based on our observations, we propose a novel mechanism by which X-TSK refines the field of positional information by integration of multiple pathways in the extracellular space.


Subject(s)
Bone Morphogenetic Proteins/metabolism , Fibroblast Growth Factors/metabolism , Gene Expression Regulation, Developmental , Germ Layers/metabolism , Signal Transduction , Xenopus Proteins/metabolism , Xenopus Proteins/physiology , Animals , Cell Communication , Endoderm/metabolism , Germ Cells , Ligands , Mesoderm , Models, Biological , Transforming Growth Factor beta/metabolism
11.
Biochem J ; 388(Pt 1): 185-94, 2005 May 15.
Article in English | MEDLINE | ID: mdl-15651977

ABSTRACT

The regulation of survival and cell death is a key determinant of cell fate. Recent evidence shows that survival and death machineries are regulated along the cell cycle. In the present paper, we show that BimEL [a BH3 (Bcl-2 homology 3)-only member of the Bcl-2 family of proteins; Bim is Bcl-2-interacting mediator of cell death; EL is the extra-long form] is phosphorylated in mitosis. This post-translational modification is dependent on MEK (mitogen-activated protein kinase/extracellular-signal-regulated kinase kinase) and growth factor signalling. Interestingly, FGF (fibroblast growth factor) signalling seems to play an essential role in this process, since, in the presence of serum, inhibition of FGF receptors abrogated phosphorylation of Bim in mitosis. Moreover, we have shown bFGF (basic FGF) to be sufficient to induce phosphorylation of Bim in serum-free conditions in any phase of the cell cycle, and also to significantly rescue cells from serum-deprivation-induced apoptosis. Our results show that, in mitosis, Bim is phosphorylated downstream of growth factor signalling in a MEK-dependent manner, with FGF signalling playing an important role. We suggest that phosphorylation of Bim is a decisive step for the survival of proliferating cells.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Apoptosis/physiology , Fibroblast Growth Factors/physiology , Membrane Proteins/metabolism , Mitosis/physiology , Proto-Oncogene Proteins/metabolism , Animals , Bcl-2-Like Protein 11 , CDC2 Protein Kinase/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , MAP Kinase Kinase Kinases/metabolism , Mice , NIH 3T3 Cells , Phosphorylation , Protein Processing, Post-Translational , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL