Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 44
Filter
Add more filters











Publication year range
1.
Dev Cell ; 58(3): 239-255.e10, 2023 02 06.
Article in English | MEDLINE | ID: mdl-36706756

ABSTRACT

The adult spinal cord stem cell potential resides within the ependymal cell population and declines with age. Ependymal cells are, however, heterogeneous, and the biological diversity this represents and how it changes with age remain unknown. Here, we present a single-cell transcriptomic census of spinal cord ependymal cells from adult and aged mice, identifying not only all known ependymal cell subtypes but also immature as well as mature cell states. By comparing transcriptomes of spinal cord and brain ependymal cells, which lack stem cell abilities, we identify immature cells as potential spinal cord stem cells. Following spinal cord injury, these cells re-enter the cell cycle, which is accompanied by a short-lived reversal of ependymal cell maturation. We further analyze ependymal cells in the human spinal cord and identify widespread cell maturation and altered cell identities. This in-depth characterization of spinal cord ependymal cells provides insight into their biology and informs strategies for spinal cord repair.


Subject(s)
Neuroglia , Spinal Cord Injuries , Adult , Animals , Humans , Mice , Cell Differentiation , Neuroglia/metabolism , Spinal Cord/metabolism , Spinal Cord Injuries/metabolism
2.
PLoS Biol ; 20(12): e3000221, 2022 12.
Article in English | MEDLINE | ID: mdl-36455041

ABSTRACT

Fibroblast growth factor (FGF) is a neural inducer in many vertebrate embryos, but how it regulates chromatin organization to coordinate the activation of neural genes is unclear. Moreover, for differentiation to progress, FGF signalling must decline. Why these signalling dynamics are required has not been determined. Here, we show that dephosphorylation of the FGF effector kinase ERK1/2 rapidly increases chromatin accessibility at neural genes in mouse embryos, and, using ATAC-seq in human embryonic stem cell derived spinal cord precursors, we demonstrate that this occurs genome-wide across neural genes. Importantly, ERK1/2 inhibition induces precocious neural gene transcription, and this involves dissociation of the polycomb repressive complex from key gene loci. This takes place independently of subsequent loss of the repressive histone mark H3K27me3 and transcriptional onset. Transient ERK1/2 inhibition is sufficient for the dissociation of the repressive complex, and this is not reversed on resumption of ERK1/2 signalling. Moreover, genomic footprinting of sites identified by ATAC-seq together with ChIP-seq for polycomb protein Ring1B revealed that ERK1/2 inhibition promotes the occupancy of neural transcription factors (TFs) at non-polycomb as well as polycomb associated sites. Together, these findings indicate that ERK1/2 signalling decline promotes global changes in chromatin accessibility and TF binding at neural genes by directing polycomb and other regulators and appears to serve as a gating mechanism that provides directionality to the process of differentiation.


Subject(s)
Chromatin , MAP Kinase Signaling System , Mice , Humans , Animals , Polycomb-Group Proteins/genetics , Polycomb-Group Proteins/metabolism , Cell Differentiation , Signal Transduction
3.
Elife ; 112022 02 21.
Article in English | MEDLINE | ID: mdl-35188104

ABSTRACT

Species-specific differentiation pace in vitro indicates that some aspects of neural differentiation are governed by cell intrinsic properties. Here we describe a novel in vitro human neural-rosette assay that recapitulates dorsal spinal cord differentiation but proceeds more rapidly than in the human embryo, suggesting that it lacks endogenous signalling dynamics. To test whether in vitro conditions represent an intrinsic differentiation pace, human iPSC-derived neural rosettes were challenged by grafting into the faster differentiating chicken embryonic neural tube iso-chronically, or hetero-chronically into older embryos. In both contexts in vitro differentiation pace was initially unchanged, while long-term analysis revealed iso-chronic slowed and hetero-chronic conditions promoted human neural differentiation. Moreover, hetero-chronic conditions did not alter the human neural differentiation programme, which progressed to neurogenesis, while the host embryo advanced into gliogenesis. This study demonstrates that intrinsic properties limit human differentiation pace, and that timely extrinsic signals are required for progression through an intrinsic human neural differentiation programme.


Subject(s)
Induced Pluripotent Stem Cells , Spinal Cord , Animals , Cell Differentiation , Chick Embryo , Humans , Neural Tube , Neurogenesis
4.
J Cell Sci ; 135(6)2022 03 15.
Article in English | MEDLINE | ID: mdl-35217862

ABSTRACT

Dynamic contacts between cells within the developing neuroepithelium are poorly understood but play important roles in cell and tissue morphology and cell signalling. Here, using live-cell imaging and electron microscopy we reveal multiple protrusive structures in neuroepithelial apical endfeet of the chick embryonic spinal cord, including sub-apical protrusions that extend laterally within the tissue, and observe similar structures in human neuroepithelium. We characterise the dynamics, shape and cytoskeleton of these lateral protrusions and distinguish them from cytonemes, filopodia and tunnelling nanotubes. We demonstrate that lateral protrusions form a latticework of membrane contacts between non-adjacent cells, depend on actin but not microtubule dynamics, and provide a lamellipodial-like platform for further extending fine actin-dependent filipodia. We find that lateral protrusions depend on the actin-binding protein WAVE1 (also known as WASF1): misexpression of mutant WAVE1 attenuated protrusion and generated a round-ended apical endfoot morphology. However, this did not alter apico-basal cell polarity or tissue integrity. During normal neuronal delamination, lateral protrusions were withdrawn, but precocious protrusion loss induced by mutant WAVE1 was insufficient to trigger neurogenesis. This study uncovers a new form of cell-cell contact within the developing neuroepithelium, regulation of which prefigures neuronal delamination. This article has an associated First Person interview with the first author of the paper.


Subject(s)
Actins , Neuroepithelial Cells , Actins/metabolism , Cytoskeleton/metabolism , Humans , Neuroepithelial Cells/metabolism , Neurogenesis , Pseudopodia/metabolism , Wiskott-Aldrich Syndrome Protein Family/metabolism
5.
PLoS Biol ; 18(3): e3000470, 2020 03.
Article in English | MEDLINE | ID: mdl-32150534

ABSTRACT

In the spinal cord, the central canal forms through a poorly understood process termed dorsal collapse that involves attrition and remodelling of pseudostratified ventricular layer (VL) cells. Here, we use mouse and chick models to show that dorsal ventricular layer (dVL) cells adjacent to dorsal midline Nestin(+) radial glia (dmNes+RG) down-regulate apical polarity proteins, including Crumbs2 (CRB2) and delaminate in a stepwise manner; live imaging shows that as one cell delaminates, the next cell ratchets up, the dmNes+RG endfoot ratchets down, and the process repeats. We show that dmNes+RG secrete a factor that promotes loss of cell polarity and delamination. This activity is mimicked by a secreted variant of Crumbs2 (CRB2S) which is specifically expressed by dmNes+RG. In cultured MDCK cells, CRB2S associates with apical membranes and decreases cell cohesion. Analysis of Crb2F/F/Nestin-Cre+/- mice, and targeted reduction of Crb2/CRB2S in slice cultures reveal essential roles for transmembrane CRB2 (CRB2TM) and CRB2S on VL cells and dmNes+RG, respectively. We propose a model in which a CRB2S-CRB2TM interaction promotes the progressive attrition of the dVL without loss of overall VL integrity. This novel mechanism may operate more widely to promote orderly progenitor delamination.


Subject(s)
Membrane Proteins/metabolism , Spinal Cord/cytology , Spinal Cord/embryology , Animals , Cell Adhesion , Chick Embryo , Dogs , Gene Expression Regulation, Developmental , HEK293 Cells , Humans , Madin Darby Canine Kidney Cells , Membrane Proteins/genetics , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , SOXB1 Transcription Factors/genetics , SOXB1 Transcription Factors/metabolism , Tight Junctions/metabolism , Time-Lapse Imaging
6.
J Anat ; 236(2): 334-350, 2020 02.
Article in English | MEDLINE | ID: mdl-31670387

ABSTRACT

The ventricular layer of the spinal cord is remodelled during embryonic development and ultimately forms the ependymal cell lining of the adult central canal, which retains neural stem cell potential. This anatomical transformation involves the process of dorsal collapse; however, accompanying changes in tissue organisation and cell behaviour as well as the precise origin of cells contributing to the central canal are not well understood. Here, we describe sequential localised cell rearrangements which accompany the gradual attrition of the spinal cord ventricular layer during development. This includes local breakdown of the pseudostratified organisation of the dorsal ventricular layer prefiguring dorsal collapse and evidence for a new phenomenon, ventral dissociation, during which the ventral-most floor plate cells separate from a subset that are retained around the central canal. Using cell proliferation markers and cell-cycle reporter mice, we further show that following dorsal collapse, ventricular layer attrition involves an overall reduction in cell proliferation, characterised by an intriguing increase in the percentage of cells in G1/S. In contrast, programmed cell death does not contribute to ventricular layer remodelling. By analysing transcript and protein expression patterns associated with key signalling pathways, we provide evidence for a gradual decline in ventral sonic hedgehog activity and an accompanying ventral expansion of initial dorsal bone morphogenetic protein signalling, which comes to dominate the forming the central canal lining. This study identifies multiple steps that may contribute to spinal cord ventricular layer attrition and adds to increasing evidence for the heterogeneous origin of the spinal cord ependymal cell population, which includes cells from the floor plate and the roof plate as well as ventral progenitor domains.


Subject(s)
Cell Proliferation/physiology , Cerebral Ventricles/cytology , Spinal Cord/cytology , Animals , Apoptosis/physiology , Bone Morphogenetic Proteins/metabolism , Cell Cycle/physiology , Cerebral Ventricles/metabolism , Ependyma/cytology , Ependyma/metabolism , Hedgehog Proteins/metabolism , Mice , SOXB1 Transcription Factors/metabolism , Signal Transduction/physiology , Spinal Cord/metabolism
7.
EMBO Rep ; 21(1): e48469, 2020 01 07.
Article in English | MEDLINE | ID: mdl-31789450

ABSTRACT

Amino acids are essential for cellular metabolism, and it is important to understand how nutrient supply is coordinated with changing energy requirements during embryogenesis. Here, we show that the amino acid transporter Slc7a5/Lat1 is highly expressed in tissues undergoing morphogenesis and that Slc7a5-null mouse embryos have profound neural and limb bud outgrowth defects. Slc7a5-null neural tissue exhibited aberrant mTORC1 activity and cell proliferation; transcriptomics, protein phosphorylation and apoptosis analyses further indicated induction of the integrated stress response as a potential cause of observed defects. The pattern of stress response gene expression induced in Slc7a5-null embryos was also detected at low level in wild-type embryos and identified stress vulnerability specifically in tissues undergoing morphogenesis. The Slc7a5-null phenotype is reminiscent of Wnt pathway mutants, and we show that Wnt/ß-catenin loss inhibits Slc7a5 expression and induces this stress response. Wnt signalling therefore normally supports the metabolic demands of morphogenesis and constrains cellular stress. Moreover, operation in the embryo of the integrated stress response, which is triggered by pathogen-mediated as well as metabolic stress, may provide a mechanistic explanation for a range of developmental defects.


Subject(s)
Large Neutral Amino Acid-Transporter 1 , Wnt Signaling Pathway , Animals , Cell Proliferation/genetics , Large Neutral Amino Acid-Transporter 1/metabolism , Mechanistic Target of Rapamycin Complex 1/genetics , Mechanistic Target of Rapamycin Complex 1/metabolism , Mice , Morphogenesis
8.
Development ; 145(19)2018 10 02.
Article in English | MEDLINE | ID: mdl-30201686

ABSTRACT

The vertebrate body forms by continuous generation of new tissue from progenitors at the posterior end of the embryo. The study of these axial progenitors has proved to be challenging in vivo largely because of the lack of unique molecular markers to identify them. Here, we elucidate the expression pattern of the transcription factor Nkx1-2 in the mouse embryo and show that it identifies axial progenitors throughout body axis elongation, including neuromesodermal progenitors and early neural and mesodermal progenitors. We create a tamoxifen-inducible Nkx1-2CreERT2 transgenic mouse and exploit the conditional nature of this line to uncover the lineage contributions of Nkx1-2-expressing cells at specific stages. We show that early Nkx1-2-expressing epiblast cells contribute to all three germ layers, mostly neuroectoderm and mesoderm, excluding notochord. Our data are consistent with the presence of some self-renewing axial progenitors that continue to generate neural and mesoderm tissues from the tail bud. This study identifies Nkx1-2-expressing cells as the source of most trunk and tail tissues in the mouse and provides a useful tool to genetically label and manipulate axial progenitors in vivo.


Subject(s)
Cell Lineage , Homeodomain Proteins/metabolism , Integrases/metabolism , Nuclear Proteins/metabolism , Stem Cells/cytology , Tail/embryology , Torso/embryology , Transcription Factors/metabolism , Animals , Body Patterning , Embryo, Mammalian/cytology , Embryo, Mammalian/metabolism , Genes, Reporter , Mesoderm/cytology , Mice, Inbred C57BL , Mice, Transgenic , Neurons/cytology , Neurons/metabolism , SOXB1 Transcription Factors/metabolism , Tail/cytology
9.
Development ; 145(14)2018 07 30.
Article in English | MEDLINE | ID: mdl-30061166

ABSTRACT

The Myc transcriptional regulators are implicated in a range of cellular functions, including proliferation, cell cycle progression, metabolism and pluripotency maintenance. Here, we investigated the expression, regulation and function of the Myc family during mouse embryonic axis elongation and segmentation. Expression of both cMyc (Myc - Mouse Genome Informatics) and MycN in the domains in which neuromesodermal progenitors (NMPs) and underlying caudal pre-somitic mesoderm (cPSM) cells reside is coincident with WNT and FGF signals, factors known to maintain progenitors in an undifferentiated state. Pharmacological inhibition of Myc activity downregulates expression of WNT/FGF components. In turn, we find that cMyc expression is WNT, FGF and Notch protein regulated, placing it centrally in the signalling circuit that operates in the tail end that both sustains progenitors and drives maturation of the PSM into somites. Interfering with Myc function in the PSM, where it displays oscillatory expression, delays the timing of segmentation clock oscillations and thus of somite formation. In summary, we identify Myc as a component that links NMP maintenance and PSM maturation during the body axis elongation stages of mouse embryogenesis.


Subject(s)
Biological Clocks/genetics , Body Patterning/genetics , CLOCK Proteins/genetics , Mesoderm/cytology , Mesoderm/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Signal Transduction/genetics , Stem Cells/metabolism , Animals , CLOCK Proteins/metabolism , Cell Differentiation , Down-Regulation/genetics , Embryo, Mammalian/metabolism , Female , Fibroblast Growth Factor 8/metabolism , Fibroblast Growth Factors/metabolism , Gene Expression Regulation, Developmental , Humans , Mesoderm/embryology , Mice , RNA, Messenger/genetics , RNA, Messenger/metabolism , Somites/embryology , Somites/metabolism , Stem Cells/cytology , Tail/embryology , Wnt Proteins/metabolism
10.
Development ; 145(16)2018 07 12.
Article in English | MEDLINE | ID: mdl-29899136

ABSTRACT

Robust protocols for directed differentiation of human pluripotent cells are required to determine whether mechanisms operating in model organisms are relevant to our own development. Recent work in vertebrate embryos has identified neuromesodermal progenitors as a bipotent cell population that contributes to paraxial mesoderm and spinal cord. However, precise protocols for in vitro differentiation of human spinal cord progenitors are lacking. Informed by signalling in amniote embryos, we show here that transient dual-SMAD inhibition, together with retinoic acid (dSMADi-RA), provides rapid and reproducible induction of human spinal cord progenitors from neuromesodermal progenitor-like cells. Using CRISPR-Cas9 to engineer human embryonic stem cells with a GFP-reporter for neuromesodermal progenitor-associated gene Nkx1.2 we facilitate selection of this cell population. RNA-sequencing was then used to identify human and conserved neuromesodermal progenitor transcriptional signatures, to validate this differentiation protocol and to reveal new pathways/processes in human neural differentiation. This optimised protocol, novel reporter line and transcriptomic data are useful resources with which to dissect molecular mechanisms regulating human spinal cord generation and allow the scaling-up of distinct cell populations for global analyses, including proteomic, biochemical and chromatin interrogation.


Subject(s)
Cell Differentiation , Cell Lineage , Gene Expression Profiling , Mesoderm/physiology , Neural Stem Cells/physiology , Neurogenesis/physiology , Spinal Cord/physiology , Animals , Body Patterning/genetics , Cell Differentiation/genetics , Cell Lineage/genetics , Cells, Cultured , Embryo, Mammalian , Gene Expression Regulation, Developmental , Humans , Mesoderm/cytology , Mesoderm/embryology , Mice , Neural Stem Cells/cytology , Neurogenesis/genetics , Spinal Cord/cytology , Stem Cells/cytology , Stem Cells/physiology
11.
Int J Dev Biol ; 62(1-2-3): 167-175, 2018.
Article in English | MEDLINE | ID: mdl-29616725

ABSTRACT

Signalling pathways that regulate neural progenitor proliferation and neuronal differentiation have been identified. However, we know much less about how transduction of such signals is regulated within neuroepithelial cells to direct cell fate choice during mitosis and subsequent neuronal differentiation. Here we review recent advances in the experimentally amenable chick embryo, which reveal that this involves association of signalling pathway components with cell biological entities, including mitotic centrosomes and ciliary structures. This includes changing centrosomal localization of protein kinase A, which regulates Sonic hedgehog signalling and so neural progenitor status, and Mindbomb1, a mediator of Notch ligand activation, which promotes Notch signalling in neighbouring cells, and so is active in presumptive neurons. We further review cell biological events that underlie the later step of neuronal delamination, during which a newborn neuron detaches from its neighbouring cells and undergoes a process known as apical abscission. This involves inter-dependent actin and microtubule dynamics and includes dissociation of the centrosome from the ciliary membrane, which potentially alters the signalling repertoire of this now post-mitotic cell. Open questions and future directions are discussed along with technological advances which improve accuracy of gene manipulation, monitoring of protein dynamics and quantification of cell biological processes in living tissues.


Subject(s)
Chick Embryo , Neurogenesis , Actins/metabolism , Animals , Cadherins/metabolism , Cell Differentiation/physiology , Centrosome/metabolism , Centrosome/ultrastructure , Chickens , Cyclic AMP-Dependent Protein Kinases/metabolism , Developmental Biology , Embryonic Development , Hedgehog Proteins/metabolism , Ligands , Mitosis , Nervous System/metabolism , Neural Tube/ultrastructure , Neurons/metabolism , Receptors, Notch/metabolism , Signal Transduction
12.
Elife ; 62017 10 23.
Article in English | MEDLINE | ID: mdl-29058679

ABSTRACT

Detachment of newborn neurons from the neuroepithelium is required for correct neuronal architecture and functional circuitry. This process, also known as delamination, involves adherens-junction disassembly and acto-myosin-mediated abscission, during which the centrosome is retained while apical/ciliary membranes are shed. Cell-biological mechanisms mediating delamination are, however, poorly understood. Using live-tissue and super-resolution imaging, we uncover a centrosome-nucleated wheel-like microtubule configuration, aligned with the apical actin cable and adherens-junctions within chick and mouse neuroepithelial cells. These microtubules maintain adherens-junctions while actin maintains microtubules, adherens-junctions and apical end-foot dimensions. During neuronal delamination, acto-myosin constriction generates a tunnel-like actin-microtubule configuration through which the centrosome translocates. This movement requires inter-dependent actin and microtubule activity, and we identify drebrin as a potential coordinator of these cytoskeletal dynamics. Furthermore, centrosome compromise revealed that this organelle is required for delamination. These findings identify new cytoskeletal configurations and regulatory relationships that orchestrate neuronal delamination and may inform mechanisms underlying pathological epithelial cell detachment.


Subject(s)
Actins/metabolism , Cell Differentiation , Centrosome/metabolism , Microtubules/metabolism , Morphogenesis , Nervous System/embryology , Neurons/physiology , Animals , Biological Transport , Chick Embryo , Intravital Microscopy , Microscopy, Fluorescence
13.
Elife ; 5: e14830, 2016 03 07.
Article in English | MEDLINE | ID: mdl-26949247

ABSTRACT

Experiments in mice shed new light on an elusive population of embryonic cells called neuromesodermal progenitors.


Subject(s)
Gene Expression Regulation, Developmental , Mesoderm/embryology , Animals , Mice
14.
Development ; 142(17): 2864-75, 2015 Sep 01.
Article in English | MEDLINE | ID: mdl-26329597

ABSTRACT

Neuromesodermal progenitors (NMps) contribute to both the elongating spinal cord and the adjacent paraxial mesoderm. It has been assumed that these cells arise as a result of patterning of the anterior neural plate. However, as the molecular mechanisms that specify NMps in vivo are uncovered, and as protocols for generating these bipotent cells from mouse and human pluripotent stem cells in vitro are established, the emerging data suggest that this view needs to be revised. Here, we review the characteristics, regulation, in vitro derivation and in vivo induction of NMps. We propose that these cells arise within primitive streak-associated epiblast via a mechanism that is separable from that which establishes neural fate in the anterior epiblast. We thus argue for the existence of two distinct routes for making central nervous system progenitors.


Subject(s)
Mesoderm/cytology , Spinal Cord/cytology , Spinal Cord/embryology , Stem Cells/cytology , Animals , Body Patterning , Embryo, Mammalian/cytology , Humans , Neural Stem Cells/cytology , Signal Transduction
15.
Development ; 141(16): 3266-76, 2014 Aug.
Article in English | MEDLINE | ID: mdl-25063452

ABSTRACT

Here, we exploit the spatial separation of temporal events of neural differentiation in the elongating chick body axis to provide the first analysis of transcriptome change in progressively more differentiated neural cell populations in vivo. Microarray data, validated against direct RNA sequencing, identified: (1) a gene cohort characteristic of the multi-potent stem zone epiblast, which contains neuro-mesodermal progenitors that progressively generate the spinal cord; (2) a major transcriptome re-organisation as cells then adopt a neural fate; and (3) increasing diversity as neural patterning and neuron production begin. Focussing on the transition from multi-potent to neural state cells, we capture changes in major signalling pathways, uncover novel Wnt and Notch signalling dynamics, and implicate new pathways (mevalonate pathway/steroid biogenesis and TGFß). This analysis further predicts changes in cellular processes, cell cycle, RNA-processing and protein turnover as cells acquire neural fate. We show that these changes are conserved across species and provide biological evidence for reduced proteasome efficiency and a novel lengthening of S phase. This latter step may provide time for epigenetic events to mediate large-scale transcriptome re-organisation; consistent with this, we uncover simultaneous downregulation of major chromatin modifiers as the neural programme is established. We further demonstrate that transcription of one such gene, HDAC1, is dependent on FGF signalling, making a novel link between signals that control neural differentiation and transcription of a core regulator of chromatin organisation. Our work implicates new signalling pathways and dynamics, cellular processes and epigenetic modifiers in neural differentiation in vivo, identifying multiple new potential cellular and molecular mechanisms that direct differentiation.


Subject(s)
Chromatin/metabolism , Neurogenesis/physiology , Neurons/cytology , Transcriptome , Animals , Body Patterning , Cell Cycle , Cell Differentiation , Cell Lineage , Chick Embryo , Epigenesis, Genetic , Fibroblast Growth Factors/metabolism , Gene Expression Regulation, Developmental , Histone Deacetylase 1/metabolism , Mice , Oligonucleotide Array Sequence Analysis , Sequence Analysis, RNA , Signal Transduction , Spinal Cord/embryology , Time Factors , Transforming Growth Factor beta/metabolism
16.
PLoS One ; 9(4): e94270, 2014.
Article in English | MEDLINE | ID: mdl-24722185

ABSTRACT

The reference annotations made for a genome sequence provide the framework for all subsequent analyses of the genome. Correct and complete annotation in addition to the underlying genomic sequence is particularly important when interpreting the results of RNA-seq experiments where short sequence reads are mapped against the genome and assigned to genes according to the annotation. Inconsistencies in annotations between the reference and the experimental system can lead to incorrect interpretation of the effect on RNA expression of an experimental treatment or mutation in the system under study. Until recently, the genome-wide annotation of 3' untranslated regions received less attention than coding regions and the delineation of intron/exon boundaries. In this paper, data produced for samples in Human, Chicken and A. thaliana by the novel single-molecule, strand-specific, Direct RNA Sequencing technology from Helicos Biosciences which locates 3' polyadenylation sites to within +/- 2 nt, were combined with archival EST and RNA-Seq data. Nine examples are illustrated where this combination of data allowed: (1) gene and 3' UTR re-annotation (including extension of one 3' UTR by 5.9 kb); (2) disentangling of gene expression in complex regions; (3) clearer interpretation of small RNA expression and (4) identification of novel genes. While the specific examples displayed here may become obsolete as genome sequences and their annotations are refined, the principles laid out in this paper will be of general use both to those annotating genomes and those seeking to interpret existing publically available annotations in the context of their own experimental data.


Subject(s)
3' Untranslated Regions , Computational Biology/methods , Expressed Sequence Tags , Sequence Analysis, RNA/methods , Animals , Arabidopsis/genetics , Cell Cycle Proteins/genetics , Chickens/genetics , Genetic Loci/genetics , Genome , Genome, Human , Genome, Plant , Genomics , Humans , Methyltransferases/genetics , Models, Statistical , RNA, Messenger/metabolism , Ribosomal Proteins/genetics , Skin/metabolism
17.
Science ; 343(6167): 200-4, 2014 Jan 10.
Article in English | MEDLINE | ID: mdl-24408437

ABSTRACT

Withdrawal of differentiating cells from proliferative tissue is critical for embryonic development and adult tissue homeostasis; however, the mechanisms that control this cell behavior are poorly understood. Using high-resolution live-cell imaging in chick neural tube, we uncover a form of cell subdivision that abscises apical cell membrane and mediates neuron detachment from the ventricle. This mechanism operates in chick and mouse, is dependent on actin-myosin contraction, and results in loss of apical cell polarity. Apical abscission also dismantles the primary cilium, known to transduce sonic-hedgehog signals, and is required for expression of cell-cycle-exit gene p27/Kip1. We further show that N-cadherin levels, regulated by neuronal-differentiation factor Neurog2, determine cilium disassembly and final abscission. This cell-biological mechanism may mediate such cell transitions in other epithelia in normal and cancerous conditions.


Subject(s)
Cell Polarity , Neurogenesis , Neurons/cytology , Actins/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cadherins/metabolism , Carcinogenesis/pathology , Cell Division , Chick Embryo , Cilia/ultrastructure , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Mice , Myosins/metabolism , Nerve Tissue Proteins/metabolism , Neural Tube/growth & development , Neural Tube/metabolism , Neurons/metabolism , Neurons/ultrastructure
18.
F1000Res ; 3: 275, 2014.
Article in English | MEDLINE | ID: mdl-25580236

ABSTRACT

Mesodermal and spinal cord progenitors originate from common founder cells from which they segregate during development. Moreover, neural and mesodermal tissues closely interact during embryogenesis to ensure timely patterning and differentiation of both head and trunk structures. For instance, the fate and morphogenesis of neural progenitors is dependent on signals produced by mesodermal cells and vice-versa. While some of the cellular and molecular signals that mediate these interactions have been described, much more remains to be uncovered. The scope of this collection will cover these interactions between neural (CNS or PNS) and mesodermal progenitors in patterning body plans and specific body systems in vertebrate embryos. This includes, but is not limited to, interactions influencing the formation of body axes, neural tube formation, neural crest migration, gut development, muscle patterning and myogenesis.

19.
PLoS Genet ; 9(7): e1003614, 2013.
Article in English | MEDLINE | ID: mdl-23874217

ABSTRACT

Changes in higher order chromatin organisation have been linked to transcriptional regulation; however, little is known about how such organisation alters during embryonic development or how it is regulated by extrinsic signals. Here we analyse changes in chromatin organisation as neural differentiation progresses, exploiting the clear spatial separation of the temporal events of differentiation along the elongating body axis of the mouse embryo. Combining fluorescence in situ hybridisation with super-resolution structured illumination microscopy, we show that chromatin around key differentiation gene loci Pax6 and Irx3 undergoes both decompaction and displacement towards the nuclear centre coincident with transcriptional onset. Conversely, down-regulation of Fgf8 as neural differentiation commences correlates with a more peripheral nuclear position of this locus. During normal neural differentiation, fibroblast growth factor (FGF) signalling is repressed by retinoic acid, and this vitamin A derivative is further required for transcription of neural genes. We show here that exposure to retinoic acid or inhibition of FGF signalling promotes precocious decompaction and central nuclear positioning of differentiation gene loci. Using the Raldh2 mutant as a model for retinoid deficiency, we further find that such changes in higher order chromatin organisation are dependent on retinoid signalling. In this retinoid deficient condition, FGF signalling persists ectopically in the elongating body, and importantly, we find that inhibiting FGF receptor (FGFR) signalling in Raldh2-/- embryos does not rescue differentiation gene transcription, but does elicit both chromatin decompaction and nuclear position change. These findings demonstrate that regulation of higher order chromatin organisation during differentiation in the embryo can be uncoupled from the machinery that promotes transcription and, for the first time, identify FGF as an extrinsic signal that can direct chromatin compaction and nuclear organisation of gene loci.


Subject(s)
Cell Differentiation/genetics , Chromatin/genetics , Fibroblast Growth Factor 8/genetics , Neurogenesis , Receptors, Fibroblast Growth Factor/genetics , Aldehyde Oxidoreductases/genetics , Animals , Embryonic Development/genetics , Eye Proteins/genetics , Homeodomain Proteins/genetics , Mice , PAX6 Transcription Factor , Paired Box Transcription Factors/genetics , Receptors, Fibroblast Growth Factor/metabolism , Repressor Proteins/genetics , Signal Transduction , Transcription Factors/genetics , Transcription, Genetic , Tretinoin/metabolism , Tretinoin/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL