Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 114
Filter
1.
Cell ; 174(4): 999-1014.e22, 2018 08 09.
Article in English | MEDLINE | ID: mdl-30096314

ABSTRACT

The mammalian nervous system executes complex behaviors controlled by specialized, precisely positioned, and interacting cell types. Here, we used RNA sequencing of half a million single cells to create a detailed census of cell types in the mouse nervous system. We mapped cell types spatially and derived a hierarchical, data-driven taxonomy. Neurons were the most diverse and were grouped by developmental anatomical units and by the expression of neurotransmitters and neuropeptides. Neuronal diversity was driven by genes encoding cell identity, synaptic connectivity, neurotransmission, and membrane conductance. We discovered seven distinct, regionally restricted astrocyte types that obeyed developmental boundaries and correlated with the spatial distribution of key glutamate and glycine neurotransmitters. In contrast, oligodendrocytes showed a loss of regional identity followed by a secondary diversification. The resource presented here lays a solid foundation for understanding the molecular architecture of the mammalian nervous system and enables genetic manipulation of specific cell types.


Subject(s)
Gene Expression Regulation, Developmental , Gene Regulatory Networks , Nervous System/metabolism , Single-Cell Analysis/methods , Transcriptome , Animals , Female , Gene Expression Profiling , High-Throughput Nucleotide Sequencing , Male , Mice , Mice, Inbred C57BL , Nervous System/growth & development
2.
Cell ; 157(2): 313-328, 2014 04 10.
Article in English | MEDLINE | ID: mdl-24656405

ABSTRACT

Glioblastoma multiforme (GBM) is the most aggressive form of brain cancer with marginal life expectancy. Based on the assumption that GBM cells gain functions not necessarily involved in the cancerous process, patient-derived glioblastoma cells (GCs) were screened to identify cellular processes amenable for development of targeted treatments. The quinine-derivative NSC13316 reliably and selectively compromised viability. Synthetic chemical expansion reveals delicate structure-activity relationship and analogs with increased potency, termed Vacquinols. Vacquinols stimulate death by membrane ruffling, cell rounding, massive macropinocytic vacuole accumulation, ATP depletion, and cytoplasmic membrane rupture of GCs. The MAP kinase MKK4, identified by a shRNA screen, represents a critical signaling node. Vacquinol-1 displays excellent in vivo pharmacokinetics and brain exposure, attenuates disease progression, and prolongs survival in a GBM animal model. These results identify a vulnerability to massive vacuolization that can be targeted by small molecules and point to the possible exploitation of this process in the design of anticancer therapies.


Subject(s)
Brain Neoplasms/drug therapy , Brain Neoplasms/pathology , Glioblastoma/drug therapy , Glioblastoma/pathology , Piperidines/pharmacology , Quinolines/pharmacology , Small Molecule Libraries/pharmacology , Animals , Cell Death/drug effects , Heterografts , Humans , Hydroxyquinolines/pharmacology , MAP Kinase Kinase 4/metabolism , Mice , Neoplasm Transplantation , Pinocytosis/drug effects , Vacuoles/metabolism , Zebrafish
4.
Cell ; 139(2): 366-79, 2009 Oct 16.
Article in English | MEDLINE | ID: mdl-19837037

ABSTRACT

Current opinion holds that pigment cells, melanocytes, are derived from neural crest cells produced at the dorsal neural tube and that migrate under the epidermis to populate all parts of the skin. Here, we identify growing nerves projecting throughout the body as a stem/progenitor niche containing Schwann cell precursors (SCPs) from which large numbers of skin melanocytes originate. SCPs arise as a result of lack of neuronal specification by Hmx1 homeobox gene function in the neural crest ventral migratory pathway. Schwann cell and melanocyte development share signaling molecules with both the glial and melanocyte cell fates intimately linked to nerve contact and regulated in an opposing manner by Neuregulin and soluble signals including insulin-like growth factor and platelet-derived growth factor. These results reveal SCPs as a cellular origin of melanocytes, and have broad implications on the molecular mechanisms regulating skin pigmentation during development, in health and pigmentation disorders.


Subject(s)
Melanocytes/cytology , Schwann Cells/cytology , Skin/innervation , Animals , Cell Differentiation , Cell Movement , Homeodomain Proteins , Mice , Neuroglia , Receptor, ErbB-3/metabolism , Stem Cells/cytology , Transcription Factors/metabolism
5.
Diabetologia ; 66(12): 2332-2345, 2023 12.
Article in English | MEDLINE | ID: mdl-37728731

ABSTRACT

AIMS/HYPOTHESIS: Our aim was to investigate structural changes of cutaneous Schwann cells (SCs), including nociceptive Schwann cells (nSCs) and axons, in individuals with diabetic polyneuropathy. We also aimed to investigate the relationship between these changes and peripheral neuropathic symptoms in type 1 diabetes. METHODS: Skin biopsies (3 mm) taken from carefully phenotyped participants with type 1 diabetes without polyneuropathy (T1D, n=25), type 1 diabetes with painless diabetic polyneuropathy (T1DPN, n=30) and type 1 diabetes with painful diabetic polyneuropathy (P-T1DPN, n=27), and from healthy control individuals (n=25) were immunostained with relevant antibodies to visualise SCs and nerve fibres. Stereological methods were used to quantify the expression of cutaneous SCs and nerve fibres. RESULTS: There was a difference in the number density of nSCs not abutting to nerve fibres between the groups (p=0.004) but not in the number density of nSCs abutting to nerve fibres, nor in solitary or total subepidermal SC soma number density. The overall dermal SC expression (measured by dermal SC area fraction and subepidermal SC process density) and peripheral nerve fibre expression (measured by intraepidermal nerve fibre density, dermal nerve fibre area fraction and subepidermal nerve fibre density) differed between the groups (all p<0.05): significant differences were seen in participants with T1DPN and P-T1DPN compared with those without diabetic polyneuropathy (healthy control and T1D groups) (all p<0.05). No difference was found between participants in the T1DPN and P-T1DPN group, nor between participants in the T1D and healthy control group (all p>0.05). Correlational analysis showed that cutaneous SC processes and nerve fibres were highly associated, and they were weakly negatively correlated with different neuropathy measures. CONCLUSIONS/INTERPRETATION: Cutaneous SC processes and nerves, but not SC soma, are degenerated and interdependent in individuals with diabetic polyneuropathy. However, an increase in structurally damaged nSCs was seen in individuals with diabetic polyneuropathy. Furthermore, dermal SC processes and nerve fibres correlate weakly with clinical measures of neuropathy and may play a partial role in the pathophysiology of diabetic polyneuropathy in type 1 diabetes.


Subject(s)
Diabetes Mellitus, Type 1 , Diabetic Neuropathies , Humans , Diabetes Mellitus, Type 1/complications , Nerve Fibers/pathology , Peripheral Nerves/pathology , Schwann Cells/pathology
6.
Development ; 146(20)2019 10 24.
Article in English | MEDLINE | ID: mdl-31575648

ABSTRACT

The control of all our motor outputs requires constant monitoring by proprioceptive sensory neurons (PSNs) that convey continuous muscle sensory inputs to the spinal motor network. Yet the molecular programs that control the establishment of this sensorimotor circuit remain largely unknown. The transcription factor RUNX3 is essential for the early steps of PSNs differentiation, making it difficult to study its role during later aspects of PSNs specification. Here, we conditionally inactivate Runx3 in PSNs after peripheral innervation and identify that RUNX3 is necessary for maintenance of cell identity of only a subgroup of PSNs, without discernable cell death. RUNX3 also controls the sensorimotor connection between PSNs and motor neurons at limb level, with muscle-by-muscle variable sensitivities to the loss of Runx3 that correlate with levels of RUNX3 in PSNs. Finally, we find that muscles and neurotrophin 3 signaling are necessary for maintenance of RUNX3 expression in PSNs. Hence, a transcriptional regulator that is crucial for specifying a generic PSN type identity after neurogenesis is later regulated by target muscle-derived signals to contribute to the specialized aspects of the sensorimotor connection selectivity.


Subject(s)
Core Binding Factor Alpha 3 Subunit/metabolism , Animals , Cell Differentiation/genetics , Cell Differentiation/physiology , Cells, Cultured , Core Binding Factor Alpha 3 Subunit/genetics , Female , Ganglia, Spinal/cytology , Ganglia, Spinal/metabolism , Gene Expression Regulation, Developmental , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , LIM-Homeodomain Proteins/genetics , LIM-Homeodomain Proteins/metabolism , Male , Mice , Mice, Inbred C57BL , Microfilament Proteins/genetics , Microfilament Proteins/metabolism , Motor Neurons/metabolism , Muscle Proteins/genetics , Muscle Proteins/metabolism , Nerve Growth Factors/genetics , Nerve Growth Factors/metabolism , Sensory Receptor Cells/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
7.
Development ; 145(18)2018 09 25.
Article in English | MEDLINE | ID: mdl-30126905

ABSTRACT

Touch and mechanical sensations require the development of several different kinds of sensory neurons dedicated to respond to certain types of mechanical stimuli. The transcription factor Shox2 (short stature homeobox 2) is involved in the generation of TRKB+ low-threshold mechanoreceptors (LTMRs), but mechanisms terminating this program and allowing alternative fates are unknown. Here, we show that the conditional loss of the miR-183-96-182 cluster in mouse leads to a failure of extinction of Shox2 during development and an increase in the proportion of Aδ LTMRs (TRKB+/NECAB2+) neurons at the expense of Aß slowly adapting (SA)-LTMRs (TRKC+/Runx3-) neurons. Conversely, overexpression of miR-183 cluster that represses Shox2 expression, or loss of Shox2, both increase the Aß SA-LTMRs population at the expense of Aδ LTMRs. Our results suggest that the miR-183 cluster determines the timing of Shox2 expression by direct targeting during development, and through this determines the population sizes of Aδ LTMRs and Aß SA-LTMRs.


Subject(s)
Homeodomain Proteins/metabolism , Mechanoreceptors/metabolism , MicroRNAs/genetics , Sensory Receptor Cells/cytology , Animals , Calcium-Binding Proteins/metabolism , Cell Differentiation/genetics , Eye Proteins/metabolism , Female , Humans , Membrane Glycoproteins/metabolism , Mice , Mice, Knockout , Pregnancy , Protein-Tyrosine Kinases/metabolism
8.
Liver Int ; 40(4): 977-987, 2020 04.
Article in English | MEDLINE | ID: mdl-32011099

ABSTRACT

BACKGROUND AND AIMS: Liver fibrosis results from cycles of liver damage and scar formation. We herein aimed at analysing neural crest cells and/or bone marrow stromal cells contribution to the liver. METHODS: Two liver fibrosis and one hepatectomy model were applied on double-transgenic loxP-Cre mouse lines. RESULTS: Increased numbers of glia with more complex processes were found in fibrotic livers. During embryonic development, only few cells were traced in the liver and bone marrow, in a minor fraction of mice of different neural crest reporter strains analysed: therefore, a neural crest origin of such cells is doubtful. In the fibrotic liver, a significantly higher incidence of endothelial cells and hepatocyte-like cells expressing the reporter gene Tomato were found in Wnt1-Cre-Tom and GLAST-CreERT2-Tom mice. Consistently, during early fibrogenesis stromal Wnt1-traced cells, with progenitor (CFU-F) properties, get likely mobilized to peripheral blood. Circulating adult Wnt1-traced cells are stromal cells and lack from the expression of other bone marrow and endothelial progenitor cells markers. Furthermore, in a 70% hepatectomy model GLAST+ Wnt1-traced pericytes were found to be mobilized from the bone marrow and the incidence of GLAST-traced hepatocyte-like cells was increased. Finally, GLAST-traced hepatocyte like-cells were found to maintain the expression of stromal markers. CONCLUSIONS: Our data suggest a gliosis process during liver fibrogenesis. While neural crest cells probably do not contribute with other liver cell types than glia, GLAST+ Wnt1-traced bone marrow pericytes are likely a source of endothelial and hepatocyte-like cells after liver injury and do not contribute to scarring.


Subject(s)
Neural Crest , Pericytes , Animals , Bone Marrow , Endothelial Cells , Liver , Liver Regeneration , Mice , Mice, Transgenic
9.
Nature ; 513(7519): 551-4, 2014 Sep 25.
Article in English | MEDLINE | ID: mdl-25079316

ABSTRACT

Mesenchymal stem cells occupy niches in stromal tissues where they provide sources of cells for specialized mesenchymal derivatives during growth and repair. The origins of mesenchymal stem cells have been the subject of considerable discussion, and current consensus holds that perivascular cells form mesenchymal stem cells in most tissues. The continuously growing mouse incisor tooth offers an excellent model to address the origin of mesenchymal stem cells. These stem cells dwell in a niche at the tooth apex where they produce a variety of differentiated derivatives. Cells constituting the tooth are mostly derived from two embryonic sources: neural crest ectomesenchyme and ectodermal epithelium. It has been thought for decades that the dental mesenchymal stem cells giving rise to pulp cells and odontoblasts derive from neural crest cells after their migration in the early head and formation of ectomesenchymal tissue. Here we show that a significant population of mesenchymal stem cells during development, self-renewal and repair of a tooth are derived from peripheral nerve-associated glia. Glial cells generate multipotent mesenchymal stem cells that produce pulp cells and odontoblasts. By combining a clonal colour-coding technique with tracing of peripheral glia, we provide new insights into the dynamics of tooth organogenesis and growth.


Subject(s)
Cell Differentiation , Cell Lineage , Incisor/cytology , Mesenchymal Stem Cells/cytology , Neuroglia/cytology , Animals , Cell Tracking , Clone Cells/cytology , Dental Pulp/cytology , Female , Incisor/embryology , Male , Mice , Models, Biological , Neural Crest/cytology , Odontoblasts/cytology , Regeneration , Schwann Cells/cytology
10.
Dev Biol ; 444 Suppl 1: S308-S324, 2018 12 01.
Article in English | MEDLINE | ID: mdl-29807017

ABSTRACT

Carotid body glomus cells mediate essential reflex responses to arterial blood hypoxia. They are dopaminergic and secrete growth factors that support dopaminergic neurons, making the carotid body a potential source of patient-specific cells for Parkinson's disease therapy. Like adrenal chromaffin cells, which are also hypoxia-sensitive, glomus cells are neural crest-derived and require the transcription factors Ascl1 and Phox2b; otherwise, their development is little understood at the molecular level. Here, analysis in chicken and mouse reveals further striking molecular parallels, though also some differences, between glomus and adrenal chromaffin cell development. Moreover, histology has long suggested that glomus cell precursors are 'émigrés' from neighbouring ganglia/nerves, while multipotent nerve-associated glial cells are now known to make a significant contribution to the adrenal chromaffin cell population in the mouse. We present conditional genetic lineage-tracing data from mice supporting the hypothesis that progenitors expressing the glial marker proteolipid protein 1, presumably located in adjacent ganglia/nerves, also contribute to glomus cells. Finally, we resolve a paradox for the 'émigré' hypothesis in the chicken - where the nearest ganglion to the carotid body is the nodose, in which the satellite glia are neural crest-derived, but the neurons are almost entirely placode-derived - by fate-mapping putative nodose neuronal 'émigrés' to the neural crest.


Subject(s)
Carotid Body/embryology , Chromaffin Cells/metabolism , Pericytes/metabolism , Adrenal Glands/metabolism , Adrenal Glands/physiology , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Body Patterning/physiology , Cell Differentiation , Cell Hypoxia/physiology , Chick Embryo , Chickens/metabolism , Mice , Mice, Knockout , Myelin Proteolipid Protein/physiology , Neural Crest/metabolism , Neurons/metabolism , Pericytes/physiology , Transcription Factors/metabolism
11.
Stem Cells ; 36(11): 1736-1751, 2018 11.
Article in English | MEDLINE | ID: mdl-29999568

ABSTRACT

Adult neurogenesis in the brain continuously seeds new neurons throughout life, but how homeostasis of adult neural stem cells (NSCs) is maintained is incompletely understood. Here, we demonstrate that the DNA methylation adapter ubiquitin-like, containing PHD and RING finger domains-1 (UHRF1) is expressed in, and regulates proliferation of, the active but not quiescent pool of adult neural progenitor cells. Mice with a neural stem cell-specific deficiency in UHRF1 exhibit a massive depletion of neurogenesis resulting in a collapse of formation of new neurons. In the absence of UHRF1, NSCs unexpectedly remain in the cell cycle but with a 17-fold increased cell cycle length due to a failure of replication phase entry caused by promoter demethylation and derepression of Cdkn1a, which encodes the cyclin-dependent kinase inhibitor p21. UHRF1 does not affect the proportion progenitor cells active within the cell cycle but among these cells, UHRF1 is critical for licensing replication re-entry. Therefore, this study shows that a UHRF1-Cdkn1a axis is essential for the control of stem cell self-renewal and neurogenesis in the adult brain. Stem Cells 2018;36:1736-1751.


Subject(s)
Adult Stem Cells/metabolism , Neural Stem Cells/metabolism , Nuclear Proteins/genetics , Animals , CCAAT-Enhancer-Binding Proteins , Humans , Mice , Nuclear Proteins/metabolism , Ubiquitin-Protein Ligases
12.
Am J Hum Genet ; 96(4): 519-31, 2015 Apr 02.
Article in English | MEDLINE | ID: mdl-25772936

ABSTRACT

The endothelin receptor type A (EDNRA) signaling pathway is essential for the establishment of mandibular identity during development of the first pharyngeal arch. We report four unrelated individuals with the syndrome mandibulofacial dysostosis with alopecia (MFDA) who have de novo missense variants in EDNRA. Three of the four individuals have the same substitution, p.Tyr129Phe. Tyr129 is known to determine the selective affinity of EDNRA for endothelin 1 (EDN1), its major physiological ligand, and the p.Tyr129Phe variant increases the affinity of the receptor for EDN3, its non-preferred ligand, by two orders of magnitude. The fourth individual has a somatic mosaic substitution, p.Glu303Lys, and was previously described as having Johnson-McMillin syndrome. The zygomatic arch of individuals with MFDA resembles that of mice in which EDNRA is ectopically activated in the maxillary prominence, resulting in a maxillary to mandibular transformation, suggesting that the p.Tyr129Phe variant causes an EDNRA gain of function in the developing upper jaw. Our in vitro and in vivo assays suggested complex, context-dependent effects of the EDNRA variants on downstream signaling. Our findings highlight the importance of finely tuned regulation of EDNRA signaling during human craniofacial development and suggest that modification of endothelin receptor-ligand specificity was a key step in the evolution of vertebrate jaws.


Subject(s)
Alopecia/genetics , Mandibulofacial Dysostosis/genetics , Receptor, Endothelin A/genetics , Alopecia/pathology , Animals , Base Sequence , Endothelin-1/metabolism , Exome/genetics , Humans , In Situ Hybridization , Mandibulofacial Dysostosis/pathology , Molecular Sequence Data , Morpholinos/genetics , Mutation, Missense/genetics , Pedigree , RNA, Messenger/administration & dosage , Real-Time Polymerase Chain Reaction , Receptor, Endothelin A/metabolism , Sequence Analysis, DNA , Syndrome , Tomography, X-Ray Computed , Zebrafish , Zygoma/pathology
13.
Nucleic Acids Res ; 44(10): 4745-62, 2016 06 02.
Article in English | MEDLINE | ID: mdl-26939887

ABSTRACT

Defects in DNA replication and repair are known to promote genomic instability, a hallmark of cancer cells. Thus, eukaryotic cells have developed complex mechanisms to ensure accurate duplication of their genomes. While DNA damage response has been extensively studied in tumour cells, the pathways implicated in the response to replication stress are less well understood especially in non-transformed cells. Here we show that in non-transformed cells, APC/C(Cdh1) is activated upon severe replication stress. Activation of APC/C(Cdh1) prevents new origin firing and induces permanent arrest in S-phase. Moreover, Rad51-mediated homologous recombination is also impaired under these conditions. APC/C(Cdh1) activation in S-phase occurs after replication forks have been processed into double strand breaks. Remarkably, this activation, which correlates with decreased Emi1 levels, is not prevented by ATR/ATM inhibition, but it is abrogated in cells depleted of p53 or p21. Importantly, we found that the lack of APC/C(Cdh1) activity correlated with an increase in genomic instability. Taken together, our results define a new APC/C(Cdh1) function that prevents cell cycle resumption after prolonged replication stress by inhibiting origin firing, which may act as an additional mechanism in safeguarding genome integrity.


Subject(s)
Cdh1 Proteins/metabolism , DNA Replication , Replication Origin , S Phase/genetics , Ataxia Telangiectasia Mutated Proteins/metabolism , Cell Line, Tumor , Cells, Cultured , Cyclin-Dependent Kinase Inhibitor p21/physiology , DNA Damage , Enzyme Activation , Genomic Instability , Humans , Hydroxyurea/toxicity , Stress, Physiological/genetics , Tumor Suppressor Protein p53/physiology
14.
EMBO J ; 32(11): 1613-25, 2013 May 29.
Article in English | MEDLINE | ID: mdl-23591430

ABSTRACT

The sympathetic nervous system relies on distinct populations of neurons that use noradrenaline or acetylcholine as neurotransmitter. We show that fating of the sympathetic lineage at early stages results in hybrid precursors from which, genetic cell-lineage tracing reveals, all types progressively emerge by principal mechanisms of maintenance, repression and induction of phenotypes. The homeobox transcription factor HMX1 represses Tlx3 and Ret, induces TrkA and maintains tyrosine hydroxylase (Th) expression in precursors, thus driving segregation of the noradrenergic sympathetic fate. Cholinergic sympathetic neurons develop through cross-regulatory interactions between TRKC and RET in precursors, which lead to Hmx1 repression and sustained Tlx3 expression, thereby resulting in failure of TrkA induction and loss of maintenance of Th expression. Our results provide direct evidence for a model in which diversification of noradrenergic and cholinergic sympathetic neurons is based on a principle of cross-repressive functions in which the specific cell fates are directed by an active suppression of the expression of transcription factors and receptors that direct the alternative fate.


Subject(s)
Cell Differentiation , Cholinergic Neurons/cytology , Homeodomain Proteins/metabolism , Receptors, Growth Factor/metabolism , Transcription Factors/metabolism , Tyrosine 3-Monooxygenase/genetics , Acetylcholine/metabolism , Adrenergic alpha-Agonists/metabolism , Animals , Cholinergic Agonists/metabolism , Cholinergic Neurons/physiology , Chromosomes, Artificial, Bacterial , Embryo, Mammalian , Female , Gene Expression Regulation, Developmental , Gene Library , Homeodomain Proteins/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Norepinephrine/metabolism , Phenotype , Proto-Oncogene Proteins c-ret/genetics , Proto-Oncogene Proteins c-ret/metabolism , Receptor, trkC/genetics , Receptor, trkC/metabolism , Receptors, Growth Factor/genetics , Sympathetic Nervous System/cytology , Sympathetic Nervous System/physiology , Transcription Factors/genetics , Tyrosine 3-Monooxygenase/metabolism
15.
EMBO J ; 31(18): 3718-29, 2012 Sep 12.
Article in English | MEDLINE | ID: mdl-22903063

ABSTRACT

The formation of functional connectivity in the nervous system is governed by axon guidance that instructs nerve growth and branching during development, implying a similarity between neuronal subtypes in terms of nerve extension. We demonstrate the molecular mechanism of another layer of complexity in vertebrates by defining a transcriptional program underlying growth differences between positionally different neurons. The rate of axon extension of the early subset of embryonic dorsal root ganglion sensory neurons is encoded in neurons at different axial levels. This code is determined by a segmental pattern of axial levels of Runx family transcription factor Runx3. Runx3 in turn determines transcription levels of genes encoding cytoskeletal proteins involved in axon extension, including Rock1 and Rock2 which have ongoing activities determining axon growth in early sensory neurons and blocking Rock activity reverses axon extension deficits of Runx3(-/-) neurons. Thus, Runx3 acts to regulate positional differences in axon extension properties apparently without affecting nerve guidance and branching, a principle that could be relevant to other parts of the nervous system.


Subject(s)
Axons/physiology , Core Binding Factor Alpha 3 Subunit/metabolism , Gene Expression Regulation , Sensory Receptor Cells/physiology , Animals , Axons/metabolism , Cell Proliferation , Chick Embryo , Ganglia, Spinal/embryology , Mice , Mice, Transgenic , Models, Genetic , Nervous System/embryology , Neurons/metabolism , RNA/metabolism , Time Factors
16.
EMBO Rep ; 15(4): 383-91, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24599748

ABSTRACT

The role of MYC proteins in somatic stem and progenitor cells during development is poorly understood. We have taken advantage of a chick in vivo model to examine their role in progenitor cells of the developing neural tube. Our results show that depletion of endogenous MYC in radial glial precursors (RGPs) is incompatible with differentiation and conversely, that overexpression of MYC induces neurogenesis independently of premature or upregulated expression of proneural gene programs. Unexpectedly, the neurogenic function of MYC depends on the integrity of the polarized neural tissue, in contrast to the situation in dissociated RGPs where MYC is mitogenic. Within the polarized RGPs of the neural tube, MYC drives differentiation by inhibiting Notch signaling and by increasing neurogenic cell division, eventually resulting in a depletion of progenitor cells. These results reveal an unexpected role of MYC in the control of stemness versus differentiation of neural stem cells in vivo.


Subject(s)
Avian Proteins/physiology , Neural Stem Cells/physiology , Oncogene Protein p55(v-myc)/physiology , Proto-Oncogene Proteins c-myc/physiology , Animals , Cell Differentiation , Cell Division , Cell Proliferation , Cells, Cultured , Chick Embryo , Chickens , Ependymoglial Cells/physiology , Gene Expression , Neural Tube/cytology , Neural Tube/embryology , Neurogenesis , Phenotype , Receptors, Notch/metabolism , Signal Transduction , Transcriptional Activation
17.
Nucleic Acids Res ; 42(11): 6972-86, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24852249

ABSTRACT

Pluripotency of embryonic stem cells (ESCs) is maintained by transcriptional activities and chromatin modifying complexes highly organized within the chromatin. Although much effort has been focused on identifying genome-binding sites, little is known on their dynamic association with chromatin across cell divisions. Here, we used a modified version of the iPOND (isolation of proteins at nascent DNA) technology to identify a large protein network enriched at nascent DNA in ESCs. This comprehensive and unbiased proteomic characterization in ESCs reveals that, in addition to the core replication machinery, proteins relevant for pluripotency of ESCs are present at DNA replication sites. In particular, we show that the chromatin remodeller HDAC1-NuRD complex is enriched at nascent DNA. Interestingly, an acute block of HDAC1 in ESCs leads to increased acetylation of histone H3 lysine 9 at nascent DNA together with a concomitant loss of methylation. Consistently, in contrast to what has been described in tumour cell lines, these chromatin marks were found to be stable during cell cycle progression of ESCs. Our results are therefore compatible with a rapid deacetylation-coupled methylation mechanism during the replication of DNA in ESCs that may participate in the preservation of pluripotency of ESCs during replication.


Subject(s)
DNA Replication , DNA-Binding Proteins/metabolism , Embryonic Stem Cells/metabolism , Epigenesis, Genetic , Animals , CCAAT-Enhancer-Binding Proteins , Cells, Cultured , DNA/metabolism , DNA Mismatch Repair , DNA-Binding Proteins/isolation & purification , Heterochromatin/metabolism , Histone Deacetylase 1/metabolism , Mi-2 Nucleosome Remodeling and Deacetylase Complex/metabolism , Mice , Nuclear Proteins/metabolism , Protein Interaction Mapping , Protein Interaction Maps , Ubiquitin-Protein Ligases
18.
Development ; 139(2): 397-410, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22186729

ABSTRACT

The cellular origin and molecular mechanisms regulating pigmentation of head and neck are largely unknown. Melanocyte specification is controlled by the transcriptional activity of Mitf, but no general logic has emerged to explain how Mitf and progenitor transcriptional activities consolidate melanocyte and progenitor cell fates. We show that cranial melanocytes arise from at least two different cellular sources: initially from nerve-associated Schwann cell precursors (SCPs) and later from a cellular source that is independent of nerves. Unlike the midbrain-hindbrain cluster from which melanoblasts arise independently of nerves, a large center of melanocytes in and around cranial nerves IX-X is derived from SCPs, as shown by genetic cell-lineage tracing and analysis of ErbB3-null mutant mice. Conditional gain- and loss-of-function experiments show genetically that cell fates in the neural crest involve both the SRY transcription factor Sox2 and Mitf, which consolidate an SCP progenitor or melanocyte fate by cross-regulatory interactions. A gradual downregulation of Sox2 in progenitors during development permits the differentiation of both neural crest- and SCP-derived progenitors into melanocytes, and an initial small pool of nerve-associated melanoblasts expands in number and disperses under the control of endothelin receptor B (Ednrb) and Wnt5a signaling.


Subject(s)
Gene Expression Regulation, Developmental/physiology , Melanocytes/cytology , Microphthalmia-Associated Transcription Factor/metabolism , Neural Crest/embryology , Pigmentation/physiology , SOXB1 Transcription Factors/metabolism , Animals , Chromatin Immunoprecipitation , Embryo, Mammalian/embryology , Imaging, Three-Dimensional , Immunohistochemistry , In Situ Hybridization , Melanocytes/metabolism , Mice , Neural Crest/metabolism , Plasmids/genetics , RNA, Small Interfering/genetics , Receptors, Endothelin/metabolism , Schwann Cells/cytology , Signal Transduction/physiology , Wnt Proteins/metabolism , Wnt-5a Protein
19.
Nat Chem Biol ; 9(2): 126-33, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23292650

ABSTRACT

Liver X receptors (Lxrα and Lxrß) are ligand-dependent nuclear receptors critical for ventral midbrain neurogenesis in vivo. However, no endogenous midbrain Lxr ligand has so far been identified. Here we used LC/MS and functional assays to identify cholic acid as a new Lxr ligand. Moreover, 24(S),25-epoxycholesterol (24,25-EC) was found to be the most potent and abundant Lxr ligand in the developing mouse midbrain. Both Lxr ligands promoted neural development in an Lxr-dependent manner in zebrafish in vivo. Notably, each ligand selectively regulated the development of distinct midbrain neuronal populations. Whereas cholic acid increased survival and neurogenesis of Brn3a-positive red nucleus neurons, 24,25-EC promoted dopaminergic neurogenesis. These results identify an entirely new class of highly selective and cell type-specific regulators of neurogenesis and neuronal survival. Moreover, 24,25-EC promoted dopaminergic differentiation of embryonic stem cells, suggesting that Lxr ligands may thus contribute to the development of cell replacement and regenerative therapies for Parkinson's disease.


Subject(s)
Mesencephalon/metabolism , Neurogenesis , Orphan Nuclear Receptors/metabolism , Animals , Brain Mapping/methods , Cell Differentiation , Cell Nucleus/metabolism , Cholesterol/analogs & derivatives , Cholesterol/metabolism , Cholic Acid/metabolism , Dopamine/metabolism , Dose-Response Relationship, Drug , Embryonic Stem Cells/cytology , Ligands , Liver X Receptors , Mice , Models, Biological , Time Factors , Transfection , Zebrafish
20.
J Neurosci ; 33(45): 17656-66, 2013 Nov 06.
Article in English | MEDLINE | ID: mdl-24198358

ABSTRACT

The principle by which unmyelinated primary sensory neurons transducing thermal, itch and pain perception are specified in early development is unknown. These classes of sensory neurons diversify from a common population of late-born neurons, which initiate expression of Runt homology domain transcription factor RUNX1 and the nerve growth factor receptor TrkA. Here, we report that signals emanating from within the mouse dorsal root ganglion mediated partly by early-born neurons destined to a myelinated phenotype participate in fating late-born RUNX1(+)/TrkA(+) neurons. Inductive factors include FGFs via activation of FGF receptor 1 (FGFR1). Consistently, FGF2 is sufficient to induce expression of RUNX1, and Fgfr1 conditional mutant mice display deficits in fating of the common population of late-born RUNX1(+)/TrkA(+) neurons that develop into unmyelinated neurons. Thus, the distinct lineages of sensory neurons are acquired in response to increasing FGF levels provided by a rising number of born neurons.


Subject(s)
Cell Lineage/physiology , Fibroblast Growth Factors/metabolism , Nerve Fibers, Unmyelinated/physiology , Sensory Receptor Cells/physiology , Animals , Chick Embryo , Core Binding Factor Alpha 2 Subunit/genetics , Core Binding Factor Alpha 2 Subunit/metabolism , Fibroblast Growth Factors/genetics , Ganglia, Spinal/metabolism , Gene Expression Regulation, Developmental , Mice , Nerve Fibers, Unmyelinated/metabolism , Receptor, trkA/genetics , Receptor, trkA/metabolism , Sensory Receptor Cells/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL