Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 115
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 174(4): 999-1014.e22, 2018 08 09.
Artículo en Inglés | MEDLINE | ID: mdl-30096314

RESUMEN

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.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Redes Reguladoras de Genes , Sistema Nervioso/metabolismo , Análisis de la Célula Individual/métodos , Transcriptoma , Animales , Femenino , Perfilación de la Expresión Génica , Secuenciación de Nucleótidos de Alto Rendimiento , Masculino , Ratones , Ratones Endogámicos C57BL , Sistema Nervioso/crecimiento & desarrollo
2.
Cell ; 157(2): 313-328, 2014 04 10.
Artículo en Inglés | MEDLINE | ID: mdl-24656405

RESUMEN

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.


Asunto(s)
Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/patología , Glioblastoma/tratamiento farmacológico , Glioblastoma/patología , Piperidinas/farmacología , Quinolinas/farmacología , Bibliotecas de Moléculas Pequeñas/farmacología , Animales , Muerte Celular/efectos de los fármacos , Xenoinjertos , Humanos , Hidroxiquinolinas/farmacología , MAP Quinasa Quinasa 4/metabolismo , Ratones , Trasplante de Neoplasias , Pinocitosis/efectos de los fármacos , Vacuolas/metabolismo , Pez Cebra
4.
Cell ; 139(2): 366-79, 2009 Oct 16.
Artículo en Inglés | MEDLINE | ID: mdl-19837037

RESUMEN

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.


Asunto(s)
Melanocitos/citología , Células de Schwann/citología , Piel/inervación , Animales , Diferenciación Celular , Movimiento Celular , Proteínas de Homeodominio , Ratones , Neuroglía , Receptor ErbB-3/metabolismo , Células Madre/citología , Factores de Transcripción/metabolismo
5.
Diabetologia ; 66(12): 2332-2345, 2023 12.
Artículo en Inglés | MEDLINE | ID: mdl-37728731

RESUMEN

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.


Asunto(s)
Diabetes Mellitus Tipo 1 , Neuropatías Diabéticas , Humanos , Diabetes Mellitus Tipo 1/complicaciones , Fibras Nerviosas/patología , Nervios Periféricos/patología , Células de Schwann/patología
6.
Development ; 146(20)2019 10 24.
Artículo en Inglés | MEDLINE | ID: mdl-31575648

RESUMEN

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.


Asunto(s)
Subunidad alfa 3 del Factor de Unión al Sitio Principal/metabolismo , Animales , Diferenciación Celular/genética , Diferenciación Celular/fisiología , Células Cultivadas , Subunidad alfa 3 del Factor de Unión al Sitio Principal/genética , Femenino , Ganglios Espinales/citología , Ganglios Espinales/metabolismo , Regulación del Desarrollo de la Expresión Génica , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Proteínas con Homeodominio LIM/genética , Proteínas con Homeodominio LIM/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Proteínas de Microfilamentos/genética , Proteínas de Microfilamentos/metabolismo , Neuronas Motoras/metabolismo , Proteínas Musculares/genética , Proteínas Musculares/metabolismo , Factores de Crecimiento Nervioso/genética , Factores de Crecimiento Nervioso/metabolismo , Células Receptoras Sensoriales/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
7.
Development ; 145(18)2018 09 25.
Artículo en Inglés | MEDLINE | ID: mdl-30126905

RESUMEN

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.


Asunto(s)
Proteínas de Homeodominio/metabolismo , Mecanorreceptores/metabolismo , MicroARNs/genética , Células Receptoras Sensoriales/citología , Animales , Proteínas de Unión al Calcio/metabolismo , Diferenciación Celular/genética , Proteínas del Ojo/metabolismo , Femenino , Humanos , Glicoproteínas de Membrana/metabolismo , Ratones , Ratones Noqueados , Embarazo , Proteínas Tirosina Quinasas/metabolismo
8.
Liver Int ; 40(4): 977-987, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32011099

RESUMEN

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.


Asunto(s)
Cresta Neural , Pericitos , Animales , Médula Ósea , Células Endoteliales , Hígado , Regeneración Hepática , Ratones , Ratones Transgénicos
9.
Nature ; 513(7519): 551-4, 2014 Sep 25.
Artículo en Inglés | MEDLINE | ID: mdl-25079316

RESUMEN

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.


Asunto(s)
Diferenciación Celular , Linaje de la Célula , Incisivo/citología , Células Madre Mesenquimatosas/citología , Neuroglía/citología , Animales , Rastreo Celular , Células Clonales/citología , Pulpa Dental/citología , Femenino , Incisivo/embriología , Masculino , Ratones , Modelos Biológicos , Cresta Neural/citología , Odontoblastos/citología , Regeneración , Células de Schwann/citología
10.
Dev Biol ; 444 Suppl 1: S308-S324, 2018 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-29807017

RESUMEN

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.


Asunto(s)
Cuerpo Carotídeo/embriología , Células Cromafines/metabolismo , Pericitos/metabolismo , Glándulas Suprarrenales/metabolismo , Glándulas Suprarrenales/fisiología , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Tipificación del Cuerpo/fisiología , Diferenciación Celular , Hipoxia de la Célula/fisiología , Embrión de Pollo , Pollos/metabolismo , Ratones , Ratones Noqueados , Proteína Proteolipídica de la Mielina/fisiología , Cresta Neural/metabolismo , Neuronas/metabolismo , Pericitos/fisiología , Factores de Transcripción/metabolismo
11.
Stem Cells ; 36(11): 1736-1751, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-29999568

RESUMEN

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.


Asunto(s)
Células Madre Adultas/metabolismo , Células-Madre Neurales/metabolismo , Proteínas Nucleares/genética , Animales , Proteínas Potenciadoras de Unión a CCAAT , Humanos , Ratones , Proteínas Nucleares/metabolismo , Ubiquitina-Proteína Ligasas
12.
Am J Hum Genet ; 96(4): 519-31, 2015 Apr 02.
Artículo en Inglés | MEDLINE | ID: mdl-25772936

RESUMEN

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.


Asunto(s)
Alopecia/genética , Disostosis Mandibulofacial/genética , Receptor de Endotelina A/genética , Alopecia/patología , Animales , Secuencia de Bases , Endotelina-1/metabolismo , Exoma/genética , Humanos , Hibridación in Situ , Disostosis Mandibulofacial/patología , Datos de Secuencia Molecular , Morfolinos/genética , Mutación Missense/genética , Linaje , ARN Mensajero/administración & dosificación , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptor de Endotelina A/metabolismo , Análisis de Secuencia de ADN , Síndrome , Tomografía Computarizada por Rayos X , Pez Cebra , Cigoma/patología
13.
Nucleic Acids Res ; 44(10): 4745-62, 2016 06 02.
Artículo en Inglés | MEDLINE | ID: mdl-26939887

RESUMEN

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.


Asunto(s)
Proteínas Cdh1/metabolismo , Replicación del ADN , Origen de Réplica , Fase S/genética , Proteínas de la Ataxia Telangiectasia Mutada/metabolismo , Línea Celular Tumoral , Células Cultivadas , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/fisiología , Daño del ADN , Activación Enzimática , Inestabilidad Genómica , Humanos , Hidroxiurea/toxicidad , Estrés Fisiológico/genética , Proteína p53 Supresora de Tumor/fisiología
14.
EMBO J ; 32(11): 1613-25, 2013 May 29.
Artículo en Inglés | MEDLINE | ID: mdl-23591430

RESUMEN

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.


Asunto(s)
Diferenciación Celular , Neuronas Colinérgicas/citología , Proteínas de Homeodominio/metabolismo , Receptores de Factores de Crecimiento/metabolismo , Factores de Transcripción/metabolismo , Tirosina 3-Monooxigenasa/genética , Acetilcolina/metabolismo , Agonistas alfa-Adrenérgicos/metabolismo , Animales , Agonistas Colinérgicos/metabolismo , Neuronas Colinérgicas/fisiología , Cromosomas Artificiales Bacterianos , Embrión de Mamíferos , Femenino , Regulación del Desarrollo de la Expresión Génica , Biblioteca de Genes , Proteínas de Homeodominio/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Norepinefrina/metabolismo , Fenotipo , Proteínas Proto-Oncogénicas c-ret/genética , Proteínas Proto-Oncogénicas c-ret/metabolismo , Receptor trkC/genética , Receptor trkC/metabolismo , Receptores de Factores de Crecimiento/genética , Sistema Nervioso Simpático/citología , Sistema Nervioso Simpático/fisiología , Factores de Transcripción/genética , Tirosina 3-Monooxigenasa/metabolismo
15.
EMBO J ; 31(18): 3718-29, 2012 Sep 12.
Artículo en Inglés | MEDLINE | ID: mdl-22903063

RESUMEN

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.


Asunto(s)
Axones/fisiología , Subunidad alfa 3 del Factor de Unión al Sitio Principal/metabolismo , Regulación de la Expresión Génica , Células Receptoras Sensoriales/fisiología , Animales , Axones/metabolismo , Proliferación Celular , Embrión de Pollo , Ganglios Espinales/embriología , Ratones , Ratones Transgénicos , Modelos Genéticos , Sistema Nervioso/embriología , Neuronas/metabolismo , ARN/metabolismo , Factores de Tiempo
16.
EMBO Rep ; 15(4): 383-91, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24599748

RESUMEN

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.


Asunto(s)
Proteínas Aviares/fisiología , Células-Madre Neurales/fisiología , Proteína Oncogénica p55(v-myc)/fisiología , Proteínas Proto-Oncogénicas c-myc/fisiología , Animales , Diferenciación Celular , División Celular , Proliferación Celular , Células Cultivadas , Embrión de Pollo , Pollos , Células Ependimogliales/fisiología , Expresión Génica , Tubo Neural/citología , Tubo Neural/embriología , Neurogénesis , Fenotipo , Receptores Notch/metabolismo , Transducción de Señal , Activación Transcripcional
17.
Nucleic Acids Res ; 42(11): 6972-86, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24852249

RESUMEN

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.


Asunto(s)
Replicación del ADN , Proteínas de Unión al ADN/metabolismo , Células Madre Embrionarias/metabolismo , Epigénesis Genética , Animales , Proteínas Potenciadoras de Unión a CCAAT , Células Cultivadas , ADN/metabolismo , Reparación de la Incompatibilidad de ADN , Proteínas de Unión al ADN/aislamiento & purificación , Heterocromatina/metabolismo , Histona Desacetilasa 1/metabolismo , Complejo Desacetilasa y Remodelación del Nucleosoma Mi-2/metabolismo , Ratones , Proteínas Nucleares/metabolismo , Mapeo de Interacción de Proteínas , Mapas de Interacción de Proteínas , Ubiquitina-Proteína Ligasas
18.
Development ; 139(2): 397-410, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22186729

RESUMEN

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.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica/fisiología , Melanocitos/citología , Factor de Transcripción Asociado a Microftalmía/metabolismo , Cresta Neural/embriología , Pigmentación/fisiología , Factores de Transcripción SOXB1/metabolismo , Animales , Inmunoprecipitación de Cromatina , Embrión de Mamíferos/embriología , Imagenología Tridimensional , Inmunohistoquímica , Hibridación in Situ , Melanocitos/metabolismo , Ratones , Cresta Neural/metabolismo , Plásmidos/genética , ARN Interferente Pequeño/genética , Receptores de Endotelina/metabolismo , Células de Schwann/citología , Transducción de Señal/fisiología , Proteínas Wnt/metabolismo , Proteína Wnt-5a
19.
Nat Chem Biol ; 9(2): 126-33, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23292650

RESUMEN

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.


Asunto(s)
Mesencéfalo/metabolismo , Neurogénesis , Receptores Nucleares Huérfanos/metabolismo , Animales , Mapeo Encefálico/métodos , Diferenciación Celular , Núcleo Celular/metabolismo , Colesterol/análogos & derivados , Colesterol/metabolismo , Ácido Cólico/metabolismo , Dopamina/metabolismo , Relación Dosis-Respuesta a Droga , Células Madre Embrionarias/citología , Ligandos , Receptores X del Hígado , Ratones , Modelos Biológicos , Factores de Tiempo , Transfección , Pez Cebra
20.
J Neurosci ; 33(45): 17656-66, 2013 Nov 06.
Artículo en Inglés | MEDLINE | ID: mdl-24198358

RESUMEN

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.


Asunto(s)
Linaje de la Célula/fisiología , Factores de Crecimiento de Fibroblastos/metabolismo , Fibras Nerviosas Amielínicas/fisiología , Células Receptoras Sensoriales/fisiología , Animales , Embrión de Pollo , Subunidad alfa 2 del Factor de Unión al Sitio Principal/genética , Subunidad alfa 2 del Factor de Unión al Sitio Principal/metabolismo , Factores de Crecimiento de Fibroblastos/genética , Ganglios Espinales/metabolismo , Regulación del Desarrollo de la Expresión Génica , Ratones , Fibras Nerviosas Amielínicas/metabolismo , Receptor trkA/genética , Receptor trkA/metabolismo , Células Receptoras Sensoriales/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA