Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
J Neurosci ; 35(18): 7287-94, 2015 May 06.
Artículo en Inglés | MEDLINE | ID: mdl-25948276

RESUMEN

Pyramidal neurons in layers 2/3 and 5 of primary somatosensory cortex (S1) exhibit somewhat modest synaptic plasticity after whisker input deprivation. Whether neurons involved at earlier steps of sensory processing show more or less plasticity has not yet been examined. Here, we used longitudinal in vivo two-photon microscopy to investigate dendritic spine dynamics in apical tufts of GFP-expressing layer 4 (L4) pyramidal neurons of the vibrissal (barrel) S1 after unilateral whisker trimming. First, we characterize the molecular, anatomical, and electrophysiological properties of identified L4 neurons in Ebf2-Cre transgenic mice. Next, we show that input deprivation results in a substantial (∼50%) increase in the rate of dendritic spine loss, acutely (4-8 d) after whisker trimming. This robust synaptic plasticity in L4 suggests that primary thalamic recipient pyramidal neurons in S1 may be particularly sensitive to changes in sensory experience. Ebf2-Cre mice thus provide a useful tool for future assessment of initial steps of sensory processing in S1.


Asunto(s)
Espinas Dendríticas/fisiología , Plasticidad Neuronal/fisiología , Células Piramidales/fisiología , Privación Sensorial/fisiología , Corteza Somatosensorial/fisiología , Vibrisas/fisiología , Animales , Femenino , Masculino , Ratones , Ratones Transgénicos , Red Nerviosa/citología , Red Nerviosa/fisiología , Neuronas/fisiología , Corteza Somatosensorial/citología , Vibrisas/inervación
2.
Proc Natl Acad Sci U S A ; 108(14): 5807-12, 2011 Apr 05.
Artículo en Inglés | MEDLINE | ID: mdl-21436036

RESUMEN

Neural stem cells (NSCs) generate new granule cells throughout life in the mammalian hippocampus. Canonical Wnt signaling regulates the differentiation of NSCs towards the neuronal lineage. Here we identified the prospero-related homeodomain transcription factor Prox1 as a target of ß-catenin-TCF/LEF signaling in vitro and in vivo. Prox1 overexpression enhanced neuronal differentiation whereas shRNA-mediated knockdown of Prox1 impaired the generation of neurons in vitro and within the hippocampal niche. In contrast, Prox1 was not required for survival of adult-generated granule cells after they had matured, suggesting a role for Prox1 in initial granule cell differentiation but not in the maintenance of mature granule cells. The data presented here characterize a molecular pathway from Wnt signaling to a transcriptional target leading to granule cell differentiation within the adult brain and identify a stage-specific function for Prox1 in the process of adult neurogenesis.


Asunto(s)
Diferenciación Celular/fisiología , Hipocampo/crecimiento & desarrollo , Proteínas de Homeodominio/metabolismo , Células-Madre Neurales/metabolismo , Neurogénesis/fisiología , Transducción de Señal/fisiología , Proteínas Supresoras de Tumor/metabolismo , Proteínas Wnt/metabolismo , Animales , Secuencia de Bases , Western Blotting , Células Cultivadas , Inmunoprecipitación de Cromatina , Cartilla de ADN/genética , Hipocampo/citología , Proteínas de Homeodominio/genética , Inmunohistoquímica , Hibridación in Situ , Luciferasas , Ratones , Datos de Secuencia Molecular , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Proteínas Supresoras de Tumor/genética
3.
Brain Pathol ; 32(2): e13019, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-34515386

RESUMEN

Neurodevelopmental disorders (NDDs) are conditions that present with brain dysfunction due to alterations in the processes of brain development. They present with neuropsychiatric, cognitive, and motor symptoms. Autism spectrum disorder (ASD) and Fragile X syndrome (FXS) are two of the most common NDDs. Human brain tissue is a scarce resource that is obtained from postmortem donations. In the case of NDDs, specifically autism, the reduced donation rate of brains prevents researchers to investigate its pathology and fine anatomy. The Hispano-American Brain Bank of Neurodevelopmental Disorders (Banco Hispanoamericano de CErebros de trastornos del NEurodesarrollo) or CENE is a large-scale brain bank for neurodevelopmental disorders in Hispano-America and the US. CENE's objectives are to collect and distribute brains of patients with NDDS, with a focus on ASD and FXS, to perform research, promote education of future scientists, and enhance public awareness about the importance of human tissue availability for scientific research on brain function and disease. CENE has thus far established a bilingual system of nodes and teams in several American countries including California-US, Pennsylvania-US, México, Puerto Rico, Colombia, and Dominican Republic. CENE ensures that postmortem NDD samples used in research better match the world's genetic and ethnic diversity. CENE enables and expands NDD brain research worldwide, particularly with respect to ASD and FXS.


Asunto(s)
Trastorno del Espectro Autista , Trastorno Autístico , Trastornos del Neurodesarrollo , Trastorno del Espectro Autista/genética , Trastorno del Espectro Autista/patología , Trastorno Autístico/patología , Encéfalo/patología , Humanos , Trastornos del Neurodesarrollo/patología
4.
J Neurosci ; 30(31): 10551-62, 2010 Aug 04.
Artículo en Inglés | MEDLINE | ID: mdl-20685998

RESUMEN

Cajal-Retzius (C-R) cells play important roles in the lamination of the mammalian cortex via reelin secretion. The genetic mechanisms underlying the development of these neurons have just begun to be unraveled. Here, we show that two closely related LIM-homeobox genes Lhx1 and Lhx5 are expressed in reelin+ cells in various regions in the mouse telencephalon at or adjacent to sites where the C-R cells are generated, including the cortical hem, the mantle region of the septal/retrobulbar area, and the ventral pallium. Whereas Lhx5 is expressed in all of these reelin-expressing domains, Lhx1 is preferentially expressed in the septal area and in a continuous domain spanning from lateral olfactory region to caudomedial territories. Genetic ablation of Lhx5 results in decreased reelin+ and p73+ cells in the neocortical anlage, in the cortical hem, and in the septal, olfactory, and caudomedial telencephalic regions. The overall reduction in number of C-R cells in Lhx5 mutants is accompanied by formation of ectopic reelin+ cell clusters at the caudal telencephalon. Based on differential expression of molecular markers and by fluorescent cell tracing in cultured embryos, we located the origin of reelin+ ectopic cell clusters at the caudomedial telencephalic region. We also confirmed the existence of a normal migration stream of reelin+ cells from the caudomedial area to telencephalic olfactory territories in wild-type embryos. These results reveal a complex role for Lhx5 in regulating the development and normal distribution of C-R cells in the developing forebrain.


Asunto(s)
Moléculas de Adhesión Celular Neuronal/metabolismo , Corteza Cerebral/metabolismo , Proteínas de la Matriz Extracelular/metabolismo , Proteínas de Homeodominio/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Neuronas/metabolismo , Serina Endopeptidasas/metabolismo , Factores de Transcripción/metabolismo , Animales , Moléculas de Adhesión Celular Neuronal/genética , Movimiento Celular , Técnicas de Cultivo de Embriones , Proteínas de la Matriz Extracelular/genética , Proteínas de Homeodominio/genética , Inmunohistoquímica , Hibridación in Situ , Proteínas con Homeodominio LIM , Ratones , Ratones Noqueados , Proteínas del Tejido Nervioso/genética , Proteína Reelina , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Serina Endopeptidasas/genética , Factores de Transcripción/genética
5.
J Neurosci Res ; 89(10): 1531-41, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21688288

RESUMEN

During early vertebrate forebrain development, pioneer axons establish a symmetrical scaffold descending longitudinally through the rostral forebrain, thus forming the tract of the postoptic commissure (TPOC). In mouse embryos, this tract begins to appear at embryonic day 9.5 (E9.5) as a bundle of axons tightly constrained at a specific dorsoventral level. We have characterized the participation of the Slit chemorepellants and their Robo receptors in the control of TPOC axon projection. In E9.5-E11.5 mouse embryos, Robo1 and Robo2 are expressed in the nucleus origin of the TPOC (nTPOC), and Slit expression domains flank the TPOC trajectory. These findings suggested that these proteins are important factors in the dorsoventral positioning of the TPOC axons. Consistently with this role, Slit2 inhibited TPOC axon growth in collagen gel cultures, and interfering with Robo function in cultured embryos induced projection errors in TPOC axons. Moreover, absence of both Slit1 and Slit2 or Robo1 and Robo2 in mutant mouse embryos revealed aberrant TPOC trajectories, resulting in abnormal spreading of the tract and misprojections into both ventral and dorsal tissues. These results reveal that Slit-Robo signaling regulates the dorsoventral position of this pioneer tract in the developing forebrain.


Asunto(s)
Axones/fisiología , Péptidos y Proteínas de Señalización Intercelular/fisiología , Proteínas del Tejido Nervioso/fisiología , Neurogénesis/fisiología , Prosencéfalo/embriología , Receptores Inmunológicos/fisiología , Transducción de Señal/fisiología , Animales , Axones/metabolismo , Núcleo Celular/genética , Núcleo Celular/fisiología , Regulación del Desarrollo de la Expresión Génica/fisiología , Ratones , Ratones Endogámicos , Ratones Noqueados , Proteínas del Tejido Nervioso/biosíntesis , Proteínas del Tejido Nervioso/genética , Vías Nerviosas/anomalías , Vías Nerviosas/embriología , Vías Nerviosas/fisiología , Prosencéfalo/anomalías , Estructura Terciaria de Proteína/genética , Estructura Terciaria de Proteína/fisiología , Receptores Inmunológicos/biosíntesis , Receptores Inmunológicos/genética , Proteínas Roundabout
6.
Cell Rep ; 30(6): 1964-1981.e3, 2020 02 11.
Artículo en Inglés | MEDLINE | ID: mdl-32049024

RESUMEN

The laminar architecture of the mammalian neocortex depends on the orderly generation of distinct neuronal subtypes by apical radial glia (aRG) during embryogenesis. Here, we identify critical roles for the autism risk gene Foxp1 in maintaining aRG identity and gating the temporal competency for deep-layer neurogenesis. Early in development, aRG express high levels of Foxp1 mRNA and protein, which promote self-renewing cell divisions and deep-layer neuron production. Foxp1 levels subsequently decline during the transition to superficial-layer neurogenesis. Sustained Foxp1 expression impedes this transition, preserving a population of cells with aRG identity throughout development and extending the early neurogenic period into postnatal life. FOXP1 expression is further associated with the initial formation and expansion of basal RG (bRG) during human corticogenesis and can promote the formation of cells exhibiting characteristics of bRG when misexpressed in the mouse cortex. Together, these findings reveal broad functions for Foxp1 in cortical neurogenesis.


Asunto(s)
Factores de Transcripción Forkhead/metabolismo , Células-Madre Neurales/metabolismo , Proteínas Represoras/metabolismo , Animales , Diferenciación Celular/fisiología , Autorrenovación de las Células/fisiología , Humanos , Ratones , Células-Madre Neurales/citología
7.
Front Neuroanat ; 12: 96, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30483071

RESUMEN

Stereotypic cell migrations in the developing brain are fundamental for the proper patterning of brain regions and formation of neural networks. In this work, we uncovered in the developing rat, a population of neurons expressing tyrosine hydroxylase (TH) that migrates posteriorly from the alar plate of the midbrain, in neurophilic interaction with axons of the mesencephalic nucleus of the trigeminal nerve. A fraction of this population was also shown to traverse the mid-hindbrain boundary, reaching the vicinity of the locus coeruleus (LC) in rhombomere 1 (r1). This migratory population, however, does not have a noradrenergic (NA) phenotype and, in keeping with its midbrain origin, expresses Otx2 which is down regulated upon migration into the hindbrain. The interaction with the trigeminal mesencephalic axons is necessary for the arrangement and distribution of migratory cells as these aspects are dramatically altered in whole embryo cultures upon disruption of trigeminal axon projection by interfering with DCC function. Moreover, in mouse embryos in an equivalent developmental stage, we detected a cell population that also migrates caudally within the midbrain apposed to mesencephalic trigeminal axons but that does not express TH; a fraction of this population expresses calbindin instead. Overall, our work identified TH-expressing neurons from the rat midbrain alar plate that migrate tangentially over long distances within the midbrain and into the hindbrain by means of a close interaction with trigeminal mesencephalic axons. A different migratory population in this region and also in mouse embryos revealed diversity among the cells that follow this descending migratory pathway.

8.
Front Neuroanat ; 11: 97, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29163070

RESUMEN

Early telencephalic development involves the migration of diverse cell types that can be identified by specific molecular markers. Most prominent among them are Cajal-Retzius (CR) cells that emanate mainly from the cortical hem and to a lesser extent from rostrolateral, septal and caudo-medial regions. One additional territory proposed to give rise to CR cells that migrate dorsally into the neocortex lies at the ventral pallium, although contradictory results question this notion. With the use of a cell-permeable fluorescent tracer in cultured embryos, we identified novel migratory paths of putative CR cells and other populations that originate from the rostrolateral telencephalon at its olfactory region. Moreover, extensive labeling on the lateral telencephalon along its rostro-caudal extent failed to reveal a dorsally-migrating CR cell population from the ventral pallium at the stages analyzed. Hence, this work reveals a novel olfactory CR cell migration and supports the idea that the ventral pallium, where diverse types of neurons converge, does not actually generate CR cells.

9.
Methods Mol Biol ; 1251: 25-42, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25391792

RESUMEN

Two-photon excitation (2PE) overcomes many challenges in fluorescence microscopy. Compared to confocal microscopy, 2PE microscopy improves depth penetration, owing to the longer excitation wavelength required and to the ability to collect scattered emission photons as a useful signal. It also minimizes photodamage because lower energy photons are used and because fluorescence is confined to the geometrical focus of the laser spot. 2PE is therefore ideal for high-resolution, deep-tissue, time-lapse imaging of dynamic processes in cell biology. Here, we provide examples of important applications of 2PE for in vivo imaging of neuronal structure and signals; we also describe how it can be combined with optogenetics or photolysis of caged molecules to simultaneously probe and control neuronal activity.


Asunto(s)
Técnicas Citológicas/métodos , Microscopía Fluorescente/instrumentación , Microscopía Fluorescente/métodos , Fotones , Imagen de Lapso de Tiempo/métodos , Procesamiento de Imagen Asistido por Computador/métodos , Neuronas/ultraestructura , Optogenética/métodos , Fotólisis
10.
Front Neuroanat ; 9: 113, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26321924

RESUMEN

Acquisition of specific neuronal identity by individual brain nuclei is a key step in brain development. However, how the mechanisms that confer neuronal identity are integrated with upstream regional specification networks is still mysterious. Expression of Sonic hedgehog (Shh), is required for hypothalamic specification and is later downregulated by Tbx3 to allow for the differentiation of the tubero-mamillary region. In this region, the mamillary body (MBO), is a large neuronal aggregate essential for memory formation. To clarify how MBO identity is acquired after regional specification, we investigated Lhx5, a transcription factor with restricted MBO expression. We first generated a hypomorph allele of Lhx5-in homozygotes, the MBO disappears after initial specification. Intriguingly, in these mutants, Tbx3 was downregulated and the Shh expression domain abnormally extended. Microarray analysis and chromatin immunoprecipitation indicated that Lhx5 appears to be involved in Shh downregulation through Tbx3 and activates several MBO-specific regulator and effector genes. Finally, by tracing the caudal hypothalamic cell lineage we show that, in the Lhx5 mutant, at least some MBO cells are present but lack characteristic marker expression. Our work shows how the Lhx5 locus contributes to integrate regional specification pathways with downstream acquisition of neuronal identity in the MBO.

11.
Front Neuroanat ; 9: 136, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26578897

RESUMEN

The mamillary body (MM) is a group of hypothalamic nuclei related to memory and spatial navigation that interconnects hippocampal, thalamic, and tegmental regions. Here we demonstrate that Lhx5, a LIM-HD domain transcription factor expressed early in the developing posterior hypothalamus, is required for the generation of the MM and its derived tracts. The MM markers Foxb1, Sim2, and Lhx1 are absent in Lhx5 knock-out mice from early embryonic stages, suggesting abnormal specification of this region. This was supported by the absence of Nkx2.1 and expansion of Shh in the prospective mamillary area. Interestingly, we also found an ectopic domain expressing Lhx2 and Lhx9 along the anterio-posterior hypothalamic axis. Our results suggest that Lhx5 controls early aspects of hypothalamic development by regulating gene expression and cellular specification in the prospective MM.

12.
Neuron ; 74(2): 314-30, 2012 Apr 26.
Artículo en Inglés | MEDLINE | ID: mdl-22542185

RESUMEN

Neuroepithelial attachments at adherens junctions are essential for the self-renewal of neural stem and progenitor cells and the polarized organization of the developing central nervous system. The balance between stem cell maintenance and differentiation depends on the precise assembly and disassembly of these adhesive contacts, but the gene regulatory mechanisms orchestrating this process are not known. Here, we demonstrate that two Forkhead transcription factors, Foxp2 and Foxp4, are progressively expressed upon neural differentiation in the spinal cord. Elevated expression of either Foxp represses the expression of a key component of adherens junctions, N-cadherin, and promotes the detachment of differentiating neurons from the neuroepithelium. Conversely, inactivation of Foxp2 and Foxp4 function in both chick and mouse results in a spectrum of neural tube defects associated with neuroepithelial disorganization and enhanced progenitor maintenance. Together, these data reveal a Foxp-based transcriptional mechanism that regulates the integrity and cytoarchitecture of neuroepithelial progenitors.


Asunto(s)
Tipificación del Cuerpo/genética , Cadherinas/metabolismo , Sistema Nervioso Central/citología , Factores de Transcripción Forkhead/metabolismo , Células Neuroepiteliales/fisiología , Células Madre/fisiología , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Adhesión Celular/genética , Diferenciación Celular/genética , Sistema Nervioso Central/enzimología , Embrión de Pollo , Electroporación , Embrión de Mamíferos , Citometría de Flujo , Factores de Transcripción Forkhead/genética , Regulación del Desarrollo de la Expresión Génica/genética , Proteínas Fluorescentes Verdes/genética , Ratones , Ratones Transgénicos , Modelos Biológicos , Mutación/genética , Proteínas del Tejido Nervioso/genética , Factor de Transcripción 2 de los Oligodendrocitos , Fosfopiruvato Hidratasa/metabolismo , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Factores de Transcripción SOXB1/metabolismo
13.
Mol Cell Endocrinol ; 333(2): 127-33, 2011 Feb 20.
Artículo en Inglés | MEDLINE | ID: mdl-21182892

RESUMEN

Embryonic neurogenesis is controlled by the activation of specific genetic programs. In the hypothalamus, neuronal thyrotropin-releasing hormone (TRH) populations control important physiological process, including energy homeostasis and autonomic function; however, the genetic program leading to the TRH expression is poorly understood. Here, we show that the Klf4 gene, encoding the transcription factor Krüppel-like factor 4 (Klf4), was expressed in the rat hypothalamus during development and regulated Trh expression. In rat fetal hypothalamic cells Klf4 regulated Trh promoter activity through CACCC and GC motifs present on the Trh gene promoter. Accordingly, hypothalamic Trh expression was down-regulated at embryonic day 15 in the Klf4(-/-) mice resulting in diminished bioactive peptide levels. Although at the neonatal stage the Trh transcript levels of the Klf4(-/-) mice were normal, the reduction in peptide levels persisted. Thus, our data indicate that Klf4 plays a key role in the maturation of TRH expression in hypothalamic neurons.


Asunto(s)
Hipotálamo/embriología , Hipotálamo/metabolismo , Factores de Transcripción de Tipo Kruppel/metabolismo , Hormona Liberadora de Tirotropina/biosíntesis , Animales , Secuencia de Bases , Regulación del Desarrollo de la Expresión Génica , Factor 4 Similar a Kruppel , Ratones , Ratones Endogámicos C57BL , Datos de Secuencia Molecular , Neuronas/metabolismo , Regiones Promotoras Genéticas/genética , Unión Proteica , Ratas , Factor de Transcripción Sp1/metabolismo , Hormona Liberadora de Tirotropina/genética , Transcripción Genética
14.
Nat Neurosci ; 12(11): 1373-80, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19838179

RESUMEN

The fate of cortical progenitors, which progressively generate neurons and glial cells during development, is determined by temporally and spatially regulated signaling mechanisms. We found that the transcription factor Sip1 (Zfhx1b), which is produced at high levels in postmitotic neocortical neurons, regulates progenitor fate non-cell autonomously. Conditional deletion of Sip1 in young neurons induced premature production of upper-layer neurons at the expense of deep layers, precocious and increased generation of glial precursors, and enhanced postnatal astrocytogenesis. The premature upper-layer generation coincided with overexpression of the neurotrophin-3 (Ntf3) gene and upregulation of fibroblast growth factor 9 (Fgf9) gene expression preceded precocious gliogenesis. Exogenous application of Fgf9 to mouse cortical slices induced excessive generation of glial precursors in the germinal zone. Our data suggest that Sip1 restrains the production of signaling factors in postmitotic neurons that feed back to progenitors to regulate the timing of cell fate switch and the number of neurons and glial cells throughout corticogenesis.


Asunto(s)
Diferenciación Celular/fisiología , Retroalimentación Fisiológica/fisiología , Neocórtex/citología , Proteínas del Tejido Nervioso/fisiología , Neuronas/fisiología , Transducción de Señal/fisiología , Células Madre/fisiología , Animales , Bromodesoxiuridina/metabolismo , Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Embrión de Mamíferos , Retroalimentación Fisiológica/efectos de los fármacos , Factor 9 de Crecimiento de Fibroblastos/genética , Factor 9 de Crecimiento de Fibroblastos/metabolismo , Factor 9 de Crecimiento de Fibroblastos/farmacología , Regulación del Desarrollo de la Expresión Génica/fisiología , Técnicas In Vitro , Ratones , Ratones Noqueados , Neocórtex/embriología , Proteínas del Tejido Nervioso/deficiencia , Neurogénesis/fisiología , Neuroglía/fisiología , Neurotrofina 3/genética , Neurotrofina 3/metabolismo , ARN Mensajero/metabolismo , Células Madre/efectos de los fármacos , Factores de Tiempo
15.
Proc Natl Acad Sci U S A ; 104(31): 12919-24, 2007 Jul 31.
Artículo en Inglés | MEDLINE | ID: mdl-17644613

RESUMEN

Smad-interacting protein-1 (Sip1) [Zinc finger homeobox (Zfhx1b)] is a transcription factor implicated in the genesis of Mowat-Wilson syndrome in humans. Sip1 expression in the dorsal telencephalon of mouse embryos was documented from E12.5. We inactivated the gene specifically in cortical precursors. This resulted in the lack of the entire hippocampal formation. Sip1 mutant mice exhibited death of differentiating cells and decreased proliferation in the region of the prospective hippocampus and dentate gyrus. The expression of the Wnt antagonist Sfrp1 was ectopically activated, whereas the activity of the noncanonical Wnt effector, JNK, was down-regulated in the embryonic hippocampus of mutant mice. In cortical cells, Sip1 protein was detected on the promoter of Sfrp1 gene and both genes showed a mutually exclusive pattern of expression suggesting that Sfrp1 expression is negatively regulated by Sip1. Sip1 is therefore essential to the development of the hippocampus and dentate gyrus, and is able to modulate Wnt signaling in these regions.


Asunto(s)
Hipocampo/metabolismo , Proteínas de Homeodominio/metabolismo , Proteínas Represoras/metabolismo , Transducción de Señal , Proteínas Wnt/metabolismo , Animales , Apoptosis , Proliferación Celular , Eliminación de Gen , Regulación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica , Hipocampo/citología , Hipocampo/embriología , Hipocampo/crecimiento & desarrollo , Proteínas de Homeodominio/genética , Péptidos y Proteínas de Señalización Intercelular/genética , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Mutación/genética , Regiones Promotoras Genéticas , Proteínas Represoras/genética , Células Madre/citología , Células Madre/metabolismo , Telencéfalo/metabolismo , Regulación hacia Arriba , Caja Homeótica 2 de Unión a E-Box con Dedos de Zinc
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA