Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 117
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
PLoS Genet ; 13(4): e1006746, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28453519

RESUMEN

Mid-hindbrain malformations can occur during embryogenesis through a disturbance of transient and localized gene expression patterns within these distinct brain structures. Rho guanine nucleotide exchange factor (ARHGEF) family members are key for controlling the spatiotemporal activation of Rho GTPase, to modulate cytoskeleton dynamics, cell division, and cell migration. We identified, by means of whole exome sequencing, a homozygous frameshift mutation in the ARHGEF2 as a cause of intellectual disability, a midbrain-hindbrain malformation, and mild microcephaly in a consanguineous pedigree of Kurdish-Turkish descent. We show that loss of ARHGEF2 perturbs progenitor cell differentiation and that this is associated with a shift of mitotic spindle plane orientation, putatively favoring more symmetric divisions. The ARHGEF2 mutation leads to reduction in the activation of the RhoA/ROCK/MLC pathway crucial for cell migration. We demonstrate that the human brain malformation is recapitulated in Arhgef2 mutant mice and identify an aberrant migration of distinct components of the precerebellar system as a pathomechanism underlying the midbrain-hindbrain phenotype. Our results highlight the crucial function of ARHGEF2 in human brain development and identify a mutation in ARHGEF2 as novel cause of a neurodevelopmental disorder.


Asunto(s)
Movimiento Celular/genética , Mutación del Sistema de Lectura/genética , Discapacidad Intelectual/genética , Factores de Intercambio de Guanina Nucleótido Rho/genética , Animales , Citoesqueleto/genética , Exoma/genética , Femenino , Secuenciación de Nucleótidos de Alto Rendimiento , Homocigoto , Humanos , Discapacidad Intelectual/diagnóstico por imagen , Discapacidad Intelectual/patología , Imagen por Resonancia Magnética , Masculino , Mesencéfalo/diagnóstico por imagen , Mesencéfalo/patología , Ratones , Linaje , Rombencéfalo/diagnóstico por imagen , Rombencéfalo/patología , Transducción de Señal , Proteína de Unión al GTP rhoA/genética
2.
Mol Cell ; 43(5): 843-50, 2011 Sep 02.
Artículo en Inglés | MEDLINE | ID: mdl-21884984

RESUMEN

Neurogenesis requires the concerted action of numerous genes that are regulated at multiple levels. However, how different layers of gene regulation are coordinated to promote neurogenesis is not well understood. We show that the neural-specific Ser/Arg repeat-related protein of 100 kDa (nSR100/SRRM4) negatively regulates REST (NRSF), a transcriptional repressor of genes required for neurogenesis. nSR100 directly promotes alternative splicing of REST transcripts to produce a REST isoform (REST4) with greatly reduced repressive activity, thereby activating expression of REST targets in neural cells. Conversely, REST directly represses nSR100 in nonneural cells to prevent the activation of neural-specific splicing events. Consistent with a critical role for nSR100 in the inhibition of REST activity, blocking nSR100 expression in the developing mouse brain impairs neurogenesis. Our results thus reveal a fundamental role for direct regulatory interactions between a splicing activator and transcription repressor in the control of the multilayered regulatory programs required for neurogenesis.


Asunto(s)
Empalme Alternativo , Neurogénesis , Factores de Transcripción/genética , Animales , Células Cultivadas , Ratones , Ratones Endogámicos , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Neuronas/citología , Neuronas/metabolismo , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Empalme del ARN , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Factores de Transcripción/metabolismo , Transcripción Genética
3.
Development ; 142(16): 2781-91, 2015 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-26209645

RESUMEN

Mammalian brain development requires coordination between neural precursor proliferation, differentiation and cellular organization to create the intricate neuronal networks of the adult brain. Here, we examined the role of the atypical cadherins Fat1 and Fat4 in this process. We show that mutation of Fat1 in mouse embryos causes defects in cranial neural tube closure, accompanied by an increase in the proliferation of cortical precursors and altered apical junctions, with perturbations in apical constriction and actin accumulation. Similarly, knockdown of Fat1 in cortical precursors by in utero electroporation leads to overproliferation of radial glial precursors. Fat1 interacts genetically with the related cadherin Fat4 to regulate these processes. Proteomic analysis reveals that Fat1 and Fat4 bind different sets of actin-regulating and junctional proteins. In vitro data suggest that Fat1 and Fat4 form cis-heterodimers, providing a mechanism for bringing together their diverse interactors. We propose a model in which Fat1 and Fat4 binding coordinates distinct pathways at apical junctions to regulate neural progenitor proliferation, neural tube closure and apical constriction.


Asunto(s)
Encéfalo/embriología , Cadherinas/metabolismo , Proliferación Celular/fisiología , Células-Madre Neurales/fisiología , Tubo Neural/embriología , Animales , Western Blotting , Cadherinas/genética , Técnicas de Silenciamiento del Gen , Ratones , Microscopía Fluorescente , Tubo Neural/metabolismo , beta-Galactosidasa
4.
Genes Dev ; 24(6): 549-60, 2010 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-20194434

RESUMEN

Mice with a complete deficiency of p73 have severe neurological and immunological defects due to the absence of all TAp73 and DeltaNp73 isoforms. As part of our ongoing program to distinguish the biological functions of these isoforms, we generated mice that are selectively deficient for the DeltaNp73 isoform. Mice lacking DeltaNp73 (DeltaNp73(-/-) mice) are viable and fertile but display signs of neurodegeneration. Cells from DeltaNp73(-/-) mice are sensitized to DNA-damaging agents and show an increase in p53-dependent apoptosis. When analyzing the DNA damage response (DDR) in DeltaNp73(-/-) cells, we discovered a completely new role for DeltaNp73 in inhibiting the molecular signal emanating from a DNA break to the DDR pathway. We found that DeltaNp73 localizes directly to the site of DNA damage, can interact with the DNA damage sensor protein 53BP1, and inhibits ATM activation and subsequent p53 phosphorylation. This novel finding may explain why human tumors with high levels of DeltaNp73 expression show enhanced resistance to chemotherapy.


Asunto(s)
Daño del ADN , Reparación del ADN/fisiología , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Transducción de Señal , Animales , Apoptosis/genética , Proteínas de la Ataxia Telangiectasia Mutada , Encéfalo/patología , Proteínas de Ciclo Celular/metabolismo , Línea Celular , Línea Celular Tumoral , Células Cultivadas , Proteínas de Unión al ADN/metabolismo , Femenino , Fertilidad/genética , Regulación de la Expresión Génica , Células HCT116 , Humanos , Longevidad/genética , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Enfermedades Neurodegenerativas/genética , Fosforilación , Isoformas de Proteínas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Proteínas Supresoras de Tumor/metabolismo
5.
J Neurosci ; 35(47): 15666-81, 2015 Nov 25.
Artículo en Inglés | MEDLINE | ID: mdl-26609159

RESUMEN

Here, we have asked about post-transcriptional mechanisms regulating murine developmental neurogenesis, focusing upon the RNA-binding proteins Smaug2 and Nanos1. We identify, in embryonic neural precursors of the murine cortex, a Smaug2 protein/nanos1 mRNA complex that is present in cytoplasmic granules with the translational repression proteins Dcp1 and 4E-T. We show that Smaug2 inhibits and Nanos1 promotes neurogenesis, with Smaug2 knockdown enhancing neurogenesis and depleting precursors, and Nanos1 knockdown inhibiting neurogenesis and maintaining precursors. Moreover, we show that Smaug2 likely regulates neurogenesis by silencing nanos1 mRNA. Specifically, Smaug2 knockdown inappropriately increases Nanos1 protein, and the Smaug2 knockdown-mediated neurogenesis is rescued by preventing this increase. Thus, Smaug2 and Nanos1 function as a bimodal translational repression switch to control neurogenesis, with Smaug2 acting in transcriptionally primed precursors to silence mRNAs important for neurogenesis, including nanos1 mRNA, and Nanos1 acting during the transition to neurons to repress the precursor state. SIGNIFICANCE STATEMENT: The mechanisms instructing neural stem cells to generate the appropriate progeny are still poorly understood. Here, we show that the RNA-binding proteins Smaug2 and Nanos1 are critical regulators of this balance and provide evidence supporting the idea that neural precursors are transcriptionally primed to generate neurons but translational regulation maintains these precursors in a stem cell state until the appropriate developmental time.


Asunto(s)
Diferenciación Celular/fisiología , Corteza Cerebral/fisiología , Células-Madre Neurales/fisiología , Neurogénesis/fisiología , Proteínas de Unión al ARN/fisiología , Proteínas Represoras/fisiología , Animales , Células Cultivadas , Corteza Cerebral/citología , Corteza Cerebral/embriología , Femenino , Masculino , Mamíferos , Ratones , Biosíntesis de Proteínas/fisiología
6.
J Neurosci ; 35(17): 6714-30, 2015 Apr 29.
Artículo en Inglés | MEDLINE | ID: mdl-25926450

RESUMEN

The transplantation of Schwann cells (SCs) holds considerable promise as a therapy for spinal cord injury, but the optimal source of these cells and the best timing for intervention remains debatable. Previously, we demonstrated that delayed transplantation of SCs generated from neonatal mouse skin-derived precursors (SKP-SCs) promoted repair and functional recovery in rats with thoracic contusions. Here, we conducted two experiments using neonatal rat cells and an incomplete cervical injury model to examine the efficacy of acute SKP-SC transplantation versus media control (Experiment 1) and versus nerve-derived SC or dermal fibroblast (Fibro) transplantation (Experiment 2). Despite limited graft survival, by 10 weeks after injury, rats that received SCs from either source showed improved functional recovery compared with media- or fibroblast-treated animals. Compared with media treatment, SKP-SC-transplanted rats showed enhanced rubrospinal tract (RST) sparing/plasticity in the gray matter (GM) rostral to injury, particularly in the absence of immunosuppression. The functional benefits of SC transplantations over fibroblast treatment correlated with the enhanced preservation of host tissue, reduced RST atrophy, and/or increased RST sparing/plasticity in the GM. In summary, our results indicate that: (1) early transplantation of neonatal SCs generated from skin or nerve promotes repair and functional recovery after incomplete cervical crush injury; (2) either of these cell types is preferable to Fibros for these purposes; and (3) age-matched SCs from these two sources do not differ in terms of their reparative effects or functional efficacy after transplantation into the injured cervical spinal cord.


Asunto(s)
Nervios Periféricos/citología , Recuperación de la Función/fisiología , Células de Schwann/trasplante , Piel/citología , Traumatismos de la Médula Espinal/cirugía , Animales , Animales Recién Nacidos , Biotina/análogos & derivados , Diferenciación Celular , Células Cultivadas , Médula Cervical , Dextranos , Modelos Animales de Enfermedad , Miembro Anterior/fisiología , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Actividad Motora/fisiología , Proteínas del Tejido Nervioso/metabolismo , Ratas , Ratas Sprague-Dawley , Ratas Transgénicas , Células de Schwann/fisiología , Células del Estroma/fisiología
7.
J Neurosci ; 34(15): 5164-75, 2014 Apr 09.
Artículo en Inglés | MEDLINE | ID: mdl-24719096

RESUMEN

The Snail transcription factor plays a key role in regulating diverse developmental processes but is not thought to play a role in mammalian neural precursors. Here, we have examined radial glial precursor cells of the embryonic murine cortex and demonstrate that Snail regulates their survival, self-renewal, and differentiation into intermediate progenitors and neurons via two distinct and separable target pathways. First, Snail promotes cell survival by antagonizing a p53-dependent death pathway because coincident p53 knockdown rescues survival deficits caused by Snail knockdown. Second, we show that the cell cycle phosphatase Cdc25b is regulated by Snail in radial precursors and that Cdc25b coexpression is sufficient to rescue the decreased radial precursor proliferation and differentiation observed upon Snail knockdown. Thus, Snail acts via p53 and Cdc25b to coordinately regulate multiple aspects of mammalian embryonic neural precursor biology.


Asunto(s)
Corteza Cerebral/embriología , Células-Madre Neurales/metabolismo , Neurogénesis , Factores de Transcripción/metabolismo , Animales , Proliferación Celular , Corteza Cerebral/citología , Corteza Cerebral/metabolismo , Células Ependimogliales/citología , Células Ependimogliales/metabolismo , Regulación del Desarrollo de la Expresión Génica , Células HEK293 , Humanos , Ratones , Células-Madre Neurales/citología , Neuronas/citología , Neuronas/metabolismo , Factores de Transcripción de la Familia Snail , Factores de Transcripción/genética , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo , Fosfatasas cdc25/genética , Fosfatasas cdc25/metabolismo
8.
Dev Biol ; 385(2): 230-41, 2014 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-24247009

RESUMEN

The mechanisms that regulate appropriate genesis and differentiation of interneurons in the developing mammalian brain are of significant interest not only because interneurons play key roles in the establishment of neural circuitry, but also because when they are deficient, this can cause epilepsy. In this regard, one genetic syndrome that is associated with deficits in neural development and epilepsy is Rubinstein-Taybi Syndrome (RTS), where the transcriptional activator and histone acetyltransferase CBP is mutated and haploinsufficient. Here, we have asked whether CBP is necessary for the appropriate genesis and differentiation of interneurons in the murine forebrain, since this could provide an explanation for the epilepsy that is associated with RTS. We show that CBP is expressed in neural precursors within the embryonic medial ganglionic eminence (MGE), an area that generates the vast majority of interneurons for the cortex. Using primary cultures of MGE precursors, we show that knockdown of CBP causes deficits in differentiation of these precursors into interneurons and oligodendrocytes, and that overexpression of CBP is by itself sufficient to enhance interneuron genesis. Moreover, we show that levels of the neurotransmitter synthesis enzyme GAD67, which is expressed in inhibitory interneurons, are decreased in the dorsal and ventral forebrain of neonatal CBP(+/-) mice, indicating that CBP plays a role in regulating interneuron development in vivo. Thus, CBP normally acts to ensure the differentiation of appropriate numbers of forebrain interneurons, and when its levels are decreased, this causes deficits in interneuron development, providing a potential explanation for the epilepsy seen in individuals with RTS.


Asunto(s)
Diferenciación Celular/fisiología , Interneuronas/citología , Prosencéfalo/citología , Factores de Transcripción p300-CBP/fisiología , Animales , Haploinsuficiencia , Ratones , Reacción en Cadena de la Polimerasa , Prosencéfalo/embriología , Factores de Transcripción p300-CBP/genética
9.
J Neurosci ; 33(1): 244-58, 2013 Jan 02.
Artículo en Inglés | MEDLINE | ID: mdl-23283338

RESUMEN

The transcription factor FoxP2 has been associated with the development of human speech but the underlying cellular function of FoxP2 is still unclear. Here we provide evidence that FoxP2 regulates genesis of some intermediate progenitors and neurons in the mammalian cortex, one of the key centers for human speech. Specifically, knockdown of FoxP2 in embryonic cortical precursors inhibits neurogenesis, at least in part by inhibiting the transition from radial glial precursors to neurogenic intermediate progenitors. Moreover, overexpression of human, but not mouse, FoxP2 enhances the genesis of intermediate progenitors and neurons. In contrast, expression of a human FoxP2 mutant that causes vocalization deficits decreases neurogenesis, suggesting that in the murine system human FoxP2 acts as a gain-of-function protein, while a human FoxP2 mutant acts as a dominant-inhibitory protein. These results support the idea that FoxP2 regulates the transition from neural precursors to transit-amplifying progenitors and ultimately neurons, and shed light upon the molecular changes that might contribute to evolution of the mammalian cortex.


Asunto(s)
Corteza Cerebral/metabolismo , Factores de Transcripción Forkhead/metabolismo , Neurogénesis/fisiología , Neuroglía/metabolismo , Neuronas/metabolismo , Proteínas Represoras/metabolismo , Animales , Corteza Cerebral/embriología , Proteínas del Ojo/genética , Proteínas del Ojo/metabolismo , Factores de Transcripción Forkhead/genética , Células HEK293 , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Humanos , Ratones , Factor de Transcripción PAX6 , Factores de Transcripción Paired Box/genética , Factores de Transcripción Paired Box/metabolismo , Proteínas Represoras/genética
10.
J Neurosci ; 33(31): 12569-85, 2013 Jul 31.
Artículo en Inglés | MEDLINE | ID: mdl-23904595

RESUMEN

The molecular mechanisms that regulate adult neural precursor cell (NPC) survival, and thus maintain adult neurogenesis, are not well defined. Here, we investigate the role of p63, a p53 family member, in adult NPC function in mice. Conditional ablation of p63 in adult NPCs or p63 haploinsufficiency led to reduced numbers of NPCs and newborn neurons in the neurogenic zones of the hippocampus and lateral ventricles and in the olfactory bulb. These reductions were attributable to enhanced apoptosis of NPCs and newborn neurons and were rescued by inhibition of caspase activity, p53, or the p53 apoptotic effector PUMA (p53-upregulated modulator of apoptosis). Moreover, these cellular deficits were functionally important because they led to perturbations in hippocampus-dependent memory formation. These results indicate that p63 regulates the numbers of adult NPCs and adult-born neurons as well as neural stem cell-dependent cognitive functions, and that it does so, at least in part, by inhibiting p53-dependent cell death.


Asunto(s)
Células Madre Adultas/fisiología , Conducta Exploratoria/fisiología , Hipocampo/fisiología , Células-Madre Neurales/fisiología , Neurogénesis/fisiología , Fosfoproteínas/metabolismo , Transactivadores/metabolismo , Animales , Bromodesoxiuridina/metabolismo , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/genética , Supervivencia Celular/fisiología , Células Cultivadas , Ventrículos Cerebrales/citología , Condicionamiento Psicológico/fisiología , Señales (Psicología) , Conducta Exploratoria/efectos de los fármacos , Miedo/psicología , Proteínas de Filamentos Intermediarios/genética , Aprendizaje por Laberinto/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteínas del Tejido Nervioso/genética , Nestina , Neurogénesis/efectos de los fármacos , Neurogénesis/genética , Fosfoproteínas/genética , Proteínas/genética , ARN no Traducido , Tamoxifeno/farmacología , Transactivadores/genética , Activación Transcripcional/efectos de los fármacos , Proteína p53 Supresora de Tumor/genética
11.
Hum Mol Genet ; 21(9): 1954-67, 2012 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-22262731

RESUMEN

Apoptosis, or programmed cell death, is a cellular pathway involved in normal cell turnover, developmental tissue remodeling, embryonic development, cellular homeostasis maintenance and chemical-induced cell death. Caspases are a family of intracellular proteases that play a key role in apoptosis. Aberrant activation of caspases has been implicated in human diseases. In particular, numerous findings implicate Caspase-6 (Casp6) in neurodegenerative diseases, including Alzheimer disease (AD) and Huntington disease (HD), highlighting the need for a deeper understanding of Casp6 biology and its role in brain development. The use of targeted caspase-deficient mice has been instrumental for studying the involvement of caspases in apoptosis. The goal of this study was to perform an in-depth neuroanatomical and behavioral characterization of constitutive Casp6-deficient (Casp6-/-) mice in order to understand the physiological function of Casp6 in brain development, structure and function. We demonstrate that Casp6-/- neurons are protected against excitotoxicity, nerve growth factor deprivation and myelin-induced axonal degeneration. Furthermore, Casp6-deficient mice show an age-dependent increase in cortical and striatal volume. In addition, these mice show a hypoactive phenotype and display learning deficits. The age-dependent behavioral and region-specific neuroanatomical changes observed in the Casp6-/- mice suggest that Casp6 deficiency has a more pronounced effect in brain regions that are involved in neurodegenerative diseases, such as the striatum in HD and the cortex in AD.


Asunto(s)
Caspasa 6/fisiología , Degeneración Nerviosa/enzimología , Envejecimiento/patología , Envejecimiento/fisiología , Enfermedad de Alzheimer/enzimología , Enfermedad de Alzheimer/patología , Animales , Apoptosis/fisiología , Secuencia de Bases , Conducta Animal/fisiología , Encéfalo/enzimología , Encéfalo/patología , Caspasa 6/deficiencia , Caspasa 6/genética , Humanos , Enfermedad de Huntington/enzimología , Enfermedad de Huntington/patología , Ratones , Ratones Noqueados , Actividad Motora/fisiología , Degeneración Nerviosa/genética , Degeneración Nerviosa/patología , Neuronas/enzimología , Neuronas/patología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores de N-Metil-D-Aspartato/fisiología
12.
Stem Cell Reports ; 19(5): 654-672, 2024 May 14.
Artículo en Inglés | MEDLINE | ID: mdl-38579710

RESUMEN

Here, we used single-cell RNA sequencing (scRNA-seq), single-cell ATAC sequencing (scATAC-seq), and single-cell spatial transcriptomics to characterize murine cortical OPCs throughout postnatal life. During development, we identified two groups of differentially localized PDGFRα+ OPCs that are transcriptionally and epigenetically distinct. One group (active, or actOPCs) is metabolically active and enriched in white matter. The second (homeostatic, or hOPCs) is less active, enriched in gray matter, and predicted to derive from actOPCs. In adulthood, these two groups are transcriptionally but not epigenetically distinct, and relative to developing OPCs are less active metabolically and have less open chromatin. When adult oligodendrogenesis is enhanced during experimentally induced remyelination, adult OPCs do not reacquire a developmental open chromatin state, and the oligodendrogenesis trajectory is distinct from that seen neonatally. These data suggest that there are two OPC groups subserving distinct postnatal functions and that neonatal and adult OPC-mediated oligodendrogenesis are fundamentally different.


Asunto(s)
Células Precursoras de Oligodendrocitos , Análisis de la Célula Individual , Animales , Células Precursoras de Oligodendrocitos/metabolismo , Células Precursoras de Oligodendrocitos/citología , Ratones , Diferenciación Celular/genética , Oligodendroglía/metabolismo , Oligodendroglía/citología , Epigénesis Genética , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/genética , Transcriptoma , Regulación del Desarrollo de la Expresión Génica , Ratones Endogámicos C57BL , Sustancia Blanca/metabolismo , Sustancia Blanca/citología
13.
Cell Rep ; 42(3): 112242, 2023 03 28.
Artículo en Inglés | MEDLINE | ID: mdl-36924490

RESUMEN

Here, we ask how developing precursors maintain the balance between cell genesis for tissue growth and establishment of adult stem cell pools, focusing on postnatal forebrain neural precursor cells (NPCs). We show that these NPCs are transcriptionally primed to differentiate and that the primed mRNAs are associated with the translational repressor 4E-T. 4E-T also broadly associates with other NPC mRNAs encoding transcriptional regulators, and these are preferentially depleted from ribosomes, consistent with repression. By contrast, a second translational regulator, Cpeb4, associates with diverse target mRNAs that are largely ribosome associated. The 4E-T-dependent mRNA association is functionally important because 4E-T knockdown or conditional knockout derepresses proneurogenic mRNA translation and perturbs maintenance versus differentiation of early postnatal NPCs in culture and in vivo. Thus, early postnatal NPCs are primed to differentiate, and 4E-T regulates the balance between cell genesis and stem cell expansion by sequestering and repressing mRNAs encoding transcriptional regulators.


Asunto(s)
Células-Madre Neurales , Diferenciación Celular/fisiología , Células-Madre Neurales/metabolismo , Neuronas/metabolismo , Cuerpos de Procesamiento , Biosíntesis de Proteínas , Proteínas Represoras/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteínas de Transporte Nucleocitoplasmático/metabolismo
14.
Arterioscler Thromb Vasc Biol ; 31(12): 2938-48, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21852558

RESUMEN

OBJECTIVE: The goal of this study was to characterize the factors and conditions required for smooth muscle cell (SMC)-directed differentiation of Sox2(+) multipotent rat and human skin-derived precursors (SKPs) and to define whether they represent a source of fully functional vascular SMCs for applications in vivo. METHODS AND RESULTS: We found that rat SKPs can differentiate almost exclusively into SMCs by reducing serum concentrations to 0.5% to 2% and plating them at low density. Human SKPs derived from foreskin required the addition of transforming growth factor-ß1 or -ß3 to differentiate into SMCs, but they did so even in the absence of serum. SMC formation was confirmed by quantitative reverse transcription-polymerase chain reaction, immunocytochemistry, and fluorescence-activated cell sorting, with increased expression of smoothelin-B and little to no expression of telokin or smooth muscle γ-actin, together indicating that SKPs differentiated into vascular rather than visceral SMCs. Rat and human SKP-derived SMCs were able to contract in vitro and also wrap around and support new capillary and larger blood vessel formation in angiogenesis assays in vivo. CONCLUSIONS: SKPs are Sox2(+) progenitors that represent an attainable autologous source of stem cells that can be easily differentiated into functional vascular SMCs in defined serum-free conditions without reprogramming. SKPs represent a clinically viable cell source for potential therapeutic applications in neovascularization.


Asunto(s)
Diferenciación Celular , Células Madre Multipotentes/citología , Músculo Liso Vascular/citología , Piel/citología , Actinas/metabolismo , Animales , Diferenciación Celular/efectos de los fármacos , Células Cultivadas , Proteínas del Citoesqueleto/metabolismo , Humanos , Masculino , Modelos Animales , Células Madre Multipotentes/efectos de los fármacos , Proteínas Musculares/metabolismo , Músculo Liso Vascular/efectos de los fármacos , Músculo Liso Vascular/metabolismo , Quinasa de Cadena Ligera de Miosina/metabolismo , Neovascularización Fisiológica/fisiología , Fragmentos de Péptidos/metabolismo , Ratas , Ratas Sprague-Dawley , Factor de Crecimiento Transformador beta1/farmacología , Factor de Crecimiento Transformador beta3/farmacología
15.
Nat Neurosci ; 11(6): 649-58, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18382462

RESUMEN

The mechanisms that regulate the pruning of mammalian axons are just now being elucidated. Here, we describe a mechanism by which, during developmental sympathetic axon competition, winning axons secrete brain-derived neurotrophic factor (BDNF) in an activity-dependent fashion, which binds to the p75 neurotrophin receptor (p75NTR) on losing axons to cause their degeneration and, ultimately, axon pruning. Specifically, we found that pruning of rat and mouse sympathetic axons that project to the eye requires both activity-dependent BDNF and p75NTR. p75NTR and BDNF are also essential for activity-dependent axon pruning in culture, where they mediate pruning by directly causing axon degeneration. p75NTR, which is enriched in losing axons, causes axonal degeneration by suppressing TrkA-mediated signaling that is essential for axonal maintenance. These data provide a mechanism that explains how active axons can eliminate less-active, competing axons during developmental pruning by directly promoting p75NTR-mediated axonal degeneration.


Asunto(s)
Axones/fisiología , Factor Neurotrófico Derivado del Encéfalo/fisiología , Degeneración Nerviosa/fisiopatología , Receptor de Factor de Crecimiento Nervioso/fisiología , Animales , Animales Recién Nacidos , Axones/efectos de los fármacos , Axotomía/métodos , Factor Neurotrófico Derivado del Encéfalo/farmacología , Células Cultivadas , Toxina del Cólera/metabolismo , Relación Dosis-Respuesta a Droga , Interacciones Farmacológicas , Inhibidores Enzimáticos/farmacología , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Regulación del Desarrollo de la Expresión Génica/fisiología , Proteínas Fluorescentes Verdes/biosíntesis , Proteínas Fluorescentes Verdes/genética , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Degeneración Nerviosa/tratamiento farmacológico , Degeneración Nerviosa/genética , Factor de Crecimiento Nervioso/farmacología , Neuronas/citología , Cloruro de Potasio/farmacología , Ratas , Ratas Sprague-Dawley , Receptor de Factor de Crecimiento Nervioso/deficiencia , Estilbamidinas/metabolismo , Ganglio Cervical Superior/citología , Ganglio Cervical Superior/crecimiento & desarrollo , Vías Visuales/crecimiento & desarrollo , Vías Visuales/metabolismo
16.
Artículo en Inglés | MEDLINE | ID: mdl-35667791

RESUMEN

Animals such as amphibians have an incredible capacity for regeneration with some being able to regrow their tail or appendages. Although some mammalian tissues like the skin and bones can repair following injury, there are only a few examples of true multilineage regeneration, including the distal portion of the digit tip. In both amphibians and mammals, however, to achieve successful repair or regeneration, it is now appreciated that intact nerve innervation is a necessity. Here, we review the current state of literature and discuss recent advances that identify axon-derived signals, Schwann cells, and nerve-derived mesenchymal cells as direct and indirect supporters of adult tissue homeostasis and repair. We posit that understanding how nerves positively influence repair and regeneration could lead to targeted regenerative medicine strategies to enhance tissue repair in humans.

17.
Cell Rep ; 41(12): 111853, 2022 12 20.
Artículo en Inglés | MEDLINE | ID: mdl-36543145

RESUMEN

Here, we ask why the nail base is essential for mammalian digit tip regeneration, focusing on the inductive nail mesenchyme. We identify a transcriptional signature for these cells that includes Lmx1b and show that the Lmx1b-expressing nail mesenchyme is essential for blastema formation. We use a combination of Lmx1bCreERT2-based lineage-tracing and single-cell transcriptional analyses to show that the nail mesenchyme contributes cells for two pro-regenerative mechanisms. One group of cells maintains their identity and regenerates the new nail mesenchyme. A second group contributes specifically to the dorsal blastema, loses their nail mesenchyme phenotype, acquires a blastema transcriptional state that is highly similar to blastema cells of other origins, and ultimately contributes to regeneration of the dorsal but not ventral dermis and bone. Thus, the regenerative necessity for an intact nail base is explained, at least in part, by a requirement for the inductive nail mesenchyme.


Asunto(s)
Células Madre Mesenquimatosas , Animales , Huesos , Células Cultivadas , Extremidades , Mamíferos
18.
Stem Cell Reports ; 17(2): 259-275, 2022 02 08.
Artículo en Inglés | MEDLINE | ID: mdl-35063124

RESUMEN

Senescent cells are responsible, in part, for tissue decline during aging. Here, we focused on CNS neural precursor cells (NPCs) to ask if this is because senescent cells in stem cell niches impair precursor-mediated tissue maintenance. We demonstrate an aging-dependent accumulation of senescent cells, largely senescent NPCs, within the hippocampal stem cell niche coincident with declining adult neurogenesis. Pharmacological ablation of senescent cells via acute systemic administration of the senolytic drug ABT-263 (Navitoclax) caused a rapid increase in NPC proliferation and neurogenesis. Genetic ablation of senescent cells similarly activated hippocampal NPCs. This acute burst of neurogenesis had long-term effects in middle-aged mice. One month post-ABT-263, adult-born hippocampal neuron numbers increased and hippocampus-dependent spatial memory was enhanced. These data support a model where senescent niche cells negatively influence neighboring non-senescent NPCs during aging, and ablation of these senescent cells partially restores neurogenesis and hippocampus-dependent cognition.


Asunto(s)
Senescencia Celular/fisiología , Células-Madre Neurales/metabolismo , Nicho de Células Madre/fisiología , Envejecimiento , Compuestos de Anilina/farmacología , Animales , Proliferación Celular/efectos de los fármacos , Senescencia Celular/efectos de los fármacos , Giro Dentado/citología , Giro Dentado/metabolismo , Femenino , Hipocampo/citología , Masculino , Ratones , Ratones Endogámicos C57BL , Células-Madre Neurales/citología , Neurogénesis/efectos de los fármacos , Memoria Espacial/efectos de los fármacos , Sulfonamidas/farmacología
19.
eNeuro ; 9(1)2022.
Artículo en Inglés | MEDLINE | ID: mdl-35027446

RESUMEN

Adult neural stem cells (NSCs) reside in two distinct niches in the mammalian brain, the ventricular-subventricular zone (V-SVZ) of the forebrain lateral ventricles and the subgranular zone (SGZ) of the hippocampal dentate gyrus. They are thought to be molecularly distinct since V-SVZ NSCs produce inhibitory olfactory bulb (OB) interneurons and SGZ NSCs excitatory dentate granule neurons. Here, we have asked whether this is so by directly comparing V-SVZ and SGZ NSCs from embryogenesis to adulthood using single-cell transcriptional data. We show that the embryonic radial glial precursor (RP) parents of these two NSC populations are very similar, but differentially express a small cohort of genes involved in glutamatergic versus GABAergic neurogenesis. These different RPs then undergo a similar gradual transition to a dormant adult NSC state over the first three postnatal weeks. This dormancy state involves transcriptional shutdown of genes that maintain an active, proliferative, prodifferentiation state and induction of genes involved in sensing and regulating their niche environment. Moreover, when reactivated to generate adult-born progeny, both populations reacquire a development-like state and re-express proneurogenic genes. Thus, V-SVZ and SGZ NSCs share a common transcriptional state throughout their lifespans and transition into and out of dormancy via similar trajectories.


Asunto(s)
Células-Madre Neurales , Adulto , Animales , Giro Dentado , Desarrollo Embrionario , Humanos , Ventrículos Laterales , Mamíferos , Neurogénesis/fisiología , Prosencéfalo
20.
Neuron ; 55(1): 1-2, 2007 Jul 05.
Artículo en Inglés | MEDLINE | ID: mdl-17610810

RESUMEN

During development, newly born neurons migrate away from their initial birth sites to their final positions in the mature brain. The neurotrophin BDNF has been shown to regulate the migration of granule cells in the cerebellum. The cellular mechanisms that mediate this chemotactic response have not been resolved. In this issue of Neuron, Zhou et al. show that vesicle trafficking is critical for allowing neurons to respond to a gradient of BDNF.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo/fisiología , Encéfalo/crecimiento & desarrollo , Movimiento Celular/fisiología , Animales , Encéfalo/embriología , Células Cultivadas , Quimiotaxis/fisiología , Humanos , Ratones , Receptor trkB/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA