Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 82
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 184(21): 5303-5305, 2021 10 14.
Artículo en Inglés | MEDLINE | ID: mdl-34653366

RESUMEN

In this issue of Cell, Wang et al. come to the unsettling conclusion that adeno-associated viruses, despite being engineered for glia-specific expression, can become widely active in endogenous neurons, misleading researchers in their quest for efficient conversion of glia into neurons for brain repair.


Asunto(s)
Neuroglía , Neuronas , Encéfalo , Dependovirus
2.
Cell ; 176(6): 1407-1419.e14, 2019 03 07.
Artículo en Inglés | MEDLINE | ID: mdl-30827680

RESUMEN

The function of somatic stem cells declines with age. Understanding the molecular underpinnings of this decline is key to counteract age-related disease. Here, we report a dramatic drop in the neural stem cells (NSCs) number in the aging murine brain. We find that this smaller stem cell reservoir is protected from full depletion by an increase in quiescence that makes old NSCs more resistant to regenerate the injured brain. Once activated, however, young and old NSCs show similar proliferation and differentiation capacity. Single-cell transcriptomics of NSCs indicate that aging changes NSCs minimally. In the aging brain, niche-derived inflammatory signals and the Wnt antagonist sFRP5 induce quiescence. Indeed, intervention to neutralize them increases activation of old NSCs during homeostasis and following injury. Our study identifies quiescence as a key feature of old NSCs imposed by the niche and uncovers ways to activate NSCs to repair the aging brain.


Asunto(s)
Encéfalo/fisiología , Factores de Edad , Animales , Encéfalo/citología , Diferenciación Celular/fisiología , División Celular/fisiología , Proliferación Celular/fisiología , Senescencia Celular/fisiología , Homeostasis , Masculino , Ratones , Ratones Endogámicos C57BL , Regeneración Nerviosa , Células-Madre Neurales/citología , Células-Madre Neurales/fisiología , Neurogénesis , Nicho de Células Madre
3.
EMBO J ; 42(11): e110384, 2023 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-37083045

RESUMEN

Most adult hippocampal neural stem cells (NSCs) remain quiescent, with only a minor portion undergoing active proliferation and neurogenesis. The molecular mechanisms that trigger the transition from quiescence to activation are still poorly understood. Here, we found the activity of the transcriptional co-activator Yap1 to be enriched in active NSCs. Genetic deletion of Yap1 led to a significant reduction in the relative proportion of active NSCs, supporting a physiological role of Yap1 in regulating the transition from quiescence to activation. Overexpression of wild-type Yap1 in adult NSCs did not induce NSC activation, suggesting tight upstream control mechanisms, but overexpression of a gain-of-function mutant (Yap1-5SA) elicited cell cycle entry in NSCs and hilar astrocytes. Consistent with a role of Yap1 in NSC activation, single cell RNA sequencing revealed a partial induction of an activated NSC gene expression program. Furthermore, Yap1-5SA expression also induced expression of Taz and other key components of the Yap/Taz regulon that were previously identified in glioblastoma stem cell-like cells. Consequently, dysregulated Yap1 activity led to repression of hippocampal neurogenesis, aberrant cell differentiation, and partial acquisition of a glioblastoma stem cell-like signature.


Asunto(s)
Glioblastoma , Células-Madre Neurales , Adulto , Humanos , Glioblastoma/metabolismo , Diferenciación Celular/fisiología , Hipocampo/metabolismo , Neurogénesis/genética , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Células-Madre Neurales/metabolismo
4.
PLoS Biol ; 21(8): e3002237, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37552690

RESUMEN

In vivo direct neuronal reprogramming relies on the implementation of an exogenous transcriptional program allowing to achieve conversion of a particular neuronal or glial cell type towards a new identity. The transcription factor (TF) Fezf2 is known for its role in neuronal subtype specification of deep-layer (DL) subcortical projection neurons. High ectopic Fezf2 expression in mice can convert both upper-layer (UL) and striatal projection neurons into a corticofugal fate, even if at low efficiency. In this study, we show that Fezf2 synergizes with the nuclear co-adaptor Lmo4 to further enhance reprogramming of UL cortical pyramidal neurons into DL corticofugal neurons, at both embryonic and early postnatal stages. Reprogrammed neurons express DL molecular markers and project toward subcerebral targets, including thalamus, cerebral peduncle (CP), and spinal cord (SC). We also show that co-expression of Fezf2 with the reprogramming factors Neurog2 and Bcl2 in early postnatal mouse glia promotes glia-to-neuron conversion with partial hallmarks of DL neurons and with Lmo4 promoting further morphological complexity. These data support a novel role for Lmo4 in synergizing with Fezf2 during direct lineage conversion in vivo.


Asunto(s)
Proteínas de Unión al ADN , Neuronas , Animales , Ratones , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Neuroglía/metabolismo , Neuronas/fisiología , Células Piramidales/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
5.
Development ; 149(4)2022 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-35195260

RESUMEN

Cellular identity is established through complex layers of genetic regulation, forged over a developmental lifetime. An expanding molecular toolbox is allowing us to manipulate these gene regulatory networks in specific cell types in vivo. In principle, if we found the right molecular tricks, we could rewrite cell identity and harness the rich repertoire of possible cellular functions and attributes. Recent work suggests that this rewriting of cell identity is not only possible, but that newly induced cells can mitigate disease phenotypes in animal models of major human diseases. So, is the sky the limit, or do we need to keep our feet on the ground? This Spotlight synthesises key concepts emerging from recent efforts to reprogramme cellular identity in vivo. We provide our perspectives on recent controversies in the field of glia-to-neuron reprogramming and identify important gaps in our understanding that present barriers to progress.


Asunto(s)
Reprogramación Celular , Animales , Linaje de la Célula , Proliferación Celular , Dependovirus/genética , Vectores Genéticos/genética , Vectores Genéticos/metabolismo , Neuroglía/citología , Neuroglía/metabolismo , Neuronas/citología , Neuronas/metabolismo , Medicina Regenerativa
6.
EMBO J ; 38(6)2019 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-30643018

RESUMEN

Adult neurogenesis is involved in cognitive performance but studies that manipulated this process to improve brain function are scarce. Here, we characterized a genetic mouse model in which neural stem cells (NSC) of the subventricular zone (SVZ) were temporarily expanded by conditional expression of the cell cycle regulators Cdk4/cyclinD1, thus increasing neurogenesis. We found that supernumerary neurons matured and integrated in the olfactory bulb similarly to physiologically generated newborn neurons displaying a correct expression of molecular markers, morphology and electrophysiological activity. Olfactory performance upon increased neurogenesis was unchanged when mice were tested on relatively easy tasks using distinct odor stimuli. In contrast, intriguingly, increasing neurogenesis improved the discrimination ability of mice when challenged with a difficult task using mixtures of highly similar odorants. Together, our study provides a mammalian model to control the expansion of somatic stem cells that can in principle be applied to any tissue for basic research and models of therapy. By applying this to NSC of the SVZ, we highlighted the importance of adult neurogenesis to specifically improve performance in a challenging olfactory task.


Asunto(s)
Aprendizaje Discriminativo , Células-Madre Neurales/fisiología , Neurogénesis/fisiología , Odorantes/análisis , Bulbo Olfatorio/fisiología , Animales , Ciclina D1/fisiología , Quinasa 4 Dependiente de la Ciclina/fisiología , Modelos Animales de Enfermedad , Masculino , Ratones , Ratones Transgénicos , Células-Madre Neurales/citología , Células-Madre Neurales/efectos de los fármacos , Neurogénesis/efectos de los fármacos , Bulbo Olfatorio/citología , Bulbo Olfatorio/efectos de los fármacos
7.
Hum Mol Genet ; 28(6): 961-971, 2019 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-30476097

RESUMEN

Spastic paraplegia gene 11(SPG11)-linked hereditary spastic paraplegia is a complex monogenic neurodegenerative disease that in addition to spastic paraplegia is characterized by childhood onset cognitive impairment, thin corpus callosum and enlarged ventricles. We have previously shown impaired proliferation of SPG11 neural progenitor cells (NPCs). For the delineation of potential defect in SPG11 brain development we employ 2D culture systems and 3D human brain organoids derived from SPG11 patients' iPSC and controls. We reveal that an increased rate of asymmetric divisions of NPCs leads to proliferation defect, causing premature neurogenesis. Correspondingly, SPG11 organoids appeared smaller than controls and had larger ventricles as well as thinner germinal wall. Premature neurogenesis and organoid size were rescued by GSK3 inhibititors including the Food and Drug Administration-approved tideglusib. These findings shed light on the neurodevelopmental mechanisms underlying disease pathology.


Asunto(s)
Corteza Cerebral/embriología , Corteza Cerebral/metabolismo , Neurogénesis/genética , Proteínas/genética , Alelos , Biomarcadores , Corteza Cerebral/fisiopatología , Trastornos del Conocimiento/genética , Trastornos del Conocimiento/fisiopatología , Susceptibilidad a Enfermedades , Técnica del Anticuerpo Fluorescente , Genotipo , Glucógeno Sintasa Quinasa 3/metabolismo , Humanos , Mutación , Organoides , Fenotipo , beta Catenina
8.
Development ; 144(24): 4604-4615, 2017 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-29061639

RESUMEN

The low-density lipoprotein receptor-related protein 4 (LRP4) is essential in muscle fibers for the establishment of the neuromuscular junction. Here, we show that LRP4 is also expressed by embryonic cortical and hippocampal neurons, and that downregulation of LRP4 in these neurons causes a reduction in density of synapses and number of primary dendrites. Accordingly, overexpression of LRP4 in cultured neurons had the opposite effect inducing more but shorter primary dendrites with an increased number of spines. Transsynaptic tracing mediated by rabies virus revealed a reduced number of neurons presynaptic to the cortical neurons in which LRP4 was knocked down. Moreover, neuron-specific knockdown of LRP4 by in utero electroporation of LRP4 miRNA in vivo also resulted in neurons with fewer primary dendrites and a lower density of spines in the developing cortex and hippocampus. Collectively, our results demonstrate an essential and novel role of neuronal LRP4 in dendritic development and synaptogenesis in the CNS.


Asunto(s)
Corteza Cerebral/metabolismo , Dendritas/metabolismo , Hipocampo/metabolismo , Receptores de LDL/metabolismo , Sinapsis/metabolismo , Animales , Células Cultivadas , Corteza Cerebral/citología , Corteza Cerebral/embriología , Técnicas de Inactivación de Genes , Hipocampo/citología , Hipocampo/embriología , Proteínas Relacionadas con Receptor de LDL , Ratones , Ratones Endogámicos C57BL , Rabia/patología , Virus de la Rabia/crecimiento & desarrollo , Receptores de LDL/genética
9.
PLoS Biol ; 15(3): e2000698, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-28350803

RESUMEN

Strategies for promoting neural regeneration are hindered by the difficulty of manipulating desired neural fates in the brain without complex genetic methods. The subventricular zone (SVZ) is the largest germinal zone of the forebrain and is responsible for the lifelong generation of interneuron subtypes and oligodendrocytes. Here, we have performed a bioinformatics analysis of the transcriptome of dorsal and lateral SVZ in early postnatal mice, including neural stem cells (NSCs) and their immediate progenies, which generate distinct neural lineages. We identified multiple signaling pathways that trigger distinct downstream transcriptional networks to regulate the diversity of neural cells originating from the SVZ. Next, we used a novel in silico genomic analysis, searchable platform-independent expression database/connectivity map (SPIED/CMAP), to generate a catalogue of small molecules that can be used to manipulate SVZ microdomain-specific lineages. Finally, we demonstrate that compounds identified in this analysis promote the generation of specific cell lineages from NSCs in vivo, during postnatal life and adulthood, as well as in regenerative contexts. This study unravels new strategies for using small bioactive molecules to direct germinal activity in the SVZ, which has therapeutic potential in neurodegenerative diseases.


Asunto(s)
Linaje de la Célula , Ventrículos Laterales/metabolismo , Transcriptoma , Animales , Simulación por Computador , Bases de Datos Genéticas , Regulación de la Expresión Génica , Redes Reguladoras de Genes , Genómica/métodos , Ratones , Regeneración Nerviosa , Células-Madre Neurales/citología , Células-Madre Neurales/metabolismo , Neurogénesis/genética , Oligodendroglía/citología , Oligodendroglía/metabolismo , Transducción de Señal , Bibliotecas de Moléculas Pequeñas
10.
EMBO J ; 34(7): 832-4, 2015 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-25691243

RESUMEN

Early development of the mammalian cerebral cortex proceeds via a sequence of proliferative and differentiative steps from neural stem cells toward neurons and glia. However, how these steps are molecularly orchestrated is still only partially understood. In this issue of The EMBO Journal, Artegiani and colleagues implicate Tox, a HMG-box transcription factor previously known only for its role in lymphocyte development, in early cortical development.


Asunto(s)
Calcineurina/metabolismo , Corteza Cerebral/embriología , Regulación del Desarrollo de la Expresión Génica/fisiología , Proteínas de Homeodominio/metabolismo , Factores de Transcripción NFATC/metabolismo , Transducción de Señal/fisiología , Animales
11.
EMBO J ; 34(16): 2162-81, 2015 Aug 13.
Artículo en Inglés | MEDLINE | ID: mdl-26157010

RESUMEN

The epithelial to mesenchymal transition (EMT) is a biological process in which cells lose cell-cell contacts and become motile. EMT is used during development, for example, in triggering neural crest migration, and in cancer metastasis. Despite progress, the dynamics of JNK signaling, its role in genomewide transcriptional reprogramming, and involved downstream effectors during EMT remain largely unknown. Here, we show that JNK is not required for initiation, but progression of phenotypic changes associated with EMT. Such dependency resulted from JNK-driven transcriptional reprogramming of critical EMT genes and involved changes in their chromatin state. Furthermore, we identified eight novel JNK-induced transcription factors that were required for proper EMT. Three of these factors were also highly expressed in invasive cancer cells where they function in gene regulation to maintain mesenchymal identity. These factors were also induced during neuronal development and function in neuronal migration in vivo. These comprehensive findings uncovered a kinetically distinct role for the JNK pathway in defining the transcriptome that underlies mesenchymal identity and revealed novel transcription factors that mediate these responses during development and disease.


Asunto(s)
Diferenciación Celular , Redes Reguladoras de Genes , MAP Quinasa Quinasa 4/metabolismo , Sistema de Señalización de MAP Quinasas , Mesodermo/fisiología , Ciclo Celular , Línea Celular , Perfilación de la Expresión Génica , Humanos , Imagen de Lapso de Tiempo , Factores de Transcripción/metabolismo
12.
Cereb Cortex ; 28(12): 4454-4471, 2018 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-30307491

RESUMEN

Neural stem cells (NSCs) in the adult mouse hippocampus occur in a specific neurogenic niche, where a multitude of extracellular signaling molecules converges to regulate NSC proliferation as well as fate and functional integration. However, the underlying mechanisms how NSCs react to extrinsic signals and convert them to intracellular responses still remains elusive. NSCs contain a functional endocannabinoid system, including the cannabinoid type-1 receptor (CB1). To decipher whether CB1 regulates adult neurogenesis directly or indirectly in vivo, we performed NSC-specific conditional inactivation of CB1 by using triple-transgenic mice. Here, we show that lack of CB1 in NSCs is sufficient to decrease proliferation of the stem cell pool, which consequently leads to a reduction in the number of newborn neurons. Furthermore, neuronal differentiation was compromised at the level of dendritic maturation pointing towards a postsynaptic role of CB1 in vivo. Deteriorated neurogenesis in NSC-specific CB1 knock-outs additionally resulted in reduced long-term potentiation in the hippocampal formation. The observed cellular and physiological alterations led to decreased short-term spatial memory and increased depression-like behavior. These results demonstrate that CB1 expressed in NSCs and their progeny controls neurogenesis in adult mice to regulate the NSC stem cell pool, dendritic morphology, activity-dependent plasticity, and behavior.


Asunto(s)
Hipocampo/fisiología , Potenciación a Largo Plazo , Células-Madre Neurales/fisiología , Neurogénesis , Receptor Cannabinoide CB1/fisiología , Animales , Conducta Animal , Hipocampo/citología , Masculino , Ratones Endogámicos C57BL , Ratones Transgénicos , Células-Madre Neurales/citología , Receptor Cannabinoide CB1/genética , Memoria Espacial/fisiología
13.
Biochim Biophys Acta ; 1859(7): 833-40, 2016 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-27080130

RESUMEN

The human genomic locus for the transcription factor TOX3 has been implicated in susceptibility to restless legs syndrome and breast cancer in genome-wide association studies, but the physiological role of TOX3 remains largely unknown. We found Tox3 to be predominantly expressed in the developing mouse brain with a peak at embryonic day E14 where it co-localizes with the neural stem and progenitor markers Nestin and Sox2 in radial glia of the ventricular zone and intermediate progenitors of the subventricular zone. Tox3 is also expressed in neural progenitor cells obtained from the ganglionic eminence of E15 mice that express Nestin, and it specifically binds the Nestin promoter in chromatin immunoprecipitation assays. In line with this, over-expression of Tox3 increased Nestin promoter activity, which was cooperatively enhanced by treatment with the stem cell self-renewal promoting Notch ligand Jagged and repressed by pharmacological inhibition of Notch signaling. Knockdown of Tox3 in the subventricular zone of E12.5 mouse embryos by in utero electroporation of Tox3 shRNA revealed a reduced Nestin expression and decreased proliferation at E14 and a reduced migration to the cortical plate in E16 embryos in electroporated cells. Together, these results argue for a role of Tox3 in the development of the nervous system.


Asunto(s)
Células-Madre Neurales/fisiología , Neurogénesis/genética , Receptores de Progesterona/fisiología , Animales , Proteínas Reguladoras de la Apoptosis , Células Cultivadas , Embrión de Mamíferos , Femenino , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Células-Madre Neurales/efectos de los fármacos , Neurogénesis/efectos de los fármacos , Neuronas/efectos de los fármacos , Neuronas/fisiología , Embarazo , ARN Interferente Pequeño/farmacología , Receptores de Progesterona/antagonistas & inhibidores , Receptores de Progesterona/genética , Transactivadores
14.
Stem Cells ; 34(12): 2861-2874, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27603577

RESUMEN

Conversion of astrocytes to neurons, via de-differentiation to neural stem cells (NSC), may be a new approach to treat neurodegenerative diseases and brain injuries. The signaling factors affecting such a cell conversion are poorly understood, and they are hard to identify in complex disease models or conventional cell cultures. To address this question, we developed a serum-free, strictly controlled culture system of pure and homogeneous "astrocytes generated from murine embryonic stem cells (ESC)." These stem cell derived astrocytes (mAGES), as well as standard primary astrocytes resumed proliferation upon addition of FGF. The signaling of FGF receptor tyrosine kinase converted GFAP-positive mAGES to nestin-positive NSC. ERK phosphorylation was necessary, but not sufficient, for cell cycle re-entry, as EGF triggered no de-differentiation. The NSC obtained by de-differentiation of mAGES were similar to those obtained directly by differentiation of ESC, as evidenced by standard phenotyping, and also by transcriptome mapping, metabolic profiling, and by differentiation to neurons or astrocytes. The de-differentiation was negatively affected by inflammatory mediators, and in particular, interferon-γ strongly impaired the formation of NSC from mAGES by a pathway involving phosphorylation of STAT1, but not the generation of nitric oxide. Thus, two antagonistic signaling pathways were identified here that affect fate conversion of astrocytes independent of genetic manipulation. The complex interplay of the respective signaling molecules that promote/inhibit astrocyte de-differentiation may explain why astrocytes do not readily form neural stem cells in most diseases. Increased knowledge of such factors may provide therapeutic opportunities to favor such conversions. Stem Cells 2016;34:2861-2874.


Asunto(s)
Astrocitos/citología , Factor 2 de Crecimiento de Fibroblastos/farmacología , Interferón gamma/farmacología , Células-Madre Neurales/citología , Neurogénesis , Animales , Astrocitos/efectos de los fármacos , Ciclo Celular/efectos de los fármacos , Desdiferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Factor de Crecimiento Epidérmico/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Ratones , Células Madre Embrionarias de Ratones/citología , Células Madre Embrionarias de Ratones/efectos de los fármacos , Células Madre Multipotentes/citología , Células Madre Multipotentes/efectos de los fármacos , Células Madre Multipotentes/metabolismo , Células-Madre Neurales/efectos de los fármacos , Neurogénesis/efectos de los fármacos , Transducción de Señal/efectos de los fármacos
15.
Stem Cells ; 34(8): 2115-29, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27068685

RESUMEN

Adult neural stem cells with the ability to generate neurons and glia cells are active throughout life in both the dentate gyrus (DG) and the subventricular zone (SVZ). Differentiation of adult neural stem cells is induced by cell fate determinants like the transcription factor Prox1. Evidence has been provided for a function of Prox1 as an inducer of neuronal differentiation within the DG. We now show that within the SVZ Prox1 induces differentiation into oligodendrocytes. Moreover, we find that loss of Prox1 expression in vivo reduces cell migration into the corpus callosum, where the few Prox1 deficient SVZ-derived remaining cells fail to differentiate into oligodendrocytes. Thus, our work uncovers a novel function of Prox1 as a fate determinant for oligodendrocytes in the adult mammalian brain. These data indicate that the neurogenic and oligodendrogliogenic lineages in the two adult neurogenic niches exhibit a distinct requirement for Prox1, being important for neurogenesis in the DG but being indispensable for oligodendrogliogenesis in the SVZ. Stem Cells 2016;34:2115-2129.


Asunto(s)
Células Madre Adultas/citología , Células Madre Adultas/metabolismo , Proteínas de Homeodominio/metabolismo , Ventrículos Laterales/citología , Células-Madre Neurales/citología , Células-Madre Neurales/metabolismo , Oligodendroglía/citología , Proteínas Supresoras de Tumor/metabolismo , Animales , Tipificación del Cuerpo/genética , Diferenciación Celular/genética , Linaje de la Célula/genética , Movimiento Celular/genética , Células Cultivadas , Elementos de Facilitación Genéticos/genética , Regulación de la Expresión Génica , Técnicas de Silenciamiento del Gen , Ratones , Neurogénesis/genética , Bulbo Olfatorio/citología , Bulbo Olfatorio/metabolismo , Factor de Transcripción 2 de los Oligodendrocitos/genética , Factor de Transcripción 2 de los Oligodendrocitos/metabolismo , Oligodendroglía/metabolismo , Regiones Promotoras Genéticas/genética , Unión Proteica , Receptores Notch/genética , Receptores Notch/metabolismo
16.
Biomacromolecules ; 18(3): 906-913, 2017 03 13.
Artículo en Inglés | MEDLINE | ID: mdl-28147484

RESUMEN

Biomaterials for cell culture allowing simple and quantitative presentation of instructive cues enable rationalization of the interplay between cells and their surrounding microenvironment. Poly(acrylamide) (PAAm) hydrogels are popular 2D-model substrates for this purpose. However, quantitative and reproducible biofunctionalization of PAAm hydrogels with multiple ligands in a trustable, controlled, and independent fashion is not trivial. Here, we describe a method for bifunctional modification of PAAm hydrogels with thiol- and amine- containing biomolecules with controlled densities in an independent, orthogonal manner. We developed copolymer networks of AAm with 9% acrylic acid and 2% N-(4-(5-(methylsulfonyl)-1,3,4-oxadiazol-2-yl)phenyl)acrylamide. The covalent binding of thiol- and amine-containing chromophores at tunable concentrations was demonstrated and quantified by UV spectroscopy. The morphology, mechanical properties, and homogeneity of the copolymerized hydrogels were characterized by scanning electron microscopy, dynamic mechanical analysis, and confocal microscopy studies. Our copolymer hydrogels were bifunctionalized with polylysine and a laminin-mimetic peptide using the specific chemistries. We analyzed the effect of binding protocol of the two components in the maturation of cultured postmitotic cortical neurons. Our substrates supported neuronal attachment, proliferation, and neuronal differentiation. We found that neurons cultured on our hydrogels bifunctionalized with ligand-specific chemistries in a sequential fashion exhibited higher maturation at comparable culture times than using a simultaneous bifunctionalization strategy, displaying a higher number of neurites, branches, and dendritic filopodia. These results demonstrate the relevance of quantitative and optimized coupling chemistries for the performance of simple biomaterials and with sensitive cell types.


Asunto(s)
Resinas Acrílicas/química , Materiales Biocompatibles/química , Hidrogeles/química , Animales , Células Cultivadas , Laminina/química , Ratones , Ratones Endogámicos C57BL , Polilisina/química , Polímeros/química
17.
Nat Rev Neurosci ; 12(2): 88-104, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21248788

RESUMEN

Astrocyte-like cells, which act as stem cells in the adult brain, reside in a few restricted stem cell niches. However, following brain injury, glia outside these niches acquire or reactivate stem cell potential as part of reactive gliosis. Recent studies have begun to uncover the molecular pathways involved in this process. A comparison of molecular pathways activated after injury with those involved in the normal neural stem cell niches highlights strategies that could overcome the inhibition of neurogenesis outside the stem cell niche and instruct parenchymal glia towards a neurogenic fate. This new view on reactive glia therefore suggests a widespread endogenous source of cells with stem cell potential, which might potentially be harnessed for local repair strategies.


Asunto(s)
Astrocitos/patología , Lesiones Encefálicas/patología , Encéfalo/patología , Gliosis/patología , Células-Madre Neurales/patología , Neurogénesis , Nicho de Células Madre/patología , Adulto , Animales , Proliferación Celular , Humanos
18.
Proc Natl Acad Sci U S A ; 110(12): E1152-61, 2013 Mar 19.
Artículo en Inglés | MEDLINE | ID: mdl-23487772

RESUMEN

Identifying the connectome of adult-generated neurons is essential for understanding how the preexisting circuitry is refined by neurogenesis. Changes in the pattern of connectivity are likely to control the differentiation process of newly generated neurons and exert an important influence on their unique capacity to contribute to information processing. Using a monosynaptic rabies virus-based tracing technique, we studied the evolving presynaptic connectivity of adult-generated neurons in the dentate gyrus (DG) of the hippocampus and olfactory bulb (OB) during the first weeks of their life. In both neurogenic zones, adult-generated neurons first receive local connections from multiple types of GABAergic interneurons before long-range projections become established, such as those originating from cortical areas. Interestingly, despite fundamental similarities in the overall pattern of evolution of presynaptic connectivity, there were notable differences with regard to the development of cortical projections: although DG granule neuron input originating from the entorhinal cortex could be traced starting only from 3 to 5 wk on, newly generated neurons in the OB received input from the anterior olfactory nucleus and piriform cortex already by the second week. This early glutamatergic input onto newly generated interneurons in the OB was matched in time by the equally early innervations of DG granule neurons by glutamatergic mossy cells. The development of connectivity revealed by our study may suggest common principles for incorporating newly generated neurons into a preexisting circuit.


Asunto(s)
Giro Dentado/fisiología , Neuronas/metabolismo , Bulbo Olfatorio/fisiología , Sinapsis/metabolismo , Animales , Giro Dentado/citología , Ratones , Ratones Transgénicos , Neuronas/citología , Bulbo Olfatorio/citología , Virus de la Rabia
19.
Development ; 138(6): 1057-68, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21343361

RESUMEN

Little is known about the intrinsic specification of adult neural stem cells (NSCs) and to what extent they depend on their local niche. To observe adult NSC division and lineage progression independent of their niche, we isolated cells from the adult mouse subependymal zone (SEZ) and cultured them at low density without growth factors. We demonstrate here that SEZ cells in this culture system are primarily neurogenic and that adult NSCs progress through stereotypic lineage trees consisting of asymmetric stem cell divisions, symmetric transit-amplifying divisions and final symmetric neurogenic divisions. Stem cells, identified by their astro/radial glial identity and their slow-dividing nature, were observed to generate asymmetrically and fast-dividing cells that maintained an astro/radial glia identity. These, in turn, gave rise to symmetrically and fast-dividing cells that lost glial hallmarks, but had not yet acquired neuronal features. The number of amplifying divisions was limited to a maximum of five in this system. Moreover, we found that cell growth correlated with the number of subsequent divisions of SEZ cells, with slow-dividing astro/radial glia exhibiting the most substantial growth prior to division. The fact that in the absence both of exogenously supplied growth factors and of signals provided by the local niche neurogenic lineage progression takes place in such stereotypic fashion, suggests that lineage progression is, to a significant degree, cell intrinsic or pre-programmed at the beginning of the lineage.


Asunto(s)
Células Madre Adultas/citología , Células Madre Adultas/fisiología , División Celular/fisiología , Linaje de la Célula/fisiología , Rastreo Celular/métodos , Células-Madre Neurales/citología , Células-Madre Neurales/fisiología , Células Madre Adultas/efectos de los fármacos , Animales , Astrocitos/citología , Astrocitos/efectos de los fármacos , Astrocitos/fisiología , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/fisiología , División Celular/efectos de los fármacos , Linaje de la Célula/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Tamaño de la Célula/efectos de los fármacos , Células Cultivadas , Péptidos y Proteínas de Señalización Intercelular/farmacología , Ratones , Sistema Nervioso/citología , Células-Madre Neurales/efectos de los fármacos , Factores de Tiempo
20.
J Neurosci ; 32(9): 3067-80, 2012 Feb 29.
Artículo en Inglés | MEDLINE | ID: mdl-22378879

RESUMEN

Neural stem cells (NSCs) generate new hippocampal dentate granule neurons throughout adulthood. The genetic programs controlling neuronal differentiation of adult NSCs are only poorly understood. Here we show that, in the adult mouse hippocampus, expression of the SoxC transcription factors Sox4 and Sox11 is initiated around the time of neuronal commitment of adult NSCs and is maintained in immature neurons. Overexpression of Sox4 and Sox11 strongly promotes in vitro neurogenesis from adult NSCs, whereas ablation of Sox4/Sox11 prevents in vitro and in vivo neurogenesis from adult NSCs. Moreover, we demonstrate that SoxC transcription factors target the promoters of genes that are induced on neuronal differentiation of adult NSCs. Finally, we show that reprogramming of astroglia into neurons is dependent on the presence of SoxC factors. These data identify SoxC proteins as essential contributors to the genetic network controlling neuronal differentiation in adult neurogenesis and neuronal reprogramming of somatic cells.


Asunto(s)
Células Madre Adultas/fisiología , Diferenciación Celular/fisiología , Hipocampo/fisiología , Neurogénesis/fisiología , Factores de Transcripción SOXC/fisiología , Animales , Células Cultivadas , Femenino , Células HEK293 , Hipocampo/citología , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neuronas/fisiología , Factores de Transcripción SOXC/biosíntesis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA