Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Nat Commun ; 13(1): 3340, 2022 06 09.
Artículo en Inglés | MEDLINE | ID: mdl-35680927

RESUMEN

Advances in 3D neuronal cultures, such as brain spheroids and organoids, are allowing unprecedented in vitro access to some of the molecular, cellular and developmental mechanisms underlying brain diseases. However, their efficacy in recapitulating brain network properties that encode brain function remains limited, thereby precluding development of effective in vitro models of complex brain disorders like schizophrenia. Here, we develop and characterize a Modular Neuronal Network (MoNNet) approach that recapitulates specific features of neuronal ensemble dynamics, segregated local-global network activities and a hierarchical modular organization. We utilized MoNNets for quantitative in vitro modelling of schizophrenia-related network dysfunctions caused by highly penetrant mutations in SETD1A and 22q11.2 risk loci. Furthermore, we demonstrate its utility for drug discovery by performing pharmacological rescue of alterations in neuronal ensembles stability and global network synchrony. MoNNets allow in vitro modelling of brain diseases for investigating the underlying neuronal network mechanisms and systematic drug discovery.


Asunto(s)
Encefalopatías , Esquizofrenia , Encéfalo , N-Metiltransferasa de Histona-Lisina , Humanos , Neuronas/fisiología , Organoides , Esquizofrenia/genética
2.
Nat Electron ; 2(8): 343-350, 2019 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-31850397

RESUMEN

Intracellular electrophysiology is a foundational method in neuroscience and uses electrolyte-filled glass electrodes and benchtop amplifiers to measure and control transmembrane voltages and currents. Commercial amplifiers perform such recordings with high signal-to-noise ratios (SNRs) but are often expensive, bulky, and not easily scalable to many channels due to reliance on board-level integration of discrete components. Here, we present a monolithic complementary-metal-oxide-semiconductor (CMOS) multi-clamp amplifier integrated circuit capable of recording both voltages and currents with performance exceeding that of commercial benchtop instrumentation. Miniaturization enables high-bandwidth current mirroring, facilitating the synthesis of large-valued active resistors with lower noise than their passive equivalents. This enables the realization of compensation modules that can account for a wide range of electrode impedances. We validate the amplifier's operation electrically, in primary neuronal cultures, and in acute slices, using both high-impedance sharp and patch electrodes. This work provides a solution for low-cost, high-performance and scalable multi-clamp amplifiers.

3.
J Cell Biol ; 216(6): 1543-1556, 2017 06 05.
Artículo en Inglés | MEDLINE | ID: mdl-28512143

RESUMEN

Efficient collective migration depends on a balance between contractility and cytoskeletal rearrangements, adhesion, and mechanical cell-cell communication, all controlled by GTPases of the RHO family. By comprehensive screening of guanine nucleotide exchange factors (GEFs) in human bronchial epithelial cell monolayers, we identified GEFs that are required for collective migration at large, such as SOS1 and ß-PIX, and RHOA GEFs that are implicated in intercellular communication. Down-regulation of the latter GEFs differentially enhanced front-to-back propagation of guidance cues through the monolayer and was mirrored by down-regulation of RHOA expression and myosin II activity. Phenotype-based clustering of knockdown behaviors identified RHOA-ARHGEF18 and ARHGEF3-ARHGEF28-ARHGEF11 clusters, indicating that the latter may signal through other RHO-family GTPases. Indeed, knockdown of RHOC produced an intermediate between the two phenotypes. We conclude that for effective collective migration, the RHOA-GEFs → RHOA/C → actomyosin pathways must be optimally tuned to compromise between generation of motility forces and restriction of intercellular communication.


Asunto(s)
Bronquios/enzimología , Movimiento Celular , Células Epiteliales/enzimología , Factores de Intercambio de Guanina Nucleótido/metabolismo , Transducción de Señal , Proteínas de Unión al GTP rho/metabolismo , Proteína de Unión al GTP rhoA/metabolismo , Actomiosina/metabolismo , Bronquios/citología , Línea Celular , Factores de Intercambio de Guanina Nucleótido/genética , Humanos , Fenotipo , Interferencia de ARN , Factores de Intercambio de Guanina Nucleótido Rho/genética , Factores de Intercambio de Guanina Nucleótido Rho/metabolismo , Proteína SOS1/genética , Proteína SOS1/metabolismo , Factores de Tiempo , Transfección , Cicatrización de Heridas , Proteínas de Unión al GTP rho/genética , Proteína de Unión al GTP rhoA/genética , Proteína rhoC de Unión a GTP
4.
Development ; 143(12): 2194-205, 2016 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-27122165

RESUMEN

Delamination of neural crest (NC) cells is a bona fide physiological model of epithelial-to-mesenchymal transition (EMT), a process that is influenced by Wnt/ß-catenin signalling. Using two in vivo models, we show that Wnt/ß-catenin signalling is transiently inhibited at the time of NC delamination. In attempting to define the mechanism underlying this inhibition, we found that the scaffold proteins Dact1 and Dact2, which are expressed in pre-migratory NC cells, are required for NC delamination in Xenopus and chick embryos, whereas they do not affect the motile properties of migratory NC cells. Dact1/2 inhibit Wnt/ß-catenin signalling upstream of the transcriptional activity of T cell factor (TCF), which is required for EMT to proceed. Dact1/2 regulate the subcellular distribution of ß-catenin, preventing ß-catenin from acting as a transcriptional co-activator to TCF, yet without affecting its stability. Together, these data identify a novel yet important regulatory element that inhibits ß-catenin signalling, which then affects NC delamination.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Cresta Neural/citología , Cresta Neural/metabolismo , Proteínas Wnt/metabolismo , Animales , Movimiento Celular , Núcleo Celular/metabolismo , Embrión de Pollo , Femenino , Células HEK293 , Humanos , Fracciones Subcelulares/metabolismo , Vía de Señalización Wnt , Xenopus laevis/embriología , Xenopus laevis/metabolismo , beta Catenina/metabolismo
5.
Genes Dev ; 28(24): 2764-77, 2014 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-25512563

RESUMEN

Collective epithelial migration is important throughout embryonic development. The underlying mechanisms are poorly understood but likely involve spatially localized activation of Rho GTPases. We previously reported that Rac1 is essential for generating the protrusive activity that drives the collective migration of anterior visceral endoderm (AVE) cells in the early mouse embryo. To identify potential regulators of Rac1, we first performed an RNAi screen of Rho family exchange factors (guanine nucleotide exchange factor [GEF]) in an in vitro collective epithelial migration assay and identified ß-Pix. Genetic deletion of ß-Pix in mice disrupts collective AVE migration, while high-resolution live imaging revealed that this is associated with randomly directed protrusive activity. We conclude that ß-Pix controls the spatial localization of Rac1 activity to drive collective AVE migration at a critical stage in mouse development.


Asunto(s)
Endodermo/citología , Factores de Intercambio de Guanina Nucleótido Rho/metabolismo , Animales , Movimiento Celular/genética , Embrión de Mamíferos , Células Epiteliales/citología , Eliminación de Gen , Ratones , Ratones Noqueados , Neuropéptidos/genética , Neuropéptidos/metabolismo , Transporte de Proteínas/genética , Factores de Intercambio de Guanina Nucleótido Rho/genética , Vísceras/citología , Proteína de Unión al GTP rac1/genética , Proteína de Unión al GTP rac1/metabolismo
6.
Cell Rep ; 4(3): 492-503, 2013 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-23891002

RESUMEN

The different modes of stem cell division are tightly regulated to balance growth and differentiation during organ development and homeostasis, and these regulatory processes are subverted in tumor formation. Here, we developed markers that provided the single-cell resolution necessary to quantify the three modes of division taking place in the developing nervous system in vivo: self-expanding, PP; self-replacing, PN; and self-consuming, NN. Using these markers and a mathematical model that predicts the dynamics of motor neuron progenitor division, we identify a role for the morphogen Sonic hedgehog in the maintenance of stem cell identity in the developing spinal cord. Moreover, our study provides insight into the process linking lineage commitment to neurogenesis with changes in cell-cycle parameters. As a result, we propose a challenging model in which the external Sonic hedgehog signal dictates stem cell identity, reflected in the consequent readjustment of cell-cycle parameters.


Asunto(s)
Proteínas Hedgehog/metabolismo , Neuronas Motoras/citología , Neuronas Motoras/metabolismo , Células-Madre Neurales/citología , Células-Madre Neurales/metabolismo , Animales , Ciclo Celular/fisiología , Diferenciación Celular/fisiología , Procesos de Crecimiento Celular/fisiología , Embrión de Pollo , Pollos , Proteínas Hedgehog/genética , Modelos Neurológicos , Neurogénesis , Transducción de Señal
7.
Dev Neurobiol ; 73(11): 815-27, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23776185

RESUMEN

Neuroblastoma, the most common extracranial tumor in children, is caused by genetic lesions in neural crest precursors of the peripheral nervous system. However, since neural crest cells are neither present after birth and nor are they readily accessible for analysis, very little is known about the genetic networks they might share with neuroblastoma cells during their development, despite their common embryonic origin. Here we have developed a novel resource for lineage tracing and for the isolation of neural crest cells in the chick embryo, enabling us to perform a genome-wide expression screen in neural crest progenitors. In this analysis, we efficiently retrieved known neural crest specific genes that validate our screening strategy and we identified new genes that participate in diverse cell activities, yet with a strong representation of genes associated to cell signaling and cell mobility, two hallmarks of migratory cells. We crossed this transcriptome data with that in the neuroblastoma gene server to search for the human orthologues of these genes associated with neuroblastoma. Accordingly, we retrieved 54 genes expressed strongly in both populations, from which we were able to validate a total of 27 genes expressed in the neural crest that are relevant to neuroblastoma formation. We propose that neural crest and neuroblastoma tumor cells share a common genetic signature that might serve to characterize neuroblastoma cancer stem cells, thereby contributing to the identification of specific targets against which new therapeutic strategies can be designed.


Asunto(s)
Cresta Neural/citología , Neuroblastoma/genética , Transcriptoma , Animales , Embrión de Pollo , Citometría de Flujo , Humanos , Inmunohistoquímica , Hibridación in Situ , Análisis de Secuencia por Matrices de Oligonucleótidos
8.
J Neurosci ; 33(7): 2773-83, 2013 Feb 13.
Artículo en Inglés | MEDLINE | ID: mdl-23407937

RESUMEN

Neuroblastoma is an embryonic tumor derived from cells of the neural crest. Taking advantage of a newly developed neural crest lineage tracer and based on the hypothesis that the molecular mechanisms that mediate neural crest delamination are also likely to be involved in the spread of neuroblastoma, we were able to identify genes that are active both in neural crest development and neuroblastoma tumor formation. A subsequent search of the neuroblastoma gene server for human orthologues of genes differentially expressed in the chick embryo neural crest screen retrieved the LIM domain only protein 4 (LMO4), which was expressed in both cell types analyzed. Functional experiments in these two model systems revealed that LMO4 activity is required for neuroblastoma cell invasion and neural crest delamination. Moreover, we identified LMO4 as an essential cofactor in Snail2-mediated cadherin repression and in the epithelial-to-mesenchymal transition of both neural crest and neuroblastoma cells. Together, our results suggest that the association of high levels of LMO4 with aggressive neuroblastomas is dependent on LMO4 regulation of cadherin expression and hence, tumor invasiveness.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/fisiología , Neoplasias Encefálicas/patología , Transición Epitelial-Mesenquimal/genética , Transición Epitelial-Mesenquimal/fisiología , Proteínas con Dominio LIM/genética , Proteínas con Dominio LIM/fisiología , Cresta Neural/patología , Neuroblastoma/patología , Factores de Transcripción/genética , Factores de Transcripción/fisiología , Animales , Western Blotting , Cadherinas/biosíntesis , Cadherinas/fisiología , Línea Celular Tumoral , Embrión de Pollo , ADN/genética , Electroforesis en Gel de Poliacrilamida , Citometría de Flujo , Vectores Genéticos , Humanos , Inmunohistoquímica , Hibridación in Situ , Lentivirus/genética , Luciferasas/fisiología , Análisis por Micromatrices , Invasividad Neoplásica/genética , Interferencia de ARN , ARN Interferente Pequeño/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Factores de Transcripción de la Familia Snail , Timidina/metabolismo
9.
Development ; 139(2): 259-68, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22159578

RESUMEN

BMP activity is essential for many steps of neural development, including the initial role in neural induction and the control of progenitor identities along the dorsal-ventral axis of the neural tube. Taking advantage of chick in ovo electroporation, we show a novel role for BMP7 at the time of neurogenesis initiation in the spinal cord. Using in vivo loss-of-function experiments, we show that BMP7 activity is required for the generation of three discrete subpopulations of dorsal interneurons: dI1-dI3-dI5. Analysis of the BMP7 mouse mutant shows the conservation of this activity in mammals. Furthermore, this BMP7 activity appears to be mediated by the canonical Smad pathway, as we demonstrate that Smad1 and Smad5 activities are similarly required for the generation of dI1-dI3-dI5. Moreover, we show that this role is independent of the patterned expression of progenitor proteins in the dorsal spinal cord, but depends on the BMP/Smad regulation of specific proneural proteins, thus narrowing this BMP7 activity to the time of neurogenesis. Together, these data establish a novel role for BMP7 in primary neurogenesis, the process by which a neural progenitor exits the cell cycle and enters the terminal differentiation pathway.


Asunto(s)
Proteína Morfogenética Ósea 7/metabolismo , Interneuronas/fisiología , Neurogénesis/fisiología , Transducción de Señal/fisiología , Proteínas Smad Reguladas por Receptores/metabolismo , Médula Espinal/embriología , Análisis de Varianza , Animales , Embrión de Pollo , Inmunohistoquímica , Hibridación in Situ , Interneuronas/metabolismo , Luciferasas , Ratones , Mutación/genética , Neurogénesis/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Proteínas Smad Reguladas por Receptores/genética
10.
Dev Biol ; 265(1): 207-18, 2004 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-14697364

RESUMEN

Palate fusion is a complex process that involves the coordination of a series of cellular changes including cell death and epithelial to mesenchymal transition (EMT). Since members of the Snail family of zinc-finger regulators are involved in both triggering of the EMT and cell survival, we decided to study their putative role in palatal fusion. Furthermore, Snail genes are induced by transforming growth factor beta gene (TGF-beta) superfamily members, and TGF-beta(3) null mutant mice (TGF-beta(3)-/-) show a cleft palate phenotype. Here we show that in the wild-type mouse at the time of fusion, Snail is expressed in a few cells of the midline epithelial seam (MES), compatible with a role in triggering of the EMT in a small subpopulation of the MES. We also find an intriguing relationship between the expression of Snail family members and cell survival associated to the cleft palate condition. Indeed, Snail is expressed in the medial edge epithelial (MEE) cells in TGF-beta(3)-/-mouse embryo palates, where it is activated by the aberrant expression of its inducer, TGF-beta(1), in the underlying mesenchyme. In contrast to Snail-deficient wild-type pre-adhesion MEE cells, Snail-expressing TGF-beta(3) mutant MEE cells survive as they do their counterparts in the chick embryo. Interestingly, Slug is the Snail family member expressed in the chick MEE, providing another example of interchange of Snail and Slug expression between avian and mammalian embryos. We propose that in the absence of TGF-beta(3), TGF-beta(1) is upregulated in the mesenchyme, and that in both physiological (avian) and pathological (TGF-beta(3)-/-mammalian) cleft palates, it induces the expression of Snail genes promoting the survival of the MEE cells and permitting their subsequent differentiation into keratinized stratified epithelium.


Asunto(s)
Fisura del Paladar/veterinaria , Proteínas de Unión al ADN/fisiología , Hueso Paladar/embriología , Factores de Transcripción/fisiología , Factor de Crecimiento Transformador beta/fisiología , Dedos de Zinc/fisiología , Animales , Apoptosis/fisiología , Fisura del Paladar/embriología , Proteínas de Unión al ADN/genética , Epitelio/embriología , Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica , Inmunohistoquímica , Hibridación in Situ , Ratones , Ratones Mutantes , Factores de Transcripción de la Familia Snail , Factores de Transcripción/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...