Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Biotechnol Bioeng ; 115(8): 2061-2066, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29679475

RESUMEN

Heterogeneity in pluripotent stem cell (PSC) aggregation leads to variability in mass transfer and signaling gradients between aggregates, which results in heterogeneous differentiation and therefore variability in product quality and yield. We have characterized a chemical-based method to control aggregate size within a specific, tunable range with low heterogeneity, thereby reducing process variability in PSC expansion. This method enables controlled, scalable, stirred suspension-based manufacturing of PSC cultures that are critical for the translation of regenerative medicine strategies to clinical products.


Asunto(s)
Biotecnología/métodos , Agregación Celular , Técnicas Citológicas/métodos , Células Madre Pluripotentes/fisiología
2.
Nat Commun ; 8(1): 186, 2017 08 04.
Artículo en Inglés | MEDLINE | ID: mdl-28775249

RESUMEN

Cranial radiotherapy improves survival of the most common childhood cancers, including brain tumors and leukemia. Unfortunately, long-term survivors are faced with consequences of secondary neoplasia, including radiation-induced meningiomas (RIMs). We characterized 31 RIMs with exome/NF2 intronic sequencing, RNA sequencing and methylation profiling, and found NF2 gene rearrangements in 12/31 of RIMs, an observation previously unreported in sporadic meningioma (SM). Additionally, known recurrent mutations characteristic of SM, including AKT1, KLF4, TRAF7 and SMO, were not observed in RIMs. Combined losses of chromosomes 1p and 22q were common in RIMs (16/18 cases) and overall, chromosomal aberrations were more complex than that observed in SM. Patterns of DNA methylation profiling supported similar cell of origin between RIMs and SMs. The findings indicate that the mutational landscape of RIMs is distinct from SMs, and have significant therapeutic implications for survivors of childhood cranial radiation and the elucidation of the molecular pathogenesis of meningiomas.Radiation-induced meningiomas are often more aggressive than sporadic ones. In this study, the authors perform an exome, methylation and RNA-seq analysis of 31 cases of radiation-induced meningioma and show NF2 rearrangement, an observation previously unreported in the sporadic tumors.


Asunto(s)
Irradiación Craneana/efectos adversos , Reordenamiento Génico/genética , Genes de la Neurofibromatosis 2 , Neoplasias Meníngeas/genética , Meningioma/genética , Neoplasias Inducidas por Radiación/genética , Adulto , Anciano , Supervivientes de Cáncer , Estudios de Casos y Controles , Neoplasias Cerebelosas/radioterapia , Metilación de ADN , Femenino , Humanos , Factor 4 Similar a Kruppel , Leucemia/radioterapia , Masculino , Meduloblastoma/radioterapia , Neoplasias Meníngeas/etiología , Meningioma/etiología , Persona de Mediana Edad , Mutación , Neoplasias Inducidas por Radiación/etiología , Análisis de Secuencia de ADN , Análisis de Secuencia de ARN , Adulto Joven
3.
Nat Genet ; 48(11): 1339-1348, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27723760

RESUMEN

Schwannomas are common peripheral nerve sheath tumors that can cause debilitating morbidities. We performed an integrative analysis to determine genomic aberrations common to sporadic schwannomas. Exome sequence analysis with validation by targeted DNA sequencing of 125 samples uncovered, in addition to expected NF2 disruption, recurrent mutations in ARID1A, ARID1B and DDR1. RNA sequencing identified a recurrent in-frame SH3PXD2A-HTRA1 fusion in 12/125 (10%) cases, and genomic analysis demonstrated the mechanism as resulting from a balanced 19-Mb chromosomal inversion on chromosome 10q. The fusion was associated with male gender predominance, occurring in one out of every six men with schwannoma. Methylation profiling identified distinct molecular subgroups of schwannomas that were associated with anatomical location. Expression of the SH3PXD2A-HTRA1 fusion resulted in elevated phosphorylated ERK, increased proliferation, increased invasion and in vivo tumorigenesis. Targeting of the MEK-ERK pathway was effective in fusion-positive Schwann cells, suggesting a possible therapeutic approach for this subset of tumors.


Asunto(s)
Metilación de ADN , Neoplasias del Oído/genética , Mutación , Neurilemoma/genética , Neoplasias de la Columna Vertebral/genética , Vestíbulo del Laberinto , Proteínas Adaptadoras del Transporte Vesicular/genética , Animales , Línea Celular Tumoral , Análisis Mutacional de ADN , ADN de Neoplasias , Exoma , Femenino , Fusión Génica , Genoma Humano , Serina Peptidasa A1 que Requiere Temperaturas Altas , Humanos , Masculino , Ratones , Ratones Endogámicos NOD , Ratones SCID , ARN Neoplásico , Análisis de Secuencia de ADN , Análisis de Secuencia de ARN , Serina Endopeptidasas/genética
4.
Oncotarget ; 7(43): 69518-69535, 2016 10 25.
Artículo en Inglés | MEDLINE | ID: mdl-27588472

RESUMEN

First-line cancer therapies such as alkylating agents and radiation have limited survival benefits for Glioblastoma (GBM) patients. Current research strongly supports the notion that inhibition of aberrant tumor metabolism holds promise as a therapeutic strategy when used in combination with radiation and chemotherapy. Hexokinase 2 (HK2) has been shown to be a key driver of altered metabolism in GBM, and presents an attractive therapeutic target. To date, no study has fully assessed the therapeutic value of targeting HK2 as a mechanism to sensitize cells to standard therapy, namely in the form of radiation and temozolomide (TMZ). Using cell lines and primary cultures of GBM, we showed that inducible knockdown of HK2 altered tumor metabolism, which could not be recapitulated by HK1 or HK3 loss. HK2 loss diminished both in vivo tumor vasculature as well as growth within orthotopic intracranial xenograft models of GBMs, and the survival benefit was additive with radiation and TMZ. Radio-sensitization following inhibition of HK2 was mediated by increased DNA damage, and could be rescued through constitutive activation of ERK signaling. This study supports HK2 as a potentially effective therapeutic target in GBM.


Asunto(s)
Neoplasias Encefálicas/terapia , Glioblastoma/terapia , Hexoquinasa/genética , Interferencia de ARN , Ensayos Antitumor por Modelo de Xenoinjerto , Animales , Antineoplásicos Alquilantes/farmacología , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Proliferación Celular/efectos de la radiación , Quimioradioterapia , Daño del ADN , Dacarbazina/análogos & derivados , Dacarbazina/farmacología , Glioblastoma/metabolismo , Glioblastoma/patología , Células HEK293 , Hexoquinasa/metabolismo , Humanos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Sistema de Señalización de MAP Quinasas/efectos de la radiación , Masculino , Ratones Endogámicos NOD , Ratones SCID , Temozolomida
5.
Proteomics ; 15(18): 3219-31, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26080932

RESUMEN

Cellular reprogramming remodels the gene expression program by re-setting the epigenome of somatic cells into an embryonic-like pluripotent state. Post-translational modifications of histones play an important role in this process. Previously, we found by ChIP-seq widespread changes of specific histone H3 marks in two divergent reprogramming routes leading to alternative pluripotent sates . Here, using an unbiased middle-down proteomics approach we have identified 72 unique isoforms of histone H4 and quantified 56 of them in the same set of samples. We found substantial differences between somatic and late-phase reprogramming cells. Also, ESCs and iPSCs displayed higher levels of H4 acetylation and tri-methylation concomitantly with lower levels of mono- and di-methylation when compared to cells undergoing reprogramming. Our data shows that the epigenetic remodeling induced by the reprogramming process goes beyond histone H3 and reveals the importance of H4 modifications as well. The presented data is a valuable resource to study the epigenetic mechanisms involved in the acquisition of induced pluripotency. All MS data have been deposited in the ProteomeXchange with identifier PXD002062 (http://proteomecentral.proteomexchange.org/dataset/PXD002062).


Asunto(s)
Reprogramación Celular/fisiología , Epigénesis Genética/fisiología , Histonas/química , Histonas/metabolismo , Células Madre Pluripotentes/metabolismo , Proteómica/métodos , Animales , Células Cultivadas , Análisis por Conglomerados , Ratones , Isoformas de Proteínas/química , Isoformas de Proteínas/metabolismo , Proteoma/análisis , Proteoma/química , Proteoma/metabolismo
6.
Nat Commun ; 6: 7329, 2015 Jun 16.
Artículo en Inglés | MEDLINE | ID: mdl-26076835

RESUMEN

Reprogramming is a dynamic process that can result in multiple pluripotent cell types emerging from divergent paths. Cell surface protein expression is a particularly desirable tool to categorize reprogramming and pluripotency as it enables robust quantification and enrichment of live cells. Here we use cell surface proteomics to interrogate mouse cell reprogramming dynamics and discover CD24 as a marker that tracks the emergence of reprogramming-responsive cells, while enabling the analysis and enrichment of transgene-dependent (F-class) and -independent (traditional) induced pluripotent stem cells (iPSCs) at later stages. Furthermore, CD24 can be used to delineate epiblast stem cells (EpiSCs) from embryonic stem cells (ESCs) in mouse pluripotent culture. Importantly, regulated CD24 expression is conserved in human pluripotent stem cells (PSCs), tracking the conversion of human ESCs to more naive-like PSC states. Thus, CD24 is a conserved marker for tracking divergent states in both reprogramming and standard pluripotent culture.


Asunto(s)
Antígeno CD24/metabolismo , Reprogramación Celular , Células Madre Embrionarias Humanas/metabolismo , Células Madre Pluripotentes Inducidas/metabolismo , Células Madre Embrionarias de Ratones/metabolismo , Animales , Estratos Germinativos/citología , Células Madre Embrionarias Humanas/citología , Humanos , Células Madre Pluripotentes Inducidas/citología , Ratones , Células Madre Embrionarias de Ratones/citología , Células Madre/citología , Células Madre/metabolismo
9.
Nature ; 516(7530): 192-7, 2014 Dec 11.
Artículo en Inglés | MEDLINE | ID: mdl-25503232

RESUMEN

Pluripotency is defined by the ability of a cell to differentiate to the derivatives of all the three embryonic germ layers: ectoderm, mesoderm and endoderm. Pluripotent cells can be captured via the archetypal derivation of embryonic stem cells or via somatic cell reprogramming. Somatic cells are induced to acquire a pluripotent stem cell (iPSC) state through the forced expression of key transcription factors, and in the mouse these cells can fulfil the strictest of all developmental assays for pluripotent cells by generating completely iPSC-derived embryos and mice. However, it is not known whether there are additional classes of pluripotent cells, or what the spectrum of reprogrammed phenotypes encompasses. Here we explore alternative outcomes of somatic reprogramming by fully characterizing reprogrammed cells independent of preconceived definitions of iPSC states. We demonstrate that by maintaining elevated reprogramming factor expression levels, mouse embryonic fibroblasts go through unique epigenetic modifications to arrive at a stable, Nanog-positive, alternative pluripotent state. In doing so, we prove that the pluripotent spectrum can encompass multiple, unique cell states.


Asunto(s)
Reprogramación Celular/genética , Reprogramación Celular/fisiología , Epigénesis Genética , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/metabolismo , Animales , Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Femenino , Fibroblastos/clasificación , Fibroblastos/citología , Fibroblastos/metabolismo , Histona Desacetilasas/metabolismo , Células Madre Pluripotentes Inducidas/clasificación , Ratones , Ratones Desnudos , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Transgenes/genética
10.
Nature ; 516(7530): 198-206, 2014 Dec 11.
Artículo en Inglés | MEDLINE | ID: mdl-25503233

RESUMEN

Somatic cell reprogramming to a pluripotent state continues to challenge many of our assumptions about cellular specification, and despite major efforts, we lack a complete molecular characterization of the reprograming process. To address this gap in knowledge, we generated extensive transcriptomic, epigenomic and proteomic data sets describing the reprogramming routes leading from mouse embryonic fibroblasts to induced pluripotency. Through integrative analysis, we reveal that cells transition through distinct gene expression and epigenetic signatures and bifurcate towards reprogramming transgene-dependent and -independent stable pluripotent states. Early transcriptional events, driven by high levels of reprogramming transcription factor expression, are associated with widespread loss of histone H3 lysine 27 (H3K27me3) trimethylation, representing a general opening of the chromatin state. Maintenance of high transgene levels leads to re-acquisition of H3K27me3 and a stable pluripotent state that is alternative to the embryonic stem cell (ESC)-like fate. Lowering transgene levels at an intermediate phase, however, guides the process to the acquisition of ESC-like chromatin and DNA methylation signature. Our data provide a comprehensive molecular description of the reprogramming routes and is accessible through the Project Grandiose portal at http://www.stemformatics.org.


Asunto(s)
Reprogramación Celular/genética , Genoma/genética , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/metabolismo , Animales , Cromatina/química , Cromatina/genética , Cromatina/metabolismo , Ensamble y Desensamble de Cromatina , Metilación de ADN , Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Epistasis Genética/genética , Fibroblastos/citología , Fibroblastos/metabolismo , Histonas/química , Histonas/metabolismo , Internet , Ratones , Proteoma/genética , Proteómica , ARN Largo no Codificante/genética , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Transcripción Genética/genética , Transcriptoma/genética , Transgenes/genética
11.
Nat Commun ; 5: 5619, 2014 Dec 10.
Artículo en Inglés | MEDLINE | ID: mdl-25493341

RESUMEN

Reprogramming of somatic cells to induced pluripotent stem cells involves a dynamic rearrangement of the epigenetic landscape. To characterize this epigenomic roadmap, we have performed MethylC-seq, ChIP-seq (H3K4/K27/K36me3) and RNA-Seq on samples taken at several time points during murine secondary reprogramming as part of Project Grandiose. We find that DNA methylation gain during reprogramming occurs gradually, while loss is achieved only at the ESC-like state. Binding sites of activated factors exhibit focal demethylation during reprogramming, while ESC-like pluripotent cells are distinguished by extension of demethylation to the wider neighbourhood. We observed that genes with CpG-rich promoters demonstrate stable low methylation and strong engagement of histone marks, whereas genes with CpG-poor promoters are safeguarded by methylation. Such DNA methylation-driven control is the key to the regulation of ESC-pluripotency genes, including Dppa4, Dppa5a and Esrrb. These results reveal the crucial role that DNA methylation plays as an epigenetic switch driving somatic cells to pluripotency.

12.
Nat Commun ; 5: 5522, 2014 Dec 10.
Artículo en Inglés | MEDLINE | ID: mdl-25494340

RESUMEN

MicroRNAs (miRNAs) are critical to somatic cell reprogramming into induced pluripotent stem cells (iPSCs), however, exactly how miRNA expression changes support the transition to pluripotency requires further investigation. Here we use a murine secondary reprogramming system to sample cellular trajectories towards iPSCs or a novel pluripotent 'F-class' state and perform small RNA sequencing. We detect sweeping changes in an early and a late wave, revealing that distinct miRNA milieus characterize alternate states of pluripotency. miRNA isoform expression is common but surprisingly varies little between cell states. Referencing other omic data sets generated in parallel, we find that miRNA expression is changed through transcriptional and post-transcriptional mechanisms. miRNA transcription is commonly regulated by dynamic histone modification, while DNA methylation/demethylation consolidates these changes at multiple loci. Importantly, our results suggest that a novel subset of distinctly expressed miRNAs supports pluripotency in the F-class state, substituting for miRNAs that serve such roles in iPSCs.

13.
Nat Commun ; 5: 5613, 2014 Dec 10.
Artículo en Inglés | MEDLINE | ID: mdl-25494451

RESUMEN

The ectopic expression of Oct4, Klf4, c-Myc and Sox2 (OKMS) transcription factors allows reprogramming of somatic cells into induced pluripotent stem cells (iPSCs). The reprogramming process, which involves a complex network of molecular events, is not yet fully characterized. Here we perform a quantitative mass spectrometry-based analysis to probe in-depth dynamic proteome changes during somatic cell reprogramming. Our data reveal defined waves of proteome resetting, with the first wave occurring 48 h after the activation of the reprogramming transgenes and involving specific biological processes linked to the c-Myc transcriptional network. A second wave of proteome reorganization occurs in a later stage of reprogramming, where we characterize the proteome of two distinct pluripotent cellular populations. In addition, the overlay of our proteome resource with parallel generated -omics data is explored to identify post-transcriptionally regulated proteins involved in key steps during reprogramming.

14.
Stem Cell Reports ; 3(1): 156-68, 2014 Jul 08.
Artículo en Inglés | MEDLINE | ID: mdl-25068129

RESUMEN

Conversion of EpiSCs to naive ESCs is a rare event that is driven by the reestablishment of the naive transcription factor network. In mice, STAT3 activation is sufficient to drive conversion of EpiSCs to the naive pluripotent stem cell (PSC) state. However, the lack of responsiveness of EpiSCs to LIF presents a bottleneck in this conversion process. Here, we demonstrate that local accumulation of BMP-SMAD1 signaling, in cooperation with GP130 ligands, enhances the recovery of LIF responsiveness by directly controlling transcription of the LIF receptor (Lif-r). Addition of BMP and LIF to EpiSCs increases both LIF responsiveness and conversion frequencies to naive PSCs. Mechanistically, we show that the transcriptional cofactor P300 plays a critical role by mediating complex formation between STAT3 and SMAD1. This demonstration of how the local microenvironment or stem cell niche reactivates dormant signaling responsiveness and developmental potential may be applicable to other stem cell niche-containing systems.


Asunto(s)
Células Madre Embrionarias/citología , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/metabolismo , Receptores OSM-LIF/metabolismo , Factor de Transcripción STAT3/metabolismo , Proteína Smad1/metabolismo , Fosfatasa Alcalina/metabolismo , Animales , Diferenciación Celular/fisiología , Células Cultivadas , Inmunoprecipitación de Cromatina , Células Madre Embrionarias/metabolismo , Humanos , Inmunohistoquímica , Ratones , Receptores OSM-LIF/genética , Factor de Transcripción STAT3/genética , Proteína Smad1/genética
15.
Integr Biol (Camb) ; 4(11): 1367-76, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22990140

RESUMEN

Embryonic stem cells (ESC) and epiblast stem cells (EpiSC) are distinct pluripotent stem cell states that require different signaling pathways for their self-renewal. Forward transitions between ESC and EpiSC can be accomplished by changing culture conditions; however reverse transitions between EpiSC and ESC are rare events that require transgene insertion or culture on feeders. We demonstrate that transgene-free reversion of EpiSCs to ESCs can be enhanced by local microenvironmental control and the subsequent reactivation of dormant LIF-STAT3 signaling. Reactivation of LIF responsiveness occurs in regions of colony constraint (high local cell density) typical of culture on feeders, a condition that can be recapitulated using micropatterned (µP) colonies under defined conditions. This increased LIF responsiveness results in a subsequent increase in the frequency of EpiSC reversion. Importantly, the resulting revertant EpiSCs are functionally indistinguishable from naïve mESC. Our findings demonstrate that signaling pathway activation and repression create barriers to cell fate transitions that can be overcome by microenvironmental control.


Asunto(s)
Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Estratos Germinativos/citología , Estratos Germinativos/metabolismo , Quinasas Janus/metabolismo , Factores de Transcripción STAT/metabolismo , Animales , Recuento de Células , Diferenciación Celular/efectos de los fármacos , Línea Celular , Receptor gp130 de Citocinas/metabolismo , Células Madre Embrionarias/efectos de los fármacos , Estratos Germinativos/efectos de los fármacos , Factor Inhibidor de Leucemia/metabolismo , Factor Inhibidor de Leucemia/farmacología , Ratones , Modelos Biológicos , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/efectos de los fármacos , Células Madre Pluripotentes/metabolismo , Factor de Transcripción STAT3/metabolismo , Transducción de Señal , Nicho de Células Madre
16.
Nat Methods ; 9(5): 509-16, 2012 Mar 25.
Artículo en Inglés | MEDLINE | ID: mdl-22447133

RESUMEN

We describe derivation of induced pluripotent stem cells (iPSCs) from terminally differentiated mouse cells in serum- and feeder-free stirred suspension cultures. Temporal analysis of global gene expression revealed high correlations between cells reprogrammed in suspension and cells reprogrammed in adhesion-dependent conditions. Suspension culture-reprogrammed iPSCs (SiPSCs) could be differentiated into all three germ layers in vitro and contributed to chimeric embryos in vivo. SiPSC generation allowed for efficient selection of reprogramming factor-expressing cells based on their differential survival and proliferation in suspension culture. Seamless integration of SiPSC reprogramming and directed differentiation enabled scalable production of beating cardiac cells in a continuous single cell- and small aggregate-based process. This method is an important step toward the development of robust PSC generation, expansion and differentiation technology.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Fibroblastos/citología , Células Madre Pluripotentes Inducidas/citología , Miocitos Cardíacos/citología , Animales , Diferenciación Celular/fisiología , Reprogramación Celular/fisiología , Quimera/fisiología , Ratones
17.
Stem Cell Res ; 7(2): 145-53, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21763622

RESUMEN

Human embryonic stem (ES) cells can undergo spontaneously differentiation in standard culture conditions, demonstrating that the undifferentiated state is relatively unstable. The heterogeneous expression of SSEA3 observed within human ES colonies, provides a means to examine undifferentiated stem cell substates. Through functional testing of single cells we have shown that undifferentiated ES cells can be segregated into functionally discrete subpopulations on the basis of SSEA3 expression: SSEA3(High), SSEA(Low) and SSEA3(Negative). Human ES subpopulations were found to be interconvertible, but they possess distinct properties when challenged to differentiate along the neural lineage. These data suggest that ES cells with pluripotent/self-renewal capacities can exhibit different responses to induction of differentiation.


Asunto(s)
Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/metabolismo , Antígenos de Carbohidratos Asociados a Tumores/metabolismo , Diferenciación Celular/fisiología , Línea Celular , Línea Celular Tumoral , Humanos , Antígenos Embrionarios Específico de Estadio/metabolismo
18.
PLoS One ; 5(5): e10901, 2010 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-20531938

RESUMEN

The mechanism by which an apparently uniform population of cells can generate a heterogeneous population of differentiated derivatives is a fundamental aspect of pluripotent and multipotent stem cell behaviour. One possibility is that the environment and the differentiation cues to which the cells are exposed are not uniform. An alternative, but not mutually exclusive possibility is that the observed heterogeneity arises from the stem cells themselves through the existence of different interconvertible substates that pre-exist before the cells commit to differentiate. We have tested this hypothesis in the case of apparently homogeneous pluripotent human embryonal carcinoma (EC) stem cells, which do not follow a uniform pattern of differentiation when exposed to retinoic acid. Instead, they produce differentiated progeny that include both neuronal and non-neural phenotypes. Our results suggest that pluripotent NTERA2 stem cells oscillate between functionally distinct substates that are primed to select distinct lineages when differentiation is induced.


Asunto(s)
Compartimento Celular , Diferenciación Celular , Células Madre/citología , Carcinoma Embrionario/patología , Linaje de la Célula , Células Clonales , Humanos , Modelos Biológicos , Fenotipo
19.
Differentiation ; 80(1): 20-30, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20427117

RESUMEN

Differentiation of human embryonic stem (ES) cells and embryonal carcinoma (EC) cells provides an in vitro model to study the process of neuronal differentiation. Retinoic acid (RA) is frequently used to promote neural differentiation of pluripotent cells under a wide variety of culture conditions. Through systematic comparison of differentiation conditions we demonstrate that RA induced neuronal differentiation of human ES and EC cells requires prolonged RA exposure and intercellular communication mediated by high cell density. These parameters are necessary for the up-regulation of neural gene expression (SOX2, PAX6 and NeuroD1) and the eventual appearance of neurons. Forced over-expression of neither SOX2 nor NEUROD1 was sufficient to overcome the density dependency of neuronal differentiation. Furthermore, inhibition of GSK3beta activity blocked the ability of RA to direct cell differentiation along the neural lineage, suggesting a role for appropriately regulated WNT signalling. These data indicate that RA mediated neuronal differentiation of human EC and ES cell lines is not a cell autonomous program but comprises of a multi-staged program that requires intercellular input.


Asunto(s)
Carcinoma Embrionario/patología , Diferenciación Celular/efectos de los fármacos , Células Madre de Carcinoma Embrionario/efectos de los fármacos , Neuronas/citología , Neuronas/efectos de los fármacos , Células Madre Pluripotentes/efectos de los fármacos , Tretinoina/farmacología , Antineoplásicos/farmacología , Western Blotting , Carcinoma Embrionario/tratamiento farmacológico , Carcinoma Embrionario/metabolismo , Ciclo Celular , Proliferación Celular , Células Madre de Carcinoma Embrionario/metabolismo , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Humanos , Neuronas/metabolismo , Células Madre Pluripotentes/metabolismo , ARN Mensajero/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA