Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
1.
Nature ; 532(7597): 107-11, 2016 Apr 07.
Artículo en Inglés | MEDLINE | ID: mdl-26982723

RESUMEN

Diploidy is a fundamental genetic feature in mammals, in which haploid cells normally arise only as post-meiotic germ cells that serve to ensure a diploid genome upon fertilization. Gamete manipulation has yielded haploid embryonic stem (ES) cells from several mammalian species, but haploid human ES cells have yet to be reported. Here we generated and analysed a collection of human parthenogenetic ES cell lines originating from haploid oocytes, leading to the successful isolation and maintenance of human ES cell lines with a normal haploid karyotype. Haploid human ES cells exhibited typical pluripotent stem cell characteristics, such as self-renewal capacity and a pluripotency-specific molecular signature. Moreover, we demonstrated the utility of these cells as a platform for loss-of-function genetic screening. Although haploid human ES cells resembled their diploid counterparts, they also displayed distinct properties including differential regulation of X chromosome inactivation and of genes involved in oxidative phosphorylation, alongside reduction in absolute gene expression levels and cell size. Surprisingly, we found that a haploid human genome is compatible not only with the undifferentiated pluripotent state, but also with differentiated somatic fates representing all three embryonic germ layers both in vitro and in vivo, despite a persistent dosage imbalance between the autosomes and X chromosome. We expect that haploid human ES cells will provide novel means for studying human functional genomics and development.


Asunto(s)
Diferenciación Celular , Estudios de Asociación Genética/métodos , Haploidia , Células Madre Embrionarias Humanas/citología , Células Madre Embrionarias Humanas/metabolismo , Autorrenovación de las Células , Separación Celular , Tamaño de la Célula , Cromosomas Humanos X/genética , Diploidia , Regulación hacia Abajo/genética , Eliminación de Gen , Estratos Germinativos/citología , Humanos , Cariotipificación , Oocitos/metabolismo , Fosforilación Oxidativa , Partenogénesis , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/metabolismo , Inactivación del Cromosoma X/genética
2.
Stem Cell Reports ; 18(5): 1049-1060, 2023 05 09.
Artículo en Inglés | MEDLINE | ID: mdl-37116485

RESUMEN

Humans are diploid organisms, and triploidy in human embryos is responsible for ∼10% of spontaneous miscarriages. Surprisingly, some pregnancies proceed to triploid newborns that suffer from many neuro-developmental disorders. To investigate the impact of triploidy on human development, we generate triploid human embryonic stem cells (hESCs) by fusing isogenic haploid and diploid hESCs. Comparison of the transcriptome, methylome, and genome-wide replication timing shows general similarity between diploid and triploid hESCs. However, triploid cells have a larger volume than diploid cells, demonstrating decreased surface-area-to-volume ratio. This leads to a significant downregulation of cell surface ion channel genes, which are more essential in neural progenitors than in undifferentiated cells, leading to inhibition of differentiation, and it affects the neuronal differentiation ability of triploid hESCs, both in vitro and in vivo. Notably, our research establishes a platform to study triploidy in humans and points to their pathology as observed in triploid embryos.


Asunto(s)
Células Madre Embrionarias Humanas , Triploidía , Recién Nacido , Embarazo , Femenino , Humanos , Diferenciación Celular/genética , Genoma , Genómica , Diploidia
3.
Cells ; 12(11)2023 05 29.
Artículo en Inglés | MEDLINE | ID: mdl-37296624

RESUMEN

ATM depletion is associated with the multisystemic neurodegenerative syndrome ataxia-telangiectasia (A-T). The exact linkage between neurodegeneration and ATM deficiency has not been established yet, and no treatment is currently available. In this study, we aimed to identify synthetic viable genes in ATM deficiency to highlight potential targets for the treatment of neurodegeneration in A-T. We inhibited ATM kinase activity using the background of a genome-wide haploid pluripotent CRISPR/Cas9 loss-of-function library and examined which mutations confer a growth advantage on ATM-deficient cells specifically. Pathway enrichment analysis of the results revealed the Hippo signaling pathway as a major negative regulator of cellular growth upon ATM inhibition. Indeed, genetic perturbation of the Hippo pathway genes SAV1 and NF2, as well as chemical inhibition of this pathway, specifically promoted the growth of ATM-knockout cells. This effect was demonstrated in both human embryonic stem cells and neural progenitor cells. Therefore, we suggest the Hippo pathway as a candidate target for the treatment of the devastating cerebellar atrophy associated with A-T. In addition to the Hippo pathway, our work points out additional genes, such as the apoptotic regulator BAG6, as synthetic viable with ATM-deficiency. These genes may help to develop drugs for the treatment of A-T patients as well as to define biomarkers for resistance to ATM inhibition-based chemotherapies and to gain new insights into the ATM genetic network.


Asunto(s)
Ataxia Telangiectasia , Células Madre Embrionarias Humanas , Humanos , Proteínas de la Ataxia Telangiectasia Mutada/genética , Proteínas de la Ataxia Telangiectasia Mutada/metabolismo , Vía de Señalización Hippo , Células Madre Embrionarias Humanas/metabolismo , Redes Reguladoras de Genes , Ataxia Telangiectasia/tratamiento farmacológico , Chaperonas Moleculares/metabolismo
4.
Cell Prolif ; 56(6): e13475, 2023 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-37086010

RESUMEN

Anticancer drugs are at the frontline of cancer therapy. However, innate resistance to these drugs occurs in one-third to one-half of patients, exposing them to the side effects of these drugs with no meaningful benefit. To identify the genes and pathways that confer resistance to such therapies, we performed a genome-wide screen in haploid human embryonic stem cells (hESCs). These cells possess the advantage of having only one copy of each gene, harbour a normal karyotype, and lack any underlying point mutations. We initially show a close correlation between the potency of anticancer drugs in cancer cell lines to those in hESCs. We then exposed a genome-wide loss-of-function library of mutations in all protein-coding genes to 10 selected anticancer drugs, which represent five different mechanisms of drug therapies. The genetic screening enabled us to identify genes and pathways which can confer resistance to these drugs, demonstrating several common pathways. We validated a few of the resistance-conferring genes, demonstrating a significant shift in the effective drug concentrations to indicate a drug-specific effect to these genes. Strikingly, the p53 signalling pathway seems to induce resistance to a large array of anticancer drugs. The data shows dramatic effects of loss of p53 on resistance to many but not all drugs, calling for clinical evaluation of mutations in this gene prior to anticancer therapy.


Asunto(s)
Antineoplásicos , Células Madre Embrionarias Humanas , Humanos , Células Madre Embrionarias Humanas/metabolismo , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo , Haploidia , Antineoplásicos/farmacología , Antineoplásicos/metabolismo , Mutación
5.
Stem Cells ; 29(4): 600-8, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21328508

RESUMEN

The Spemann-Mangold organizer is the structure that provides the signals, which initiate pattern formation in the developing vertebrate embryo, affecting the main body axes. Very little is known about axial induction in the gastrulating human embryo, as research is hindered by obvious ethical restrictions. Human embryonic stem cells (hESCs) are pluripotent cells derived from the pregastrula embryo that can differentiate in culture following a program similar to normal embryonic development but without pattern formation. Here, we show that in hESC-derived embryoid bodies, we can induce differentiation of cells that harbor markers and characteristics of the gastrula-organizer. Moreover, genetic labeling of these cells enabled their purification, and the discovery of a comprehensive set of their secreted proteins, cell surface receptors, and nuclear factors characteristic of the organizer. Remarkably, transplantation of cell populations enriched for the putative human organizer into frog embryos induced a secondary axis. Our research demonstrates that the human organizer can be induced in vitro and paves the way for the study of pattern formation and the initial regulation of body axis establishment in humans.


Asunto(s)
Células Madre Embrionarias/metabolismo , Gastrulación , Organizadores Embrionarios/metabolismo , Tipificación del Cuerpo , Diferenciación Celular/fisiología , Cuerpos Embrioides/metabolismo , Inducción Embrionaria , Regulación del Desarrollo de la Expresión Génica , Proteína Goosecoide/biosíntesis , Humanos , Transducción de Señal , Factor de Crecimiento Transformador beta/metabolismo , Proteínas Wnt/metabolismo
6.
Mol Cell Neurosci ; 46(1): 187-99, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20849957

RESUMEN

The development of the neural tube into a complex central nervous system involves morphological, cellular and molecular changes, all of which are tightly regulated. The floor plate (FP) is a critical organizing center located at the ventral-most midline of the neural tube. FP cells regulate dorsoventral patterning, differentiation and axon guidance by secreting morphogens. Here we show that the bHLH transcription factor Nato3 (Ferd3l) is specifically expressed in the spinal FP of chick and mouse embryos. Using in ovo electroporation to understand the regulation of the FP-specific expression of Nato3, we have identified an evolutionarily conserved 204 bp genomic region, which is necessary and sufficient to drive expression to the chick FP. This promoter contains two Foxa2-binding sites, which are highly conserved among distant phyla. The two sites can bind Foxa2 in vitro, and are necessary for the expression in the FP in vivo. Gain and loss of Foxa2 function in vivo further emphasize its role in Nato3 promoter activity. Thus, our data suggest that Nato3 is a direct target of Foxa2, a transcription activator and effector of Sonic hedgehog, the hallmark regulator of FP induction and spinal cord development. The identification of the FP-specific promoter is an important step towards a better understanding of the molecular mechanisms through which Nato3 transcription is regulated and for uncovering its function during nervous system development. Moreover, the promoter provides us with a powerful tool for conditional genetic manipulations in the FP.


Asunto(s)
Evolución Molecular , Regulación del Desarrollo de la Expresión Génica , Factor Nuclear 3-beta del Hepatocito/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Tubo Neural/anatomía & histología , Tubo Neural/fisiología , Regiones Promotoras Genéticas , Factores de Transcripción/metabolismo , Animales , Secuencia de Bases , Sitios de Unión , Embrión de Pollo , Factor Nuclear 3-beta del Hepatocito/genética , Humanos , Ratones , Ratones Endogámicos C57BL , Datos de Secuencia Molecular , Proteínas del Tejido Nervioso/genética , Proteínas Represoras , Alineación de Secuencia , Médula Espinal/anatomía & histología , Médula Espinal/embriología , Factores de Transcripción/genética
7.
Cell Rep ; 38(13): 110573, 2022 03 29.
Artículo en Inglés | MEDLINE | ID: mdl-35354027

RESUMEN

Haploinsufficiency describes a phenomenon where one functioning allele is insufficient for a normal phenotype, underlying several human diseases. The effect of haploinsufficiency on human embryonic stem cells (hESC) has not been thoroughly studied. To establish a genome-wide loss-of-function screening for heterozygous mutations, we fuse normal haploid hESCs with a library of mutant haploid hESCs. We identify over 600 genes with a negative effect on hESC growth in a haploinsufficient manner and characterize them as genes showing less tolerance to mutations, conservation during evolution, and depletion from telomeres and X chromosome. Interestingly, a large fraction of these genes is associated with extracellular matrix and plasma membrane and enriched for genes within WNT and TGF-ß pathways. We thus identify haploinsufficiency-related genes that show growth retardation in early embryonic cells, suggesting dosage-dependent phenotypes in hESCs. Overall, we construct a unique model for studying haploinsufficiency and identified important dosage-dependent pathways involved in hESC growth and survival.


Asunto(s)
Células Madre Embrionarias Humanas , Alelos , Haploidia , Haploinsuficiencia/genética , Heterocigoto , Humanos
8.
Stem Cells ; 28(1): 75-83, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19921748

RESUMEN

Human embryoid bodies (HEBs) are cell aggregates that are produced during the course of embryonic stem cell differentiation in suspension. Mature HEBs have been shown to contain derivatives of the three embryonic germ layers. In this study, using a combination of laser capture microscopy followed by DNA microarray analysis and cell sorting, we demonstrate that early HEBs are composed of three major cell populations. These cell populations can be defined by the expression of specific cell markers, namely: (i) OCT4(+), REX1(-); (ii) NCAD(+), OCT4(-); and (iii) EPOR(+), OCT4(-). By analyzing gene expression in embryonic tissues, these cell populations could respectively be assigned to the embryonic ectoderm, mesendoderm, and extraembryonic endoderm lineages. We show that the extraembryonic endoderm, which selectively expresses platelet-derived growth factor B (PDGF-B), negatively affects the mesendoderm lineage, which selectively expresses the receptor PDGFRA. Our analysis suggests that early HEBs are spatially patterned and that cell differentiation is governed by interactions between the different cell types.


Asunto(s)
Comunicación Celular , Diferenciación Celular , Linaje de la Célula , Células Madre Embrionarias/fisiología , Endodermo/fisiología , Gástrula/fisiología , Regulación del Desarrollo de la Expresión Génica , Mesodermo/fisiología , Animales , Antígenos CD/genética , Antígenos CD/metabolismo , Biomarcadores/metabolismo , Cadherinas/genética , Cadherinas/metabolismo , Comunicación Celular/genética , Diferenciación Celular/genética , Línea Celular , Linaje de la Célula/genética , Separación Celular , Células Madre Embrionarias/metabolismo , Endodermo/citología , Endodermo/metabolismo , Citometría de Flujo , Gástrula/citología , Gástrula/metabolismo , Perfilación de la Expresión Génica/métodos , Humanos , Factores de Transcripción de Tipo Kruppel/genética , Factores de Transcripción de Tipo Kruppel/metabolismo , Rayos Láser , Mesodermo/citología , Mesodermo/metabolismo , Ratones , Microdisección/instrumentación , Factor 3 de Transcripción de Unión a Octámeros/genética , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , Proteínas Proto-Oncogénicas c-sis/genética , Proteínas Proto-Oncogénicas c-sis/metabolismo , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/genética , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Receptores de Eritropoyetina/genética , Receptores de Eritropoyetina/metabolismo , Transcripción Genética
9.
Stem Cells ; 28(9): 1530-40, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20641042

RESUMEN

Syndromes caused by chromosomal aneuploidies are widely recognized genetic disorders in humans and often lead to spontaneous miscarriage. Preimplantation genetic screening is used to detect chromosomal aneuploidies in early embryos. Our aim was to derive aneuploid human embryonic stem cell (hESC) lines that may serve as models for human syndromes caused by aneuploidies. We have established 25 hESC lines from blastocysts diagnosed as aneuploid on day 3 of their in vitro development. The hESC lines exhibited morphology and expressed markers typical of hESCs. They demonstrated long-term proliferation capacity and pluripotent differentiation. Karyotype analysis revealed that two-third of the cell lines carry a normal euploid karyotype, while one-third remained aneuploid throughout the derivation, resulting in eight hESC lines carrying either trisomy 13 (Patau syndrome), 16, 17, 21 (Down syndrome), X (Triple X syndrome), or monosomy X (Turner syndrome). On the basis of the level of single nucleotide polymorphism heterozygosity in the aneuploid chromosomes, we determined whether the aneuploidy originated from meiotic or mitotic chromosomal nondisjunction. Gene expression profiles of the trisomic cell lines suggested that all three chromosomes are actively transcribed. Our analysis allowed us to determine which tissues are most affected by the presence of a third copy of either chromosome 13, 16, 17 or 21 and highlighted the effects of trisomies on embryonic development. The results presented here suggest that aneuploid embryos can serve as an alternative source for either normal euploid or aneuploid hESC lines, which represent an invaluable tool to study developmental aspects of chromosomal abnormalities in humans.


Asunto(s)
Aneuploidia , Aberraciones Cromosómicas , Trastornos de los Cromosomas/genética , Cromosomas Humanos , Células Madre Embrionarias/patología , Regulación del Desarrollo de la Expresión Génica , Células Madre Pluripotentes/patología , Diferenciación Celular/genética , Línea Celular , Proliferación Celular , Trastornos de los Cromosomas/diagnóstico , Trastornos de los Cromosomas/patología , Cromosomas Humanos Par 13 , Cromosomas Humanos Par 16 , Cromosomas Humanos Par 17 , Cromosomas Humanos Par 21 , Análisis por Conglomerados , Perfilación de la Expresión Génica , Pruebas Genéticas , Humanos , Cariotipificación , Diagnóstico Preimplantación/métodos , Síndrome
10.
Stem Cell Reports ; 15(6): 1275-1286, 2020 12 08.
Artículo en Inglés | MEDLINE | ID: mdl-32559458

RESUMEN

Chromatin regulators play fundamental roles in controlling pluripotency and differentiation. We examined the effect of mutations in 703 genes from nearly 70 chromatin-modifying complexes on human embryonic stem cell (ESC) growth. While the vast majority of chromatin-associated complexes are essential for ESC growth, the only complexes that conferred growth advantage upon mutation of their members, were the repressive complexes LSD-CoREST and BHC. Both complexes include the most potent growth-restricting chromatin-related protein, ZMYM2. Interestingly, while ZMYM2 expression is rather low in human blastocysts, its expression peaks in primed ESCs and is again downregulated upon differentiation. ZMYM2-null ESCs overexpress pluripotency genes and show genome-wide promotor-localized histone H3 hyper-acetylation. These mutant cells were also refractory to differentiate in vitro and failed to produce teratomas upon injection into immunodeficient mice. Our results suggest a central role for ZMYM2 in the transcriptional regulation of the undifferentiated state and in the exit-from-pluripotency of human ESCs.


Asunto(s)
Proliferación Celular , Proteínas de Unión al ADN/metabolismo , Mutación , Proteínas de Neoplasias/metabolismo , Teratoma/metabolismo , Factores de Transcripción/metabolismo , Animales , Proteínas de Unión al ADN/genética , Células Madre Embrionarias Humanas , Humanos , Ratones , Ratones SCID , Proteínas de Neoplasias/genética , Teratoma/genética , Teratoma/patología , Factores de Transcripción/genética
11.
Stem Cells ; 26(7): 1874-82, 2008 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-18450823

RESUMEN

Human embryonic stem cells (HESCs) are pluripotent cells derived from the inner cell mass of preimplantation embryos. In this study, to isolate new lines of HESCs, we used blastocyst-stage embryos diagnosed as aneuploid in preimplantation genetic screening (PGS). During in vitro fertilization treatments, PGS is widely applied to identify chromosomal aneuploidies, especially in cases of advanced maternal age. Embryos that are detected as carrying aneuploidies are destined to be discarded unless donated for research. From 74 fresh PGS-defined aneuploid embryos, we derived seven HESC lines. Most of the embryos were left to hatch spontaneously through the hole created for blastomere biopsy and further treated by immunosurgery. The seven HESC lines exhibited morphology and markers typical of HESCs and the capacity for long-term proliferation. The derived HESC lines manifested pluripotent differentiation potential both in vivo and in vitro. Surprisingly, karyotype analysis of the HESC lines that were derived from these aneuploid embryos showed that the cell lines carry a normal euploid karyotype. We show that the euploidy was not achieved through chromosome duplication. Alternatively, we suggest that the euploid HESC lines originated from mosaic embryos consisting of aneuploid and euploid cells, and in vitro selection occurred to favor euploid cells. We assume that aneuploid HESC lines could be isolated mostly from embryos that are uniform for the aneuploidy. These results led us to conclude that the aneuploid mosaic embryos that are destined to be discarded can serve as an alternative source for normal euploid HESC lines.


Asunto(s)
Aneuploidia , Técnicas de Cultivo de Embriones , Células Madre Embrionarias/citología , Animales , Diferenciación Celular , Proliferación Celular , Células Cultivadas , Femenino , Fertilización In Vitro , Humanos , Cariotipificación , Ratones , Fenotipo , Embarazo , Diagnóstico Preimplantación
12.
Cell Rep ; 26(10): 2531-2539.e4, 2019 03 05.
Artículo en Inglés | MEDLINE | ID: mdl-30840878

RESUMEN

Fragile X syndrome (FXS) is caused primarily by a CGG repeat expansion in the FMR1 gene that triggers its transcriptional silencing. In order to investigate the regulatory layers involved in FMR1 inactivation, we tested a collection of chromatin modulators for the ability to reactivate the FMR1 locus. Although inhibitors of DNA methyltransferase (DNMT) induced the highest levels of FMR1 expression, a combination of a DNMT inhibitor and another compound potentiated the effect of reactivating treatment. To better assess the rescue effect following direct demethylation, we characterized the long-term and genome-wide effects of FMR1 reactivation and established an in vivo system to analyze FMR1-reactivating therapies. Systemic treatment with a DNMT inhibitor in mice carrying FXS induced pluripotent stem cell (iPSC)-derived transplants robustly induced FMR1 expression in the affected tissue, which was maintained for a prolonged period of time. Finally, we show a proof of principle for FMR1-reactivating therapy in the context of the CNS.


Asunto(s)
Inhibidores Enzimáticos/farmacología , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/metabolismo , Síndrome del Cromosoma X Frágil/tratamiento farmacológico , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Células Madre Pluripotentes Inducidas/metabolismo , Animales , Metilasas de Modificación del ADN/antagonistas & inhibidores , Metilasas de Modificación del ADN/metabolismo , Femenino , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/genética , Síndrome del Cromosoma X Frágil/genética , Síndrome del Cromosoma X Frágil/metabolismo , Síndrome del Cromosoma X Frágil/patología , Humanos , Células Madre Pluripotentes Inducidas/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Ratones SCID , Células-Madre Neurales
13.
iScience ; 11: 398-408, 2019 Jan 25.
Artículo en Inglés | MEDLINE | ID: mdl-30660107

RESUMEN

Human pluripotent stem cells (hPSCs) acquire genetic changes during their propagation in culture that can affect their use in research and future therapies. To identify the key genes involved in selective advantage during culture adaptation and tumorigenicity of hPSCs, we generated a genome-wide screening system for genes and pathways that provide a growth advantage either in vitro or in vivo. We found that hyperactivation of the RAS pathway confers resistance to selection with the hPSC-specific drug PluriSIn-1. We also identified that inactivation of the RHO-ROCK pathway gives growth advantage during culture adaptation. Last, we demonstrated the importance of the PI3K-AKT and HIPPO pathways for the teratoma formation process. Our screen revealed key genes and pathways relevant to the tumorigenicity and survival of hPSCs and should thus assist in understanding and confronting their tumorigenic potential.

14.
Cell Stem Cell ; 25(3): 419-432.e9, 2019 09 05.
Artículo en Inglés | MEDLINE | ID: mdl-31491396

RESUMEN

Genomic imprinting is an epigenetic mechanism that results in parent-of-origin monoallelic expression of specific genes, which precludes uniparental development and underlies various diseases. Here, we explored molecular and developmental aspects of imprinting in humans by generating exclusively paternal human androgenetic embryonic stem cells (aESCs) and comparing them with exclusively maternal parthenogenetic ESCs (pESCs) and bi-parental ESCs, establishing a pluripotent cell system of distinct parental backgrounds. Analyzing the transcriptomes and methylomes of human aESCs, pESCs, and bi-parental ESCs enabled the characterization of regulatory relations at known imprinted regions and uncovered imprinted gene candidates within and outside known imprinted regions. Investigating the consequences of uniparental differentiation, we showed the known paternal-genome preference for placental contribution, revealed a similar bias toward liver differentiation, and implicated the involvement of the imprinted gene IGF2 in this process. Our results demonstrate the utility of parent-specific human ESCs for dissecting the role of imprinting in human development and disease.


Asunto(s)
Células Madre Embrionarias/fisiología , Partenogénesis/fisiología , Células Madre Pluripotentes/fisiología , Caracteres Sexuales , Diferenciación Celular , Células Cultivadas , Metilación de ADN , Epigénesis Genética , Femenino , Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica , Impresión Genómica , Humanos , Factor II del Crecimiento Similar a la Insulina/genética , Masculino , Padres , Transcriptoma
15.
Stem Cell Reports ; 7(4): 777-786, 2016 10 11.
Artículo en Inglés | MEDLINE | ID: mdl-27618722

RESUMEN

Down syndrome (DS) is the leading genetic cause of mental retardation and is caused by a third copy of human chromosome 21. The different pathologies of DS involve many tissues with a distinct array of neural phenotypes. Here we characterize embryonic stem cell lines with DS (DS-ESCs), and focus on the neural aspects of the disease. Our results show that neural progenitor cells (NPCs) differentiated from five independent DS-ESC lines display increased apoptosis and downregulation of forehead developmental genes. Analysis of differentially expressed genes suggested RUNX1 as a key transcription regulator in DS-NPCs. Using genome editing we were able to disrupt all three copies of RUNX1 in DS-ESCs, leading to downregulation of several RUNX1 target developmental genes accompanied by reduced apoptosis and neuron migration. Our work sheds light on the role of RUNX1 and the importance of dosage balance in the development of neural phenotypes in DS.


Asunto(s)
Diferenciación Celular , Subunidad alfa 2 del Factor de Unión al Sitio Principal/genética , Síndrome de Down/genética , Síndrome de Down/metabolismo , Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Células-Madre Neurales/citología , Células-Madre Neurales/metabolismo , Neurogénesis , Apoptosis/genética , Diferenciación Celular/genética , Línea Celular , Células Cultivadas , Subunidad alfa 2 del Factor de Unión al Sitio Principal/metabolismo , Dosificación de Gen , Edición Génica , Humanos , Cariotipo , Neurogénesis/genética , Fenotipo
16.
Cell Stem Cell ; 18(2): 253-61, 2016 Feb 04.
Artículo en Inglés | MEDLINE | ID: mdl-26669899

RESUMEN

Human pluripotent stem cells (hPSCs) frequently acquire chromosomal aberrations such as aneuploidy in culture. These aberrations progressively increase over time and may compromise the properties and clinical utility of the cells. The underlying mechanisms that drive initial genomic instability and its continued progression are largely unknown. Here, we show that aneuploid hPSCs undergo DNA replication stress, resulting in defective chromosome condensation and segregation. Aneuploid hPSCs show altered levels of actin cytoskeletal genes controlled by the transcription factor SRF, and overexpression of SRF rescues impaired chromosome condensation and segregation defects in aneuploid hPSCs. Furthermore, SRF downregulation in diploid hPSCs induces replication stress and perturbed condensation similar to that seen in aneuploid cells. Together, these results suggest that decreased SRF expression induces replicative stress and chromosomal condensation defects that underlie the ongoing chromosomal instability seen in aneuploid hPSCs. A similar mechanism may also operate during initiation of instability in diploid cells.


Asunto(s)
Aberraciones Cromosómicas , Replicación del ADN , Inestabilidad Genómica , Células Madre Pluripotentes/metabolismo , Estrés Fisiológico , Citoesqueleto de Actina/genética , Actinas/metabolismo , Anafase , Aneuploidia , Segregación Cromosómica/genética , Replicación del ADN/genética , Diploidia , Regulación hacia Abajo/genética , Humanos , Metafase , Factor de Respuesta Sérica/genética , Factor de Respuesta Sérica/metabolismo , Estrés Fisiológico/genética , Transcripción Genética
17.
Nat Commun ; 6: 7094, 2015 May 11.
Artículo en Inglés | MEDLINE | ID: mdl-25959793

RESUMEN

Recurrent genomic instability in cancer is attributed to positive selection and/or the sensitivity of specific genomic regions to breakage. Among these regions are fragile sites (FSs), genomic regions sensitive to replication stress conditions induced by the DNA polymerase inhibitor aphidicolin. However, the basis for the majority of cancer genomic instability hotspots remains unclear. Aberrant oncogene expression induces replication stress, leading to DNA breaks and genomic instability. Here we map the cytogenetic locations of oncogene-induced FSs and show that in the same cells, each oncogene creates a unique fragility landscape that only partially overlaps with aphidicolin-induced FSs. Oncogene-induced FSs colocalize with cancer breakpoints and large genes, similar to aphidicolin-induced FSs. The observed plasticity in the fragility landscape of the same cell type following oncogene expression highlights an additional level of complexity in the molecular basis for recurrent fragility in cancer.


Asunto(s)
Sitios Frágiles del Cromosoma/fisiología , Fragilidad Cromosómica/fisiología , Fibroblastos/fisiología , Inestabilidad Genómica , Oncogenes/fisiología , Línea Celular Tumoral , Sitios Frágiles del Cromosoma/genética , Fragilidad Cromosómica/genética , Eliminación de Gen , Regulación de la Expresión Génica/fisiología , Humanos , Familia de Multigenes , Oncogenes/genética , Plásmidos
18.
Nat Commun ; 5: 4825, 2014 Sep 08.
Artículo en Inglés | MEDLINE | ID: mdl-25198699

RESUMEN

Human pluripotent stem cells (hPSCs) tend to acquire genomic aberrations in culture, the most common of which is trisomy of chromosome 12. Here we dissect the cellular and molecular implications of this trisomy in hPSCs. Global gene expression analyses reveal that trisomy 12 profoundly affects the gene expression profile of hPSCs, inducing a transcriptional programme similar to that of germ cell tumours. Comparison of proliferation, differentiation and apoptosis between diploid and aneuploid hPSCs shows that trisomy 12 significantly increases the proliferation rate of hPSCs, mainly as a consequence of increased replication. Furthermore, trisomy 12 increases the tumorigenicity of hPSCs in vivo, inducing transcriptionally distinct teratomas from which pluripotent cells can be recovered. Last, a chemical screen of 89 anticancer drugs discovers that trisomy 12 raises the sensitivity of hPSCs to several replication inhibitors. Together, these findings demonstrate the extensive effect of trisomy 12 and highlight its perils for successful hPSC applications.


Asunto(s)
Carcinogénesis/genética , Proliferación Celular/genética , Cromosomas Humanos Par 12/genética , Regulación Neoplásica de la Expresión Génica/genética , Neoplasias/genética , Células Madre Pluripotentes/metabolismo , ARN Mensajero/metabolismo , Trisomía/genética , Aneuploidia , Técnicas de Cultivo de Célula , Línea Celular Tumoral , Células Madre de Carcinoma Embrionario/metabolismo , Expresión Génica/genética , Perfilación de la Expresión Génica , Humanos , Técnicas In Vitro , Neoplasias de Células Germinales y Embrionarias/genética , Teratocarcinoma/genética , Teratoma/genética
19.
Cell Stem Cell ; 15(5): 634-42, 2014 Nov 06.
Artículo en Inglés | MEDLINE | ID: mdl-25517467

RESUMEN

The recent finding that reprogrammed human pluripotent stem cells can be derived by nuclear transfer into human oocytes as well as by induced expression of defined factors has revitalized the debate on whether one approach might be advantageous over the other. Here we compare the genetic and epigenetic integrity of human nuclear-transfer embryonic stem cell (NT-ESC) lines and isogenic induced pluripotent stem cell (iPSC) lines, derived from the same somatic cell cultures of fetal, neonatal, and adult origin. The two cell types showed similar genome-wide gene expression and DNA methylation profiles. Importantly, NT-ESCs and iPSCs had comparable numbers of de novo coding mutations, but significantly more than parthenogenetic ESCs. As iPSCs, NT-ESCs displayed clone- and gene-specific aberrations in DNA methylation and allele-specific expression of imprinted genes. The occurrence of these genetic and epigenetic defects in both NT-ESCs and iPSCs suggests that they are inherent to reprogramming, regardless of derivation approach.


Asunto(s)
Impresión Genómica/genética , Tasa de Mutación , Técnicas de Transferencia Nuclear , Sistemas de Lectura Abierta/genética , Células Madre Pluripotentes/citología , Adulto , Secuencia de Bases , Células Clonales , Metilación de ADN/genética , Regulación de la Expresión Génica , Sitios Genéticos , Humanos , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/metabolismo , Masculino , Datos de Secuencia Molecular , Células Madre Pluripotentes/metabolismo
20.
Cell Stem Cell ; 12(2): 167-79, 2013 Feb 07.
Artículo en Inglés | MEDLINE | ID: mdl-23318055

RESUMEN

The use of human pluripotent stem cells (hPSCs) in cell therapy is hindered by the tumorigenic risk from residual undifferentiated cells. Here we performed a high-throughput screen of over 52,000 small molecules and identified 15 pluripotent cell-specific inhibitors (PluriSIns), nine of which share a common structural moiety. The PluriSIns selectively eliminated hPSCs while sparing a large array of progenitor and differentiated cells. Cellular and molecular analyses demonstrated that the most selective compound, PluriSIn #1, induces ER stress, protein synthesis attenuation, and apoptosis in hPSCs. Close examination identified this molecule as an inhibitor of stearoyl-coA desaturase (SCD1), the key enzyme in oleic acid biosynthesis, revealing a unique role for lipid metabolism in hPSCs. PluriSIn #1 was also cytotoxic to mouse blastocysts, indicating that the dependence on oleate is inherent to the pluripotent state. Finally, application of PluriSIn #1 prevented teratoma formation from tumorigenic undifferentiated cells. These findings should increase the safety of hPSC-based treatments.


Asunto(s)
Inhibidores Enzimáticos/farmacología , Ácido Oléico/síntesis química , Células Madre Pluripotentes/efectos de los fármacos , Estearoil-CoA Desaturasa/antagonistas & inhibidores , Animales , Blastocisto/citología , Blastocisto/efectos de los fármacos , Blastocisto/metabolismo , Células Cultivadas , Humanos , Ratones , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA