Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
Mol Biol Cell ; 17(8): 3543-56, 2006 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16760426

RESUMEN

Mesenchymal stem cells from adipose tissue can differentiate into mesodermal lineages. Differentiation potential, however, varies between clones of adipose stem cells (ASCs), raising the hypothesis that epigenetic differences account for this variability. We report here a bisulfite sequencing analysis of CpG methylation of adipogenic (leptin [LEP], peroxisome proliferator-activated receptor gamma 2 [PPARG2], fatty acid-binding protein 4 [FABP4], and lipoprotein lipase [LPL]) promoters and of nonadipogenic (myogenin [MYOG], CD31, and GAPDH) loci in freshly isolated human ASCs and in cultured ASCs, in relation to gene expression and differentiation potential. Uncultured ASCs display hypomethylated adipogenic promoters, in contrast to myogenic and endothelial loci, which are methylated. Adipogenic promoters exhibit mosaic CpG methylation, on the basis of heterogeneous methylation between cells and of variation in the extent of methylation of a given CpG between donors, and both between and within clonal cell lines. DNA methylation reflects neither transcriptional status nor potential for gene expression upon differentiation. ASC culture preserves hypomethylation of adipogenic promoters; however, between- and within-clone mosaic methylation is detected. Adipogenic differentiation also maintains the overall CpG hypomethylation of LEP, PPARG2, FABP4, and LPL despite demethylation of specific CpGs and transcriptional induction. Furthermore, enhanced methylation at adipogenic loci in primary differentiated cells unrelated to adipogenesis argues for ASC specificity of the hypomethylated state of these loci. Therefore, mosaic hypomethylation of adipogenic promoters may constitute a molecular signature of ASCs, and DNA methylation does not seem to be a determinant of differentiation potential of these cells.


Asunto(s)
Adipogénesis/genética , Tejido Adiposo/citología , Islas de CpG/genética , Metilación de ADN , Células Madre Mesenquimatosas/citología , Regiones Promotoras Genéticas/genética , Linaje de la Célula , Separación Celular , Células Cultivadas , Células Clonales , Femenino , Regulación de la Expresión Génica , Humanos , Leptina/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Análisis de Secuencia de ADN
2.
Mol Biol Cell ; 16(12): 5719-35, 2005 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16195347

RESUMEN

Functional reprogramming of a differentiated cell toward pluripotency may have long-term applications in regenerative medicine. We report the induction of dedifferentiation, associated with genomewide programming of gene expression and epigenetic reprogramming of an embryonic gene, in epithelial 293T cells treated with an extract of undifferentiated human NCCIT carcinoma cells. 293T cells exposed for 1 h to extract of NCCIT cells, but not of 293T or Jurkat T-cells, form defined colonies that are maintained for at least 23 passages in culture. Microarray and quantitative analyses of gene expression reveal that the transition from a 293T to a pluripotent cell phenotype involves a dynamic up-regulation of hundreds of NCCIT genes, concomitant with down-regulation of 293T genes and of indicators of differentiation such as A-type lamins. Up-regulated genes encompass embryonic and stem cell markers, including OCT4, SOX2, NANOG, and Oct4-responsive genes. OCT4 activation is associated with DNA demethylation in the OCT4 promoter and nuclear targeting of Oct4 protein. In fibroblasts exposed to extract of mouse embryonic stem cells, Oct4 activation is biphasic and RNA-PolII dependent, with the first transient rise of Oct4 up-regulation being necessary for the second, long-term activation of Oct4. Genes characteristic of multilineage differentiation potential are also up-regulated in NCCIT extract-treated cells, suggesting the establishment of "multilineage priming." Retinoic acid triggers Oct4 down-regulation, de novo activation of A-type lamins, and nestin. Furthermore, the cells can be induced to differentiate toward neurogenic, adipogenic, osteogenic, and endothelial lineages. The data provide a proof-of-concept that an extract of undifferentiated carcinoma cells can elicit differentiation plasticity in an otherwise more developmentally restricted cell type.


Asunto(s)
Carcinoma/genética , Regulación de la Expresión Génica/fisiología , Células Madre/citología , Células Madre/fisiología , Transcripción Genética , Diferenciación Celular , Línea Celular , Línea Celular Tumoral , Daño del ADN , Genoma Humano , Humanos , Células Jurkat , Mesodermo/citología , Mesodermo/fisiología , Neuronas/citología , Neuronas/fisiología , Reacción en Cadena de la Polimerasa , Extractos de Tejidos/fisiología
3.
Transfus Med Hemother ; 35(3): 205-215, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-21547118

RESUMEN

SUMMARY: Stem cells have the ability to self-renew, and give rise to one or more differentiated cell types. Embryonic stem cells can differentiate into all cell types of the body and have unlimited self-renewal capacity. Somatic stem cells are found in many adult tissues. They have an extensive but finite lifespan and can differentiate into a more restricted range of cell types. Increasing evidence indicates that the multilineage differentiation ability of stem cells is defined by the potential for expression of developmentally regulated transcription factors and of lineage specification genes. Gene expression, or as emphasized here, the potential for gene expression, is largely controlled by epigenetic modifications of DNA (DNA methylation) and chromatin (such as post-translational histone modifications) in the regulatory regions of specific genes. Epigenetic modifications can also influence the timing of DNA replication. We highlight here how mechanisms by which genes are poised for transcription in undifferentiated stem cells are being uncovered through the mapping of DNA methylation profiles on differentiation-regulated promoters and at the genome-wide level, histone modifications, and transcription factor binding. Epigenetic marks on developmentally regulated and lineage specification genes in stem cells seem to define a state of pluripotency.

4.
Front Cell Dev Biol ; 6: 73, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30057899

RESUMEN

At the nuclear periphery, the genome is anchored to A- and B-type nuclear lamins in the form of heterochromatic lamina-associated domains. A-type lamins also associate with chromatin in the nuclear interior, away from the peripheral nuclear lamina. This nucleoplasmic lamin A environment tends to be euchromatic, suggesting distinct roles of lamin A in the regulation of gene expression in peripheral and more central regions of the nucleus. The hot-spot lamin A R482W mutation causing familial partial lipodystrophy of Dunnigan-type (FPLD2), affects lamin A association with chromatin at the nuclear periphery and in the nuclear interior, and is associated with 3-dimensional (3D) rearrangements of chromatin. Here, we highlight features of nuclear lamin association with the genome at the nuclear periphery and in the nuclear interior. We address recent data showing a rewiring of such interactions in cells from FPLD2 patients, and in adipose progenitor and induced pluripotent stem cell models of FPLD2. We discuss associated epigenetic and genome conformation changes elicited by the lamin A R482W mutation at the gene level. The findings argue that the mutation adversely impacts both global and local genome architecture throughout the nucleus space. The results, together with emerging new computational modeling tools, mark the start of a new era in our understanding of the 3D genomics of laminopathies.

5.
J Cell Biol ; 216(9): 2731-2743, 2017 09 04.
Artículo en Inglés | MEDLINE | ID: mdl-28751304

RESUMEN

Mutations in the Lamin A/C (LMNA) gene-encoding nuclear LMNA cause laminopathies, which include partial lipodystrophies associated with metabolic syndromes. The lipodystrophy-associated LMNA p.R482W mutation is known to impair adipogenic differentiation, but the mechanisms involved are unclear. We show in this study that the lamin A p.R482W hot spot mutation prevents adipogenic gene expression by epigenetically deregulating long-range enhancers of the anti-adipogenic MIR335 microRNA gene in human adipocyte progenitor cells. The R482W mutation results in a loss of function of differentiation-dependent lamin A binding to the MIR335 locus. This impairs H3K27 methylation and instead favors H3K27 acetylation on MIR335 enhancers. The lamin A mutation further promotes spatial clustering of MIR335 enhancer and promoter elements along with overexpression of the MIR355 gene after adipogenic induction. Our results link a laminopathy-causing lamin A mutation to an unsuspected deregulation of chromatin states and spatial conformation of an miRNA locus critical for adipose progenitor cell fate.


Asunto(s)
Adipocitos , Adipogénesis/genética , Epigénesis Genética , Fibroblastos , Lamina Tipo A/genética , Lipodistrofia Parcial Familiar/genética , MicroARNs/genética , Mutación , Células Madre , Acetilación , Adipocitos/metabolismo , Adipocitos/patología , Células Cultivadas , Cromatina/genética , Cromatina/metabolismo , Ensamble y Desensamble de Cromatina , Fibroblastos/metabolismo , Fibroblastos/patología , Predisposición Genética a la Enfermedad , Histonas/metabolismo , Humanos , Lamina Tipo A/metabolismo , Lipodistrofia Parcial Familiar/metabolismo , Lipodistrofia Parcial Familiar/patología , Lipodistrofia Parcial Familiar/fisiopatología , Metilación , MicroARNs/química , MicroARNs/metabolismo , Conformación de Ácido Nucleico , Fenotipo , Regiones Promotoras Genéticas , Células Madre/metabolismo , Células Madre/patología , Relación Estructura-Actividad , Regulación hacia Arriba
6.
Genome Biol ; 18(1): 21, 2017 01 30.
Artículo en Inglés | MEDLINE | ID: mdl-28137286

RESUMEN

Current three-dimensional (3D) genome modeling platforms are limited by their inability to account for radial placement of loci in the nucleus. We present Chrom3D, a user-friendly whole-genome 3D computational modeling framework that simulates positions of topologically-associated domains (TADs) relative to each other and to the nuclear periphery. Chrom3D integrates chromosome conformation capture (Hi-C) and lamin-associated domain (LAD) datasets to generate structure ensembles that recapitulate radial distributions of TADs detected in single cells. Chrom3D reveals unexpected spatial features of LAD regulation in cells from patients with a laminopathy-causing lamin mutation. Chrom3D is freely available on github.


Asunto(s)
Cromatina/genética , Biología Computacional/métodos , Lámina Nuclear/genética , Adulto , Femenino , Genoma , Células HeLa , Humanos , Masculino , Modelos Genéticos , Adulto Joven
7.
Oncotarget ; 6(29): 26729-45, 2015 Sep 29.
Artículo en Inglés | MEDLINE | ID: mdl-26353929

RESUMEN

BACKGROUND & AIMS: Liver fibrogenesis - scarring of the liver that can lead to cirrhosis and liver cancer - is characterized by hepatocyte impairment, capillarization of liver sinusoidal endothelial cells (LSECs) and hepatic stellate cell (HSC) activation. To date, the molecular determinants of a healthy human liver cell phenotype remain largely uncharacterized. Here, we assess the transcriptome and the genome-wide promoter methylome specific for purified, non-cultured human hepatocytes, LSECs and HSCs, and investigate the nature of epigenetic changes accompanying transcriptional changes associated with activation of HSCs. MATERIAL AND METHODS: Gene expression profile and promoter methylome of purified, uncultured human liver cells and culture-activated HSCs were respectively determined using Affymetrix HG-U219 genechips and by methylated DNA immunoprecipitation coupled to promoter array hybridization. Histone modification patterns were assessed at the single-gene level by chromatin immunoprecipitation and quantitative PCR. RESULTS: We unveil a DNA-methylation-based epigenetic relationship between hepatocytes, LSECs and HSCs despite their distinct ontogeny. We show that liver cell type-specific DNA methylation targets early developmental and differentiation-associated functions. Integrative analysis of promoter methylome and transcriptome reveals partial concordance between DNA methylation and transcriptional changes associated with human HSC activation. Further, we identify concordant histone methylation and acetylation changes in the promoter and putative novel enhancer elements of genes involved in liver fibrosis. CONCLUSIONS: Our study provides the first epigenetic blueprint of three distinct freshly isolated, human hepatic cell types and of epigenetic changes elicited upon HSC activation.


Asunto(s)
Metilación de ADN , Perfilación de la Expresión Génica , Estudio de Asociación del Genoma Completo , Células Estrelladas Hepáticas/citología , Hígado/citología , Adolescente , Anciano , Animales , Células Cultivadas , Niño , Inmunoprecipitación de Cromatina , Epigénesis Genética , Femenino , Hepatocitos/citología , Humanos , Lactante , Recién Nacido , Cirrosis Hepática/patología , Masculino , Ratones , Ratones Endogámicos BALB C , Análisis de Secuencia por Matrices de Oligonucleótidos , Fenotipo , Regiones Promotoras Genéticas , Transcripción Genética , Transcriptoma
8.
Mol Biol Cell ; 21(12): 2066-77, 2010 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-20410135

RESUMEN

Mesenchymal stem cells (MSCs) isolated from various tissues share common phenotypic and functional properties. However, intrinsic molecular evidence supporting these observations has been lacking. Here, we unravel overlapping genome-wide promoter DNA methylation patterns between MSCs from adipose tissue, bone marrow, and skeletal muscle, whereas hematopoietic progenitors are more epigenetically distant from MSCs as a whole. Commonly hypermethylated genes are enriched in signaling, metabolic, and developmental functions, whereas genes hypermethylated only in MSCs are associated with early development functions. We find that most lineage-specification promoters are DNA hypomethylated and harbor a combination of trimethylated H3K4 and H3K27, whereas early developmental genes are DNA hypermethylated with or without H3K27 methylation. Promoter DNA methylation patterns of differentiated cells are largely established at the progenitor stage; yet, differentiation segregates a minor fraction of the commonly hypermethylated promoters, generating greater epigenetic divergence between differentiated cell types than between their undifferentiated counterparts. We also show an effect of promoter CpG content on methylation dynamics upon differentiation and distinct methylation profiles on transcriptionally active and inactive promoters. We infer that methylation state of lineage-specific promoters in MSCs is not a primary determinant of differentiation capacity. Our results support the view of a common origin of mesenchymal progenitors.


Asunto(s)
Diferenciación Celular/genética , Reprogramación Celular/genética , Metilación de ADN/genética , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Regiones Promotoras Genéticas , Tejido Adiposo/citología , Células de la Médula Ósea/citología , Linaje de la Célula/genética , Cromatina/genética , Inmunoprecipitación de Cromatina , Islas de CpG/genética , Epigénesis Genética , Regulación del Desarrollo de la Expresión Génica , Histonas/metabolismo , Humanos , Lisina/metabolismo , Músculos/citología , Análisis de Secuencia por Matrices de Oligonucleótidos , Transcripción Genética
9.
Mol Biol Cell ; 21(11): 1872-84, 2010 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-20375147

RESUMEN

In contrast to canonical histones, histone variant H3.3 is incorporated into chromatin in a replication-independent manner. Posttranslational modifications of H3.3 have been identified; however, the epigenetic environment of incorporated H3.3 is unclear. We have investigated the genomic distribution of epitope-tagged H3.3 in relation to histone modifications, DNA methylation, and transcription in mesenchymal stem cells. Quantitative imaging at the nucleus level shows that H3.3, relative to replicative H3.2 or canonical H2B, is enriched in chromatin domains marked by histone modifications of active or potentially active genes. Chromatin immunoprecipitation of epitope-tagged H3.3 and array hybridization identified 1649 H3.3-enriched promoters, a fraction of which is coenriched in H3K4me3 alone or together with H3K27me3, whereas H3K9me3 is excluded, corroborating nucleus-level imaging data. H3.3-enriched promoters are predominantly CpG-rich and preferentially DNA methylated, relative to the proportion of methylated RefSeq promoters in the genome. Most but not all H3.3-enriched promoters are transcriptionally active, and coenrichment of H3.3 with repressive H3K27me3 correlates with an enhanced proportion of expressed genes carrying this mark. H3.3-target genes are enriched in mesodermal differentiation and signaling functions. Our data suggest that in mesenchymal stem cells, H3.3 targets lineage-priming genes with a potential for activation facilitated by H3K4me3 in facultative association with H3K27me3.


Asunto(s)
Inmunoprecipitación de Cromatina/métodos , Cromatina/química , ADN/metabolismo , Genoma , Histonas/química , ADN/genética , Metilación de ADN , Regulación de la Expresión Génica , Histonas/genética , Histonas/metabolismo , Humanos , Análisis por Micromatrices , Regiones Promotoras Genéticas , Procesamiento Proteico-Postraduccional , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo
10.
Stem Cells Dev ; 19(8): 1257-66, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19886822

RESUMEN

Mesenchymal stem cells (MSCs) can differentiate into multiple mesodermal cell types in vitro; however, their differentiation capacity is influenced by their tissue of origin. To what extent epigenetic information on promoters of lineage-specification genes in human progenitors influences transcriptional activation and differentiation potential remains unclear. We produced bisulfite sequencing maps of DNA methylation in adipogenic, myogenic, and endothelial promoters in relation to gene expression and differentiation capacity, and unravel a similarity in DNA methylation profiles between MSCs isolated from human adipose tissue, bone marrow (BM), and muscle. This similarity is irrespective of promoter CpG content. Methylation patterns of MSCs are distinct from those of hematopoietic progenitor cells (HPCs), pluripotent human embryonic stem cells (hESCs), and multipotent hESC-derived mesenchymal cells (MCs). Moreover, in vitro MSC differentiation does not affect lineage-specific promoter methylation states, arguing that these methylation patterns in differentiated cells are already established at the progenitor stage. Further, we find a correlation between lineage-specific promoter hypermethylation and lack of differentiation capacity toward that lineage, but no relationship between weak promoter methylation and capacity of transcriptional activation or differentiation. Thus, only part of the restriction in differentiation capacity of tissue-specific stem cells is programmed by promoter DNA methylation: hypermethylation seems to constitute a barrier to differentiation, however, no or weak methylation has no predictive value for differentiation potential.


Asunto(s)
Células Madre Adultas/citología , Células Madre Adultas/metabolismo , Linaje de la Célula/fisiología , Metilación de ADN , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Regiones Promotoras Genéticas/genética , Adipocitos/citología , Adipocitos/metabolismo , Tejido Adiposo/citología , Células de la Médula Ósea/citología , Células de la Médula Ósea/metabolismo , Diferenciación Celular/fisiología , Línea Celular , Islas de CpG/genética , Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Expresión Génica/genética , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Humanos , Células Musculares/citología , Células Musculares/metabolismo , Músculo Esquelético/citología , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/genética
11.
Methods Mol Biol ; 567: 249-62, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19588097

RESUMEN

DNA methylation contributes to the regulation of long-term gene repression by enabling the recruitment of transcriptional repressor complexes to methylated cytosines. Several methods for detecting DNA methylation at the gene-specific and genome-wide levels have been developed. Methylated DNA immunoprecipitation, or MeDIP, consists of the selective immunoprecipitation of methylated DNA fragments using antibodies to 5-methylcytosine. The genomic site of interest can be detected by PCR, hybridization to DNA arrays, or by direct sequencing. This chapter describes the MeDIP protocol and quality control tests that should be performed throughout the procedure.


Asunto(s)
Inmunoprecipitación de Cromatina/métodos , Metilación de ADN , ADN/metabolismo , Animales , ADN/análisis , ADN/aislamiento & purificación , Metilación de ADN/fisiología , Humanos , Modelos Biológicos , Técnicas de Amplificación de Ácido Nucleico/métodos , Análisis de Secuencia por Matrices de Oligonucleótidos/métodos
12.
Stem Cells ; 25(4): 852-61, 2007 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17170064

RESUMEN

In vivo endothelial commitment of adipose stem cells (ASCs) has scarcely been reported, and controversy remains on the contribution of ASCs to vascularization. We address the epigenetic commitment of ASCs to the endothelial lineage. We report a bisulfite sequencing analysis of CpG methylation in the promoters of two endothelial-cell-specific genes, CD31 and CD144, in freshly isolated and in cultures of ASCs before and after induction of endothelial differentiation. In contrast to adipose tissue-derived endothelial (CD31(+)) cells, freshly isolated ASCs display a heavily methylated CD31 promoter and a mosaically methylated CD144 promoter despite basal transcription of both genes. Methylation state of both promoters remains globally stable upon culture. Endothelial stimulation of ASCs in methylcellulose elicits phenotypic changes, marginal upregulation of CD31, and CD144 expression and restrictive induction of a CD31(+)CD144(+) immunophenotype. These events are accompanied by discrete changes in CpG methylation in CD31 and CD144 promoters; however, no global demethylation that marks CD31(+) cells and human umbilical vein endothelial cells occurs. Immunoselection of CD31(+) cells after endothelial stimulation reveals consistent demethylation of one CpG immediately 3' of the transcription start site of the CD31 promoter. Adipogenic or osteogenic differentiation maintains CD31 and CD144 methylation patterns of undifferentiated cells. Methylation profiles of CD31 and CD144 promoters suggest a limited commitment of ASCs to the endothelial lineage. This contrasts with the reported hypomethylation of adipogenic promoters, which reflects a propensity of ASCs toward adipogenic differentiation. Analysis of CpG methylation at lineage-specific promoters provides a robust assessment of epigenetic commitment of stem cells to a specific lineage.


Asunto(s)
Tejido Adiposo/citología , Diferenciación Celular/fisiología , Fosfatos de Dinucleósidos/genética , Endotelio Vascular/citología , Regiones Promotoras Genéticas , Células Madre/citología , Células Madre/fisiología , Tejido Adiposo/fisiología , Antígenos CD , Humanos , Inmunofenotipificación , Metilación , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/análisis , Venas Umbilicales
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA