Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 3 de 3
Filter
Add more filters










Database
Language
Publication year range
1.
Stem Cells Dev ; 19(8): 1257-66, 2010 Aug.
Article in English | MEDLINE | ID: mdl-19886822

ABSTRACT

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.


Subject(s)
Adult Stem Cells/cytology , Adult Stem Cells/metabolism , Cell Lineage/physiology , DNA Methylation , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Promoter Regions, Genetic/genetics , Adipocytes/cytology , Adipocytes/metabolism , Adipose Tissue/cytology , Bone Marrow Cells/cytology , Bone Marrow Cells/metabolism , Cell Differentiation/physiology , Cell Line , CpG Islands/genetics , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Gene Expression/genetics , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Humans , Muscle Cells/cytology , Muscle Cells/metabolism , Muscle, Skeletal/cytology , Platelet Endothelial Cell Adhesion Molecule-1/genetics
2.
J Cell Mol Med ; 11(4): 602-20, 2007.
Article in English | MEDLINE | ID: mdl-17760828

ABSTRACT

In opposition to terminally differentiated cells, stem cells can self-renew and give rise to multiple cell types. Embryonic stem cells retain the ability of the inner cell mass of blastocysts to differentiate into all cell types of the body and have acquired in culture unlimited self-renewal capacity. Somatic stem cells are found in many adult tissues, have an extensive but finite lifespan and can differentiate into a more restricted array of cell types. A growing body of evidence indicates that multi-lineage differentiation ability of stem cells can be defined by the potential for expression of lineage-specification genes. Gene expression, or as emphasized here, potential for gene expression, is largely controlled by epigenetic modifications of DNA and chromatin on genomic regulatory and coding regions. These modifications modulate chromatin organization not only on specific genes but also at the level of the whole nucleus; they can also affect timing of DNA replication. This review highlights how mechanisms by which genes are poised for transcription in undifferentiated stem cells are being uncovered through primarily the mapping of DNA methylation, histone modifications and transcription factor binding throughout the genome. The combinatorial association of epigenetic marks on developmentally regulated and lineage-specifying genes in undifferentiated cells seems to define a pluripotent state.


Subject(s)
Embryonic Stem Cells/metabolism , Epigenesis, Genetic , Genome , Stem Cells/metabolism , Amino Acid Sequence , Animals , Chromatin/metabolism , DNA Methylation , Embryonic Stem Cells/cytology , Humans , Molecular Sequence Data , Stem Cells/cytology
3.
Mol Biol Cell ; 18(5): 1543-53, 2007 May.
Article in English | MEDLINE | ID: mdl-17314394

ABSTRACT

Analyses of molecular events associated with reprogramming somatic nuclei to pluripotency are scarce. We previously reported the reprogramming of epithelial cells by extract of undifferentiated embryonal carcinoma (EC) cells. We now demonstrate reprogramming of DNA methylation and histone modifications on regulatory regions of the developmentally regulated OCT4 and NANOG genes by exposure of 293T cells to EC cell extract. OCT4 and NANOG are transcriptionally up-regulated and undergo mosaic cytosine-phosphate-guanosine demethylation. OCT4 demethylation occurs as early as week 1, is enhanced by week 2, and is most prominent in the proximal promoter and distal enhancer. Targeted OCT4 and NANOG demethylation does not occur in 293T extract-treated cells. Retinoic acid-mediated differentiation of reprogrammed cells elicits OCT4 promoter remethylation and transcriptional repression. Chromatin immunoprecipitation analyses of lysines K4, K9, and K27 of histone H3 on OCT4 and NANOG indicate that primary chromatin remodeling determinants are acetylation of H3K9 and demethylation of dimethylated H3K9. H3K4 remains di- and trimethylated. Demethylation of trimethylated H3K9 and H3K27 also occurs; however, trimethylation seems more stable than dimethylation. We conclude that a central epigenetic reprogramming event is relaxation of chromatin at loci associated with pluripotency to create a conformation compatible with transcriptional activation.


Subject(s)
Carcinoma, Embryonal/genetics , DNA-Binding Proteins/genetics , Homeodomain Proteins/genetics , Octamer Transcription Factor-3/genetics , Acetylation , Carcinoma, Embryonal/pathology , Cell Line , Chromatin Assembly and Disassembly/genetics , DNA Methylation/drug effects , Epigenesis, Genetic , Histones/chemistry , Histones/metabolism , Humans , In Vitro Techniques , Lysine/chemistry , Methylation , Nanog Homeobox Protein , Promoter Regions, Genetic , Protein Processing, Post-Translational , Regulatory Sequences, Nucleic Acid , Tretinoin/pharmacology , Tumor Cells, Cultured , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL
...