Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
1.
Cell ; 153(5): 1134-48, 2013 May 23.
Article in English | MEDLINE | ID: mdl-23664764

ABSTRACT

Epigenetic mechanisms have been proposed to play crucial roles in mammalian development, but their precise functions are only partially understood. To investigate epigenetic regulation of embryonic development, we differentiated human embryonic stem cells into mesendoderm, neural progenitor cells, trophoblast-like cells, and mesenchymal stem cells and systematically characterized DNA methylation, chromatin modifications, and the transcriptome in each lineage. We found that promoters that are active in early developmental stages tend to be CG rich and mainly engage H3K27me3 upon silencing in nonexpressing lineages. By contrast, promoters for genes expressed preferentially at later stages are often CG poor and primarily employ DNA methylation upon repression. Interestingly, the early developmental regulatory genes are often located in large genomic domains that are generally devoid of DNA methylation in most lineages, which we termed DNA methylation valleys (DMVs). Our results suggest that distinct epigenetic mechanisms regulate early and late stages of ES cell differentiation.


Subject(s)
DNA Methylation , Embryonic Stem Cells/metabolism , Epigenomics , Gene Expression Regulation, Developmental , Animals , Cell Differentiation , Chromatin/metabolism , CpG Islands , Embryonic Stem Cells/cytology , Histones/metabolism , Humans , Methylation , Neoplasms/genetics , Promoter Regions, Genetic , Zebrafish/embryology
2.
Nature ; 471(7336): 68-73, 2011 Mar 03.
Article in English | MEDLINE | ID: mdl-21289626

ABSTRACT

Induced pluripotent stem cells (iPSCs) offer immense potential for regenerative medicine and studies of disease and development. Somatic cell reprogramming involves epigenomic reconfiguration, conferring iPSCs with characteristics similar to embryonic stem (ES) cells. However, it remains unknown how complete the reestablishment of ES-cell-like DNA methylation patterns is throughout the genome. Here we report the first whole-genome profiles of DNA methylation at single-base resolution in five human iPSC lines, along with methylomes of ES cells, somatic cells, and differentiated iPSCs and ES cells. iPSCs show significant reprogramming variability, including somatic memory and aberrant reprogramming of DNA methylation. iPSCs share megabase-scale differentially methylated regions proximal to centromeres and telomeres that display incomplete reprogramming of non-CG methylation, and differences in CG methylation and histone modifications. Lastly, differentiation of iPSCs into trophoblast cells revealed that errors in reprogramming CG methylation are transmitted at a high frequency, providing an iPSC reprogramming signature that is maintained after differentiation.


Subject(s)
Cellular Reprogramming/genetics , DNA Methylation/genetics , Epistasis, Genetic/genetics , Genome, Human/genetics , Induced Pluripotent Stem Cells/metabolism , Cell Differentiation/genetics , Cell Line , CpG Islands/genetics , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Epigenomics , Fibroblasts/cytology , Fibroblasts/metabolism , Histones/metabolism , Humans , Induced Pluripotent Stem Cells/cytology , Trophoblasts/cytology , Trophoblasts/metabolism
3.
J Bacteriol ; 191(16): 5240-52, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19502398

ABSTRACT

We have generated extreme ionizing radiation resistance in a relatively sensitive bacterial species, Escherichia coli, by directed evolution. Four populations of Escherichia coli K-12 were derived independently from strain MG1655, with each specifically adapted to survive exposure to high doses of ionizing radiation. D(37) values for strains isolated from two of the populations approached that exhibited by Deinococcus radiodurans. Complete genomic sequencing was carried out on nine purified strains derived from these populations. Clear mutational patterns were observed that both pointed to key underlying mechanisms and guided further characterization of the strains. In these evolved populations, passive genomic protection is not in evidence. Instead, enhanced recombinational DNA repair makes a prominent but probably not exclusive contribution to genome reconstitution. Multiple genes, multiple alleles of some genes, multiple mechanisms, and multiple evolutionary pathways all play a role in the evolutionary acquisition of extreme radiation resistance. Several mutations in the recA gene and a deletion of the e14 prophage both demonstrably contribute to and partially explain the new phenotype. Mutations in additional components of the bacterial recombinational repair system and the replication restart primosome are also prominent, as are mutations in genes involved in cell division, protein turnover, and glutamate transport. At least some evolutionary pathways to extreme radiation resistance are constrained by the temporally ordered appearance of specific alleles.


Subject(s)
Directed Molecular Evolution , Escherichia coli/genetics , Escherichia coli/radiation effects , Radiation, Ionizing , Chromatography, High Pressure Liquid , Electrophoresis, Gel, Pulsed-Field , Escherichia coli/growth & development , Mutation , Phylogeny , Rec A Recombinases/genetics , Rec A Recombinases/physiology
4.
Cell Res ; 21(10): 1393-409, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21876557

ABSTRACT

Pluripotency, the ability of a cell to differentiate and give rise to all embryonic lineages, defines a small number of mammalian cell types such as embryonic stem (ES) cells. While it has been generally held that pluripotency is the product of a transcriptional regulatory network that activates and maintains the expression of key stem cell genes, accumulating evidence is pointing to a critical role for epigenetic processes in establishing and safeguarding the pluripotency of ES cells, as well as maintaining the identity of differentiated cell types. In order to better understand the role of epigenetic mechanisms in pluripotency, we have examined the dynamics of chromatin modifications genome-wide in human ES cells (hESCs) undergoing differentiation into a mesendodermal lineage. We found that chromatin modifications at promoters remain largely invariant during differentiation, except at a small number of promoters where a dynamic switch between acetylation and methylation at H3K27 marks the transition between activation and silencing of gene expression, suggesting a hierarchy in cell fate commitment over most differentially expressed genes. We also mapped over 50 000 potential enhancers, and observed much greater dynamics in chromatin modifications, especially H3K4me1 and H3K27ac, which correlate with expression of their potential target genes. Further analysis of these enhancers revealed potentially key transcriptional regulators of pluripotency and a chromatin signature indicative of a poised state that may confer developmental competence in hESCs. Our results provide new evidence supporting the role of chromatin modifications in defining enhancers and pluripotency.


Subject(s)
Cell Differentiation/physiology , Embryonic Stem Cells/metabolism , Epigenesis, Genetic/physiology , Pluripotent Stem Cells/metabolism , Transcription, Genetic/physiology , Cell Line , Cell Lineage/physiology , Chromatin/genetics , Chromatin/metabolism , Embryonic Stem Cells/cytology , Enhancer Elements, Genetic/physiology , Genome-Wide Association Study , Humans , Pluripotent Stem Cells/cytology
5.
Nat Struct Mol Biol ; 18(1): 91-3, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21131980

ABSTRACT

We have tested the specificity and utility of more than 200 antibodies raised against 57 different histone modifications in Drosophila melanogaster, Caenorhabditis elegans and human cells. Although most antibodies performed well, more than 25% failed specificity tests by dot blot or western blot. Among specific antibodies, more than 20% failed in chromatin immunoprecipitation experiments. We advise rigorous testing of histone-modification antibodies before use, and we provide a website for posting new test results (http://compbio.med.harvard.edu/antibodies/).


Subject(s)
Antibody Specificity , Histones/immunology , Animals , Antibodies/chemistry , Blotting, Western , Caenorhabditis elegans/genetics , Caenorhabditis elegans Proteins/chemistry , Chromatin Immunoprecipitation , Drosophila Proteins/chemistry , Drosophila melanogaster/genetics , Histones/chemistry , Histones/metabolism , Immunoblotting , Protein Processing, Post-Translational , Quality Control , Reproducibility of Results
6.
Cell Stem Cell ; 6(5): 479-91, 2010 May 07.
Article in English | MEDLINE | ID: mdl-20452322

ABSTRACT

Human embryonic stem cells (hESCs) share an identical genome with lineage-committed cells, yet possess the remarkable properties of self-renewal and pluripotency. The diverse cellular properties in different cells have been attributed to their distinct epigenomes, but how much epigenomes differ remains unclear. Here, we report that epigenomic landscapes in hESCs and lineage-committed cells are drastically different. By comparing the chromatin-modification profiles and DNA methylomes in hESCs and primary fibroblasts, we find that nearly one-third of the genome differs in chromatin structure. Most changes arise from dramatic redistributions of repressive H3K9me3 and H3K27me3 marks, which form blocks that significantly expand in fibroblasts. A large number of potential regulatory sequences also exhibit a high degree of dynamics in chromatin modifications and DNA methylation. Additionally, we observe novel, context-dependent relationships between DNA methylation and chromatin modifications. Our results provide new insights into epigenetic mechanisms underlying properties of pluripotency and cell fate commitment.


Subject(s)
Cell Lineage/genetics , Epigenesis, Genetic , Fibroblasts/cytology , Fibroblasts/metabolism , Genome, Human/genetics , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Cell Line , Chromatin/genetics , CpG Islands/genetics , DNA Methylation/genetics , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Genes, Developmental , Histones/metabolism , Humans , Lysine/metabolism , Protein Processing, Post-Translational , Regulatory Sequences, Nucleic Acid/genetics
SELECTION OF CITATIONS
SEARCH DETAIL