Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 69
Filter
Add more filters

Publication year range
1.
Cell ; 158(6): 1254-1269, 2014 Sep 11.
Article in English | MEDLINE | ID: mdl-25215486

ABSTRACT

Current human pluripotent stem cells lack the transcription factor circuitry that governs the ground state of mouse embryonic stem cells (ESC). Here, we report that short-term expression of two components, NANOG and KLF2, is sufficient to ignite other elements of the network and reset the human pluripotent state. Inhibition of ERK and protein kinase C sustains a transgene-independent rewired state. Reset cells self-renew continuously without ERK signaling, are phenotypically stable, and are karyotypically intact. They differentiate in vitro and form teratomas in vivo. Metabolism is reprogrammed with activation of mitochondrial respiration as in ESC. DNA methylation is dramatically reduced and transcriptome state is globally realigned across multiple cell lines. Depletion of ground-state transcription factors, TFCP2L1 or KLF4, has marginal impact on conventional human pluripotent stem cells but collapses the reset state. These findings demonstrate feasibility of installing and propagating functional control circuitry for ground-state pluripotency in human cells.


Subject(s)
Homeodomain Proteins/metabolism , Kruppel-Like Transcription Factors/metabolism , Pluripotent Stem Cells/metabolism , Animals , Cytological Techniques , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Epigenesis, Genetic , Humans , Kruppel-Like Factor 4 , Mice , Mitochondria/metabolism , Nanog Homeobox Protein , Pluripotent Stem Cells/cytology , Transcription Factors/metabolism , Transcriptome
2.
Development ; 150(2)2023 01 15.
Article in English | MEDLINE | ID: mdl-36621005

ABSTRACT

Gene duplication events can drive evolution by providing genetic material for new gene functions, and they create opportunities for diverse developmental strategies to emerge between species. To study the contribution of duplicated genes to human early development, we examined the evolution and function of NANOGP1, a tandem duplicate of the transcription factor NANOG. We found that NANOGP1 and NANOG have overlapping but distinct expression profiles, with high NANOGP1 expression restricted to early epiblast cells and naïve-state pluripotent stem cells. Sequence analysis and epitope-tagging revealed that NANOGP1 is protein coding with an intact homeobox domain. The duplication that created NANOGP1 occurred earlier in primate evolution than previously thought and has been retained only in great apes, whereas Old World monkeys have disabled the gene in different ways, including homeodomain point mutations. NANOGP1 is a strong inducer of naïve pluripotency; however, unlike NANOG, it is not required to maintain the undifferentiated status of human naïve pluripotent cells. By retaining expression, sequence and partial functional conservation with its ancestral copy, NANOGP1 exemplifies how gene duplication and subfunctionalisation can contribute to transcription factor activity in human pluripotency and development.


Subject(s)
Genes, Homeobox , Pluripotent Stem Cells , Animals , Humans , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Nanog Homeobox Protein/genetics , Nanog Homeobox Protein/metabolism , Pluripotent Stem Cells/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
4.
Nature ; 576(7787): 487-491, 2019 12.
Article in English | MEDLINE | ID: mdl-31827285

ABSTRACT

Formation of the three primary germ layers during gastrulation is an essential step in the establishment of the vertebrate body plan and is associated with major transcriptional changes1-5. Global epigenetic reprogramming accompanies these changes6-8, but the role of the epigenome in regulating early cell-fate choice remains unresolved, and the coordination between different molecular layers is unclear. Here we describe a single-cell multi-omics map of chromatin accessibility, DNA methylation and RNA expression during the onset of gastrulation in mouse embryos. The initial exit from pluripotency coincides with the establishment of a global repressive epigenetic landscape, followed by the emergence of lineage-specific epigenetic patterns during gastrulation. Notably, cells committed to mesoderm and endoderm undergo widespread coordinated epigenetic rearrangements at enhancer marks, driven by ten-eleven translocation (TET)-mediated demethylation and a concomitant increase of accessibility. By contrast, the methylation and accessibility landscape of ectodermal cells is already established in the early epiblast. Hence, regulatory elements associated with each germ layer are either epigenetically primed or remodelled before cell-fate decisions, providing the molecular framework for a hierarchical emergence of the primary germ layers.


Subject(s)
DNA Methylation , Epigenesis, Genetic , Gastrula/cytology , Gastrula/metabolism , Gastrulation/genetics , Gene Expression Regulation, Developmental , RNA/genetics , Single-Cell Analysis , Animals , Cell Differentiation/genetics , Cell Lineage/genetics , Chromatin/genetics , Chromatin/metabolism , Demethylation , Embryoid Bodies/cytology , Endoderm/cytology , Endoderm/embryology , Endoderm/metabolism , Enhancer Elements, Genetic/genetics , Epigenome/genetics , Erythropoiesis , Factor Analysis, Statistical , Gastrula/embryology , Gastrulation/physiology , Mesoderm/cytology , Mesoderm/embryology , Mesoderm/metabolism , Mice , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , RNA/analysis , Time Factors , Zinc Fingers
5.
PLoS Biol ; 19(3): e3000886, 2021 03.
Article in English | MEDLINE | ID: mdl-33760805

ABSTRACT

Faithful replication of the entire genome requires replication forks to copy large contiguous tracts of DNA, and sites of persistent replication fork stalling present a major threat to genome stability. Understanding the distribution of sites at which replication forks stall, and the ensuing fork processing events, requires genome-wide methods that profile replication fork position and the formation of recombinogenic DNA ends. Here, we describe Transferase-Activated End Ligation sequencing (TrAEL-seq), a method that captures single-stranded DNA 3' ends genome-wide and with base pair resolution. TrAEL-seq labels both DNA breaks and replication forks, providing genome-wide maps of replication fork progression and fork stalling sites in yeast and mammalian cells. Replication maps are similar to those obtained by Okazaki fragment sequencing; however, TrAEL-seq is performed on asynchronous populations of wild-type cells without incorporation of labels, cell sorting, or biochemical purification of replication intermediates, rendering TrAEL-seq far simpler and more widely applicable than existing replication fork direction profiling methods. The specificity of TrAEL-seq for DNA 3' ends also allows accurate detection of double-strand break sites after the initiation of DNA end resection, which we demonstrate by genome-wide mapping of meiotic double-strand break hotspots in a dmc1Δ mutant that is competent for end resection but not strand invasion. Overall, TrAEL-seq provides a flexible and robust methodology with high sensitivity and resolution for studying DNA replication and repair, which will be of significant use in determining mechanisms of genome instability.


Subject(s)
DNA Replication/genetics , DNA Replication/physiology , Sequence Analysis, DNA/methods , 3' Untranslated Regions/genetics , Animals , DNA/chemistry , DNA/genetics , DNA Breaks, Double-Stranded , DNA Repair/genetics , DNA-Binding Proteins/metabolism , Genome/genetics , Humans
6.
Nucleic Acids Res ; 50(2): 915-936, 2022 01 25.
Article in English | MEDLINE | ID: mdl-35018465

ABSTRACT

Adaptive mutations can cause drug resistance in cancers and pathogens, and increase the tolerance of agricultural pests and diseases to chemical treatment. When and how adaptive mutations form is often hard to discern, but we have shown that adaptive copy number amplification of the copper resistance gene CUP1 occurs in response to environmental copper due to CUP1 transcriptional activation. Here we dissect the mechanism by which CUP1 transcription in budding yeast stimulates copy number variation (CNV). We show that transcriptionally stimulated CNV requires TREX-2 and Mediator, such that cells lacking TREX-2 or Mediator respond normally to copper but cannot acquire increased resistance. Mediator and TREX-2 can cause replication stress by tethering transcribed loci to nuclear pores, a process known as gene gating, and transcription at the CUP1 locus causes a TREX-2-dependent accumulation of replication forks indicative of replication fork stalling. TREX-2-dependent CUP1 gene amplification occurs by a Rad52 and Rad51-mediated homologous recombination mechanism that is enhanced by histone H3K56 acetylation and repressed by Pol32 and Pif1. CUP1 amplification is also critically dependent on late-firing replication origins present in the CUP1 repeats, and mutations that remove or inactivate these origins strongly suppress the acquisition of copper resistance. We propose that replicative stress imposed by nuclear pore association causes replication bubbles from these origins to collapse soon after activation, leaving a tract of H3K56-acetylated chromatin that promotes secondary recombination events during elongation after replication fork re-start events. The capacity for inefficient replication origins to promote copy number variation renders certain genomic regions more fragile than others, and therefore more likely to undergo adaptive evolution through de novo gene amplification.


Subject(s)
DNA, Fungal/metabolism , Exodeoxyribonucleases/metabolism , Histones/metabolism , Metallothionein/metabolism , Saccharomyces cerevisiae/metabolism , DNA Replication , Homologous Recombination , Replication Origin
7.
Magn Reson Med ; 89(3): 1002-1015, 2023 03.
Article in English | MEDLINE | ID: mdl-36336877

ABSTRACT

PURPOSE: Subject-tailored parallel transmission pulses for ultra-high fields body applications are typically calculated based on subject-specific B 1 + $$ {\mathrm{B}}_1^{+} $$ -maps of all transmit channels, which require lengthy adjustment times. This study investigates the feasibility of using deep learning to estimate complex, channel-wise, relative 2D B 1 + $$ {\mathrm{B}}_1^{+} $$ -maps from a single gradient echo localizer to overcome long calibration times. METHODS: 126 channel-wise, complex, relative 2D B 1 + $$ {\mathrm{B}}_1^{+} $$ -maps of the human heart from 44 subjects were acquired at 7T using a Cartesian, cardiac gradient-echo sequence obtained under breath-hold to create a library for network training and cross-validation. The deep learning predicted maps were qualitatively compared to the ground truth. Phase-only B 1 + $$ {\mathrm{B}}_1^{+} $$ -shimming was subsequently performed on the estimated B 1 + $$ {\mathrm{B}}_1^{+} $$ -maps for a region of interest covering the heart. The proposed network was applied at 7T to 3 unseen test subjects. RESULTS: The deep learning-based B 1 + $$ {\mathrm{B}}_1^{+} $$ -maps, derived in approximately 0.2 seconds, match the ground truth for the magnitude and phase. The static, phase-only pulse design performs best when maximizing the mean transmission efficiency. In-vivo application of the proposed network to unseen subjects demonstrates the feasibility of this approach: the network yields predicted B 1 + $$ {\mathrm{B}}_1^{+} $$ -maps comparable to the acquired ground truth and anatomical scans reflect the resulting B 1 + $$ {\mathrm{B}}_1^{+} $$ -pattern using the deep learning-based maps. CONCLUSION: The feasibility of estimating 2D relative B 1 + $$ {\mathrm{B}}_1^{+} $$ -maps from initial localizer scans of the human heart at 7T using deep learning is successfully demonstrated. Because the technique requires only sub-seconds to derive channel-wise B 1 + $$ {\mathrm{B}}_1^{+} $$ -maps, it offers high potential for advancing clinical body imaging at ultra-high fields.


Subject(s)
Deep Learning , Magnetic Resonance Imaging , Humans , Magnetic Resonance Imaging/methods , Image Interpretation, Computer-Assisted/methods , Heart/diagnostic imaging , Calibration
8.
PLoS Biol ; 17(12): e3000471, 2019 12.
Article in English | MEDLINE | ID: mdl-31794573

ABSTRACT

Extrachromosomal circular DNA (eccDNA) facilitates adaptive evolution by allowing rapid and extensive gene copy number variation and is implicated in the pathology of cancer and ageing. Here, we demonstrate that yeast aged under environmental copper accumulate high levels of eccDNA containing the copper-resistance gene CUP1. Transcription of the tandemly repeated CUP1 gene causes CUP1 eccDNA accumulation, which occurs in the absence of phenotypic selection. We have developed a sensitive and quantitative eccDNA sequencing pipeline that reveals CUP1 eccDNA accumulation on copper exposure to be exquisitely site specific, with no other detectable changes across the eccDNA complement. eccDNA forms de novo from the CUP1 locus through processing of DNA double-strand breaks (DSBs) by Sae2, Mre11 and Mus81, and genome-wide analyses show that other protein coding eccDNA species in aged yeast share a similar biogenesis pathway. Although abundant, we find that CUP1 eccDNA does not replicate efficiently, and high-copy numbers in aged cells arise through frequent formation events combined with asymmetric DNA segregation. The transcriptional stimulation of CUP1 eccDNA formation shows that age-linked genetic change varies with transcription pattern, resulting in gene copy number profiles tailored by environment.


Subject(s)
DNA Copy Number Variations/genetics , DNA, Circular/genetics , Transcription, Genetic/genetics , Age Factors , Copper/metabolism , Copper/pharmacology , DNA, Circular/metabolism , Endonucleases , Gene Dosage/genetics , Metallothionein/genetics , Metallothionein/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics , Tandem Repeat Sequences/genetics
9.
Nucleic Acids Res ; 48(16): e92, 2020 09 18.
Article in English | MEDLINE | ID: mdl-32621604

ABSTRACT

Genomic imprinting is an epigenetic phenomenon leading to parental allele-specific expression. Dosage of imprinted genes is crucial for normal development and its dysregulation accounts for several human disorders. This unusual expression pattern is mostly dictated by differences in DNA methylation between parental alleles at specific regulatory elements known as imprinting control regions (ICRs). Although several approaches can be used for methylation inspection, we lack an easy and cost-effective method to simultaneously measure DNA methylation at multiple imprinted regions. Here, we present IMPLICON, a high-throughput method measuring DNA methylation levels at imprinted regions with base-pair resolution and over 1000-fold coverage. We adapted amplicon bisulfite-sequencing protocols to design IMPLICON for ICRs in adult tissues of inbred mice, validating it in hybrid mice from reciprocal crosses for which we could discriminate methylation profiles in the two parental alleles. Lastly, we developed a human version of IMPLICON and detected imprinting errors in embryonic and induced pluripotent stem cells. We also provide rules and guidelines to adapt this method for investigating the DNA methylation landscape of any set of genomic regions. In summary, IMPLICON is a rapid, cost-effective and scalable method, which could become the gold standard in both imprinting research and diagnostics.


Subject(s)
CpG Islands , DNA Methylation , Genomic Imprinting , High-Throughput Nucleotide Sequencing/methods , Animals , Cells, Cultured , Female , Fibroblasts , Human Embryonic Stem Cells , Humans , Induced Pluripotent Stem Cells , Male , Mice , Mice, Inbred C57BL
10.
Mol Cell ; 48(6): 849-62, 2012 Dec 28.
Article in English | MEDLINE | ID: mdl-23219530

ABSTRACT

Genome-wide DNA methylation reprogramming occurs in mouse primordial germ cells (PGCs) and preimplantation embryos, but the precise dynamics and biological outcomes are largely unknown. We have carried out whole-genome bisulfite sequencing (BS-Seq) and RNA-Seq across key stages from E6.5 epiblast to E16.5 PGCs. Global loss of methylation takes place during PGC expansion and migration with evidence for passive demethylation, but sequences that carry long-term epigenetic memory (imprints, CpG islands on the X chromosome, germline-specific genes) only become demethylated upon entry of PGCs into the gonads. The transcriptional profile of PGCs is tightly controlled despite global hypomethylation, with transient expression of the pluripotency network, suggesting that reprogramming and pluripotency are inextricably linked. Our results provide a framework for the understanding of the epigenetic ground state of pluripotency in the germline.


Subject(s)
DNA Methylation/genetics , Genome , Genomic Imprinting , Germ Cells/metabolism , Transcriptome , Animals , CpG Islands , Female , Germ Layers/cytology , Male , Mice , Mice, Inbred C57BL , Sequence Analysis, DNA , Transcription, Genetic , X Chromosome/genetics , X Chromosome/metabolism
11.
Nat Methods ; 13(3): 229-232, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26752769

ABSTRACT

We report scM&T-seq, a method for parallel single-cell genome-wide methylome and transcriptome sequencing that allows for the discovery of associations between transcriptional and epigenetic variation. Profiling of 61 mouse embryonic stem cells confirmed known links between DNA methylation and transcription. Notably, the method revealed previously unrecognized associations between heterogeneously methylated distal regulatory elements and transcription of key pluripotency genes.


Subject(s)
Embryonic Stem Cells/physiology , Epigenesis, Genetic/genetics , High-Throughput Nucleotide Sequencing/methods , Regulatory Elements, Transcriptional/genetics , Transcription Factors/genetics , Animals , Base Sequence , Cells, Cultured , Mice , Molecular Sequence Data
12.
Nucleic Acids Res ; 45(20): 11559-11569, 2017 Nov 16.
Article in English | MEDLINE | ID: mdl-29036576

ABSTRACT

DNA methylation is an important epigenetic modification in many species that is critical for development, and implicated in ageing and many complex diseases, such as cancer. Many cost-effective genome-wide analyses of DNA modifications rely on restriction enzymes capable of digesting genomic DNA at defined sequence motifs. There are hundreds of restriction enzyme families but few are used to date, because no tool is available for the systematic evaluation of restriction enzyme combinations that can enrich for certain sites of interest in a genome. Herein, we present customised Reduced Representation Bisulfite Sequencing (cuRRBS), a novel and easy-to-use computational method that solves this problem. By computing the optimal enzymatic digestions and size selection steps required, cuRRBS generalises the traditional MspI-based Reduced Representation Bisulfite Sequencing (RRBS) protocol to all restriction enzyme combinations. In addition, cuRRBS estimates the fold-reduction in sequencing costs and provides a robustness value for the personalised RRBS protocol, allowing users to tailor the protocol to their experimental needs. Moreover, we show in silico that cuRRBS-defined restriction enzymes consistently out-perform MspI digestion in many biological systems, considering both CpG and CHG contexts. Finally, we have validated the accuracy of cuRRBS predictions for single and double enzyme digestions using two independent experimental datasets.


Subject(s)
Computational Biology/methods , DNA Methylation/genetics , Sequence Analysis, DNA/economics , Sequence Analysis, DNA/methods , Whole Genome Sequencing/methods , Animals , Arabidopsis/genetics , Binding Sites/genetics , CCCTC-Binding Factor/genetics , CCCTC-Binding Factor/metabolism , CpG Islands/genetics , DNA Restriction Enzymes/chemistry , Humans , Induced Pluripotent Stem Cells/cytology , Mice , Nuclear Respiratory Factor 1/genetics , Nuclear Respiratory Factor 1/metabolism
13.
Nucleic Acids Res ; 45(4): 1703-1713, 2017 02 28.
Article in English | MEDLINE | ID: mdl-27899645

ABSTRACT

DNA methylation plays a critical role in the regulation and maintenance of cell-type specific transcriptional programs. Targeted epigenome editing is an emerging technology to specifically regulate cellular gene expression in order to modulate cell phenotypes or dissect the epigenetic mechanisms involved in their control. In this work, we employed a DNA methyltransferase Dnmt3a-Dnmt3L construct fused to the nuclease-inactivated dCas9 programmable targeting domain to introduce DNA methylation into the human genome specifically at the EpCAM, CXCR4 and TFRC gene promoters. We show that targeting of these loci with single gRNAs leads to efficient and widespread methylation of the promoters. Multiplexing of several guide RNAs does not increase the efficiency of methylation. Peaks of targeted methylation were observed around 25 bp upstream and 40 bp downstream of the PAM site, while 20-30 bp of the binding site itself are protected against methylation. Potent methylation is dependent on the multimerization of Dnmt3a/Dnmt3L complexes on the DNA. Furthermore, the introduced methylation causes transcriptional repression of the targeted genes. These new programmable epigenetic editors allow unprecedented control of the DNA methylation status in cells and will lead to further advances in the understanding of epigenetic signaling.


Subject(s)
DNA (Cytosine-5-)-Methyltransferases/metabolism , DNA Methylation , DNA/genetics , DNA/metabolism , Endonucleases/metabolism , Animals , Cell Line, Tumor , Cluster Analysis , CpG Islands , DNA (Cytosine-5-)-Methyltransferases/chemistry , DNA (Cytosine-5-)-Methyltransferases/genetics , Endonucleases/genetics , Epigenesis, Genetic , Epigenomics/methods , Gene Expression Regulation , High-Throughput Nucleotide Sequencing , Humans , Mice , Models, Biological , Promoter Regions, Genetic , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism
14.
Proc Natl Acad Sci U S A ; 112(45): 13970-5, 2015 Nov 10.
Article in English | MEDLINE | ID: mdl-26483466

ABSTRACT

Phenotypic plasticity is important in adaptation and shapes the evolution of organisms. However, we understand little about what aspects of the genome are important in facilitating plasticity. Eusocial insect societies produce plastic phenotypes from the same genome, as reproductives (queens) and nonreproductives (workers). The greatest plasticity is found in the simple eusocial insect societies in which individuals retain the ability to switch between reproductive and nonreproductive phenotypes as adults. We lack comprehensive data on the molecular basis of plastic phenotypes. Here, we sequenced genomes, microRNAs (miRNAs), and multiple transcriptomes and methylomes from individual brains in a wasp (Polistes canadensis) and an ant (Dinoponera quadriceps) that live in simple eusocial societies. In both species, we found few differences between phenotypes at the transcriptional level, with little functional specialization, and no evidence that phenotype-specific gene expression is driven by DNA methylation or miRNAs. Instead, phenotypic differentiation was defined more subtly by nonrandom transcriptional network organization, with roles in these networks for both conserved and taxon-restricted genes. The general lack of highly methylated regions or methylome patterning in both species may be an important mechanism for achieving plasticity among phenotypes during adulthood. These findings define previously unidentified hypotheses on the genomic processes that facilitate plasticity and suggest that the molecular hallmarks of social behavior are likely to differ with the level of social complexity.


Subject(s)
Ants/genetics , Gene Expression Regulation/genetics , Hierarchy, Social , Models, Genetic , Phenotype , Social Behavior , Wasps/genetics , Animals , Ants/physiology , Base Sequence , Brain/metabolism , DNA Methylation/genetics , Genome, Insect/genetics , High-Throughput Nucleotide Sequencing , MicroRNAs/genetics , Molecular Sequence Data , Transcriptome/genetics , Wasps/physiology
15.
Nat Methods ; 11(8): 817-820, 2014 Aug.
Article in English | MEDLINE | ID: mdl-25042786

ABSTRACT

We report a single-cell bisulfite sequencing (scBS-seq) method that can be used to accurately measure DNA methylation at up to 48.4% of CpG sites. Embryonic stem cells grown in serum or in 2i medium displayed epigenetic heterogeneity, with '2i-like' cells present in serum culture. Integration of 12 individual mouse oocyte datasets largely recapitulated the whole DNA methylome, which makes scBS-seq a versatile tool to explore DNA methylation in rare cells and heterogeneous populations.


Subject(s)
Epigenesis, Genetic , Genome , Sulfites/chemistry , Animals , DNA Methylation , Mice
16.
Nature ; 473(7347): 398-402, 2011 May 19.
Article in English | MEDLINE | ID: mdl-21460836

ABSTRACT

Methylation at the 5' position of cytosine in DNA has important roles in genome function and is dynamically reprogrammed during early embryonic and germ cell development. The mammalian genome also contains 5-hydroxymethylcytosine (5hmC), which seems to be generated by oxidation of 5-methylcytosine (5mC) by the TET family of enzymes that are highly expressed in embryonic stem (ES) cells. Here we use antibodies against 5hmC and 5mC together with high throughput sequencing to determine genome-wide patterns of methylation and hydroxymethylation in mouse wild-type and mutant ES cells and differentiating embryoid bodies. We find that 5hmC is mostly associated with euchromatin and that whereas 5mC is under-represented at gene promoters and CpG islands, 5hmC is enriched and is associated with increased transcriptional levels. Most, if not all, 5hmC in the genome depends on pre-existing 5mC and the balance between these two modifications is different between genomic regions. Knockdown of Tet1 and Tet2 causes downregulation of a group of genes that includes pluripotency-related genes (including Esrrb, Prdm14, Dppa3, Klf2, Tcl1 and Zfp42) and a concomitant increase in methylation of their promoters, together with an increased propensity of ES cells for extraembryonic lineage differentiation. Declining levels of TETs during differentiation are associated with decreased hydroxymethylation levels at the promoters of ES cell-specific genes together with increased methylation and gene silencing. We propose that the balance between hydroxymethylation and methylation in the genome is inextricably linked with the balance between pluripotency and lineage commitment.


Subject(s)
Cell Differentiation/genetics , Cytosine/analogs & derivatives , DNA Methylation , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Gene Expression Regulation, Developmental , 5-Methylcytosine/analogs & derivatives , Animals , Antibodies/immunology , Cell Line , Cell Lineage/genetics , CpG Islands/genetics , Cytosine/analysis , Cytosine/immunology , Cytosine/metabolism , DNA-Binding Proteins/deficiency , Dioxygenases , Down-Regulation , Embryoid Bodies/cytology , Embryoid Bodies/metabolism , Euchromatin/genetics , Euchromatin/metabolism , Exons/genetics , Gene Silencing , Genome/genetics , Mice , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Promoter Regions, Genetic/genetics , Proto-Oncogene Proteins/deficiency , Reproducibility of Results , Sequence Analysis, DNA , Transcription, Genetic
17.
Nat Methods ; 9(2): 145-51, 2012 Jan 30.
Article in English | MEDLINE | ID: mdl-22290186

ABSTRACT

Bisulfite conversion of genomic DNA combined with next-generation sequencing (BS-seq) is widely used to measure the methylation state of a whole genome, the methylome, at single-base resolution. However, analysis of BS-seq data still poses a considerable challenge. Here we summarize the challenges of BS-seq mapping as they apply to both base and color-space data. We also explore the effect of sequencing errors and contaminants on inferred methylation levels and recommend the most appropriate way to analyze this type of data.


Subject(s)
DNA Methylation , DNA/metabolism , Sequence Analysis, DNA , Sulfites/chemistry , DNA/genetics
18.
PLoS Genet ; 8(9): e1002974, 2012 Sep.
Article in English | MEDLINE | ID: mdl-23028372

ABSTRACT

Budding yeast centromeres are sequence-defined point centromeres and are, unlike in many other organisms, not embedded in heterochromatin. Here we show that Fun30, a poorly understood SWI/SNF-like chromatin remodeling factor conserved in humans, promotes point centromere function through the formation of correct chromatin architecture at centromeres. Our determination of the genome-wide binding and nucleosome positioning properties of Fun30 shows that this enzyme is consistently enriched over centromeres and that a majority of CENs show Fun30-dependent changes in flanking nucleosome position and/or CEN core micrococcal nuclease accessibility. Fun30 deletion leads to defects in histone variant Htz1 occupancy genome-wide, including at and around most centromeres. FUN30 genetically interacts with CSE4, coding for the centromere-specific variant of histone H3, and counteracts the detrimental effect of transcription through centromeres on chromosome segregation and suppresses transcriptional noise over centromere CEN3. Previous work has shown a requirement for fission yeast and mammalian homologs of Fun30 in heterochromatin assembly. As centromeres in budding yeast are not embedded in heterochromatin, our findings indicate a direct role of Fun30 in centromere chromatin by promoting correct chromatin architecture.


Subject(s)
Centromere/genetics , Chromosomal Proteins, Non-Histone , DNA-Binding Proteins , Saccharomyces cerevisiae Proteins , Saccharomyces cerevisiae , Transcription Factors , Chromatin Assembly and Disassembly/genetics , Chromosomal Proteins, Non-Histone/genetics , Chromosomal Proteins, Non-Histone/metabolism , Chromosome Segregation/genetics , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Heterochromatin/genetics , Histones/genetics , Humans , Kinetochores , Nucleosomes/genetics , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
19.
Bioinformatics ; 28(3): 428-9, 2012 Feb 01.
Article in English | MEDLINE | ID: mdl-22155865

ABSTRACT

SUMMARY: Bisulfite sequencing, a combination of bisulfite treatment and high-throughput sequencing, has proved to be a valuable method for measuring DNA methylation at single base resolution. Here, we present B-SOLANA, an approach for the analysis of two-base encoding (colorspace) bisulfite sequencing data on the SOLiD platform of Life Technologies. It includes the alignment of bisulfite sequences and the determination of methylation levels in CpG as well as non-CpG sequence contexts. B-SOLANA enables a fast and accurate analysis of large raw sequence datasets. AVAILABILITY AND IMPLEMENTATION: The source code, released under the GNU GPLv3 licence, is freely available at http://code.google.com/p/bsolana/. CONTACT: b.kreck@ikmb.uni-kiel.de SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.


Subject(s)
DNA Methylation , Sequence Analysis, DNA/methods , Software , Genome, Human , Genome-Wide Association Study , Humans , Sequence Alignment , Sulfites
20.
Stem Cells ; 30(12): 2732-45, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23034951

ABSTRACT

Embryonic (ES) and epiblast (EpiSC) stem cells are pluripotent but committed to an embryonic lineage fate. Conversely, trophoblast (TS) and extraembryonic endoderm (XEN) stem cells contribute predominantly to tissues of the placenta and yolk sac, respectively. Here we show that each of these four stem cell types is defined by a unique DNA methylation profile. Despite their distinct developmental origin, TS and XEN cells share key epigenomic hallmarks, chiefly characterized by robust DNA methylation of embryo-specific developmental regulators, as well as a subordinate role of 5-hydroxymethylation. We also observe a substantial methylation reinforcement of pre-existing epigenetic repressive marks that specifically occurs in extraembryonic stem cells compared to in vivo tissue, presumably due to continued high Dnmt3b expression levels. These differences establish a major epigenetic barrier between the embryonic and extraembryonic stem cell types. In addition, epigenetic lineage boundaries also separate the two extraembryonic stem cell types by mutual repression of key lineage-specific transcription factors. Thus, global DNA methylation patterns are a defining feature of each stem cell type that underpin lineage commitment and differentiative potency of early embryo-derived stem cells. Our detailed methylation profiles identify a cohort of developmentally regulated sequence elements, such as orphan CpG islands, that will be most valuable to uncover novel transcriptional regulators and pivotal "gatekeeper" genes in pluripotency and lineage differentiation.


Subject(s)
DNA Methylation , Embryonic Stem Cells/classification , Pluripotent Stem Cells/classification , Animals , Cell Differentiation/physiology , Cell Lineage , Embryonic Stem Cells/cytology , Embryonic Stem Cells/physiology , Endoderm/cytology , Female , Genomic Imprinting , Male , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/physiology , Pregnancy , Trophoblasts/cytology , X Chromosome Inactivation
SELECTION OF CITATIONS
SEARCH DETAIL