Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
Add more filters










Publication year range
1.
Nat Genet ; 54(12): 1907-1918, 2022 12.
Article in English | MEDLINE | ID: mdl-36471076

ABSTRACT

In mammals, interactions between sequences within topologically associating domains enable control of gene expression across large genomic distances. Yet it is unknown how frequently such contacts occur, how long they last and how they depend on the dynamics of chromosome folding and loop extrusion activity of cohesin. By imaging chromosomal locations at high spatial and temporal resolution in living cells, we show that interactions within topologically associating domains are transient and occur frequently during the course of a cell cycle. Interactions become more frequent and longer in the presence of convergent CTCF sites, resulting in suppression of variability in chromosome folding across time. Supported by physical models of chromosome dynamics, our data suggest that CTCF-anchored loops last around 10 min. Our results show that long-range transcriptional regulation might rely on transient physical proximity, and that cohesin and CTCF stabilize highly dynamic chromosome structures, facilitating selected subsets of chromosomal interactions.


Subject(s)
Chromosomes , Chromosomes/genetics
2.
Nature ; 604(7906): 571-577, 2022 04.
Article in English | MEDLINE | ID: mdl-35418676

ABSTRACT

Chromosome structure in mammals is thought to regulate transcription by modulating three-dimensional interactions between enhancers and promoters, notably through CTCF-mediated loops and topologically associating domains (TADs)1-4. However, how chromosome interactions are actually translated into transcriptional outputs remains unclear. Here, to address this question, we use an assay to position an enhancer at large numbers of densely spaced chromosomal locations relative to a fixed promoter, and measure promoter output and interactions within a genomic region with minimal regulatory and structural complexity. A quantitative analysis of hundreds of cell lines reveals that the transcriptional effect of an enhancer depends on its contact probabilities with the promoter through a nonlinear relationship. Mathematical modelling suggests that nonlinearity might arise from transient enhancer-promoter interactions being translated into slower promoter bursting dynamics in individual cells, therefore uncoupling the temporal dynamics of interactions from those of transcription. This uncovers a potential mechanism of how distal enhancers act from large genomic distances, and of how topologically associating domain boundaries block distal enhancers. Finally, we show that enhancer strength also determines absolute transcription levels as well as the sensitivity of a promoter to CTCF-mediated transcriptional insulation. Our measurements establish general principles for the context-dependent role of chromosome structure in long-range transcriptional regulation.


Subject(s)
Chromosomes , Enhancer Elements, Genetic , Animals , Chromatin/genetics , Enhancer Elements, Genetic/genetics , Gene Expression Regulation , Genomics , Mammals/genetics , Promoter Regions, Genetic/genetics
3.
Sci Adv ; 7(50): eabj6897, 2021 Dec 10.
Article in English | MEDLINE | ID: mdl-34890235

ABSTRACT

Mechanical input shapes cell fate decisions during development and regeneration in many systems, yet the mechanisms of this cross-talk are often unclear. In regenerating Hydra tissue spheroids, periodic osmotically driven inflation and deflation cycles generate mechanical stimuli in the form of tissue stretching. Here, we demonstrate that tissue stretching during inflation is important for the appearance of the head organizer­a group of cells that secrete the Wnt3 ligand. Exploiting time series RNA expression profiles, we identify the up-regulation of Wnt signaling as a key readout of the mechanical input. In this system, the levels of Wnt3 expression correspond to the levels of stretching, and Wnt3 overexpression alone enables successful regeneration in the absence of mechanical stimulation. Our findings enable the incorporation of mechanical signals in the framework of Hydra patterning and highlight the broad significance of mechanochemical feedback loops for patterning epithelial lumens.

4.
J Biol Chem ; 297(2): 100947, 2021 08.
Article in English | MEDLINE | ID: mdl-34270961

ABSTRACT

Transcription factors (TFs) harboring broad-complex, tramtrack, and bric-a-brac (BTB) domains play important roles in development and disease. These BTB domains are thought to recruit transcriptional modulators to target DNA regions. However, a systematic molecular understanding of the mechanism of action of this TF family is lacking. Here, we identify the zinc finger BTB-TF Zbtb2 from a genetic screen for regulators of exit from pluripotency and demonstrate that its absence perturbs embryonic stem cell differentiation and the gene expression dynamics underlying peri-implantation development. We show that ZBTB2 binds the chromatin remodeler Ep400 to mediate downstream transcription. Independently, the BTB domain directly interacts with nucleosome remodeling and deacetylase and histone chaperone histone regulator A. Nucleosome remodeling and deacetylase recruitment is a common feature of BTB TFs, and based on phylogenetic analysis, we propose that this is a conserved evolutionary property. Binding to UBN2, in contrast, is specific to ZBTB2 and requires a C-terminal extension of the BTB domain. Taken together, this study identifies a BTB-domain TF that recruits chromatin modifiers and a histone chaperone during a developmental cell state transition and defines unique and shared molecular functions of the BTB-domain TF family.


Subject(s)
Repressor Proteins , Transcription Factors , BTB-POZ Domain , Histone Chaperones , Humans , Phylogeny , Zinc Fingers
5.
Nature ; 596(7870): 133-137, 2021 08.
Article in English | MEDLINE | ID: mdl-34234345

ABSTRACT

The majority of gene transcripts generated by RNA polymerase II in mammalian genomes initiate at CpG island (CGI) promoters1,2, yet our understanding of their regulation remains limited. This is in part due to the incomplete information that we have on transcription factors, their DNA-binding motifs and which genomic binding sites are functional in any given cell type3-5. In addition, there are orphan motifs without known binders, such as the CGCG element, which is associated with highly expressed genes across human tissues and enriched near the transcription start site of a subset of CGI promoters6-8. Here we combine single-molecule footprinting with interaction proteomics to identify BTG3-associated nuclear protein (BANP) as the transcription factor that binds this element in the mouse and human genome. We show that BANP is a strong CGI activator that controls essential metabolic genes in pluripotent stem and terminally differentiated neuronal cells. BANP binding is repelled by DNA methylation of its motif in vitro and in vivo, which epigenetically restricts most binding to CGIs and accounts for differential binding at aberrantly methylated CGI promoters in cancer cells. Upon binding to an unmethylated motif, BANP opens chromatin and phases nucleosomes. These findings establish BANP as a critical activator of a set of essential genes and suggest a model in which the activity of CGI promoters relies on methylation-sensitive transcription factors that are capable of chromatin opening.


Subject(s)
Cell Cycle Proteins/metabolism , Chromatin Assembly and Disassembly , Chromatin/genetics , Chromatin/metabolism , CpG Islands/genetics , DNA-Binding Proteins/metabolism , Nuclear Proteins/metabolism , Animals , Base Sequence , Cell Line, Tumor , Chromatin/chemistry , Chromatin Assembly and Disassembly/genetics , DNA Methylation , Gene Expression Regulation , Genes, Essential , Humans , Mice , Single Molecule Imaging
6.
EMBO J ; 40(12): e106818, 2021 06 15.
Article in English | MEDLINE | ID: mdl-33909924

ABSTRACT

Mouse embryonic stem cells (mESCs) are biased toward producing embryonic rather than extraembryonic endoderm fates. Here, we identify the mechanism of this barrier and report that the histone deacetylase Hdac3 and the transcriptional corepressor Dax1 cooperatively limit the lineage repertoire of mESCs by silencing an enhancer of the extraembryonic endoderm-specifying transcription factor Gata6. This restriction is opposed by the pluripotency transcription factors Nr5a2 and Esrrb, which promote cell type conversion. Perturbation of the barrier extends mESC potency and allows formation of 3D spheroids that mimic the spatial segregation of embryonic epiblast and extraembryonic endoderm in early embryos. Overall, this study shows that transcriptional repressors stabilize pluripotency by biasing the equilibrium between embryonic and extraembryonic lineages that is hardwired into the mESC transcriptional network.


Subject(s)
DAX-1 Orphan Nuclear Receptor , Histone Deacetylases , Mouse Embryonic Stem Cells/cytology , Animals , Cell Differentiation , Cells, Cultured , DAX-1 Orphan Nuclear Receptor/genetics , DAX-1 Orphan Nuclear Receptor/metabolism , Female , GATA6 Transcription Factor/genetics , Histone Deacetylases/genetics , Histone Deacetylases/metabolism , Male , Mice , RNA, Small Interfering/genetics , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Cytoplasmic and Nuclear/metabolism , Receptors, Estrogen/genetics , Receptors, Estrogen/metabolism
7.
Nat Commun ; 11(1): 2680, 2020 05 29.
Article in English | MEDLINE | ID: mdl-32471981

ABSTRACT

DNA methylation is considered a stable epigenetic mark, yet methylation patterns can vary during differentiation and in diseases such as cancer. Local levels of DNA methylation result from opposing enzymatic activities, the rates of which remain largely unknown. Here we developed a theoretical and experimental framework enabling us to infer methylation and demethylation rates at 860,404 CpGs in mouse embryonic stem cells. We find that enzymatic rates can vary as much as two orders of magnitude between CpGs with identical steady-state DNA methylation. Unexpectedly, de novo and maintenance methylation activity is reduced at transcription factor binding sites, while methylation turnover is elevated in transcribed gene bodies. Furthermore, we show that TET activity contributes substantially more than passive demethylation to establishing low methylation levels at distal enhancers. Taken together, our work unveils a genome-scale map of methylation kinetics, revealing highly variable and context-specific activity for the DNA methylation machinery.


Subject(s)
CpG Islands/genetics , DNA (Cytosine-5-)-Methyltransferase 1/metabolism , DNA Demethylation , DNA Methylation/genetics , DNA-Binding Proteins/metabolism , Mouse Embryonic Stem Cells/metabolism , Proto-Oncogene Proteins/metabolism , Animals , Binding Sites/genetics , Cell Line , Chromosome Mapping , DNA (Cytosine-5-)-Methyltransferase 1/genetics , DNA (Cytosine-5-)-Methyltransferases/genetics , DNA (Cytosine-5-)-Methyltransferases/metabolism , DNA Methyltransferase 3A , DNA-Binding Proteins/genetics , Dioxygenases/genetics , Dioxygenases/metabolism , Epigenesis, Genetic/genetics , Genome/genetics , Histones/metabolism , Mice , Mice, Inbred C57BL , Proto-Oncogene Proteins/genetics , Regulatory Sequences, Nucleic Acid/genetics , Transcription Factors/metabolism , Transcription, Genetic/genetics , DNA Methyltransferase 3B
8.
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
9.
Nat Struct Mol Biol ; 26(6): 471-480, 2019 06.
Article in English | MEDLINE | ID: mdl-31133702

ABSTRACT

Current understanding of chromosome folding is largely reliant on chromosome conformation capture (3C)-based experiments, where chromosomal interactions are detected as ligation products after chromatin crosslinking. To measure chromosome structure in vivo, quantitatively and without crosslinking and ligation, we implemented a modified version of DNA adenine methyltransferase identification (DamID) named DamC, which combines DNA methylation-based detection of chromosomal interactions with next-generation sequencing and biophysical modeling of methylation kinetics. DamC performed in mouse embryonic stem cells provides the first in vivo validation of the existence of topologically associating domains (TADs), CTCF loops and confirms 3C-based measurements of the scaling of contact probabilities. Combining DamC with transposon-mediated genomic engineering shows that new loops can be formed between ectopic and endogenous CTCF sites, which redistributes physical interactions within TADs. DamC provides the first crosslinking- and ligation-free demonstration of the existence of key structural features of chromosomes and provides novel insights into how chromosome structure within TADs can be manipulated.


Subject(s)
CCCTC-Binding Factor/metabolism , Chromatin/metabolism , DNA Methylation , Site-Specific DNA-Methyltransferase (Adenine-Specific)/metabolism , Animals , Bacterial Proteins/metabolism , Cell Line , Chromatin/chemistry , Chromosomes/chemistry , Chromosomes/metabolism , Mice , Mouse Embryonic Stem Cells/chemistry , Mouse Embryonic Stem Cells/metabolism , Nucleic Acid Conformation , Recombinant Fusion Proteins/metabolism
10.
Cell Stem Cell ; 24(2): 257-270.e8, 2019 02 07.
Article in English | MEDLINE | ID: mdl-30595499

ABSTRACT

Self-renewal and differentiation of pluripotent murine embryonic stem cells (ESCs) is regulated by extrinsic signaling pathways. It is less clear whether cellular metabolism instructs developmental progression. In an unbiased genome-wide CRISPR/Cas9 screen, we identified components of a conserved amino-acid-sensing pathway as critical drivers of ESC differentiation. Functional analysis revealed that lysosome activity, the Ragulator protein complex, and the tumor-suppressor protein Folliculin enable the Rag GTPases C and D to bind and seclude the bHLH transcription factor Tfe3 in the cytoplasm. In contrast, ectopic nuclear Tfe3 represses specific developmental and metabolic transcriptional programs that are associated with peri-implantation development. We show differentiation-specific and non-canonical regulation of Rag GTPase in ESCs and, importantly, identify point mutations in a Tfe3 domain required for cytoplasmic inactivation as potentially causal for a human developmental disorder. Our work reveals an instructive and biomedically relevant role of metabolic signaling in licensing embryonic cell fate transitions.


Subject(s)
Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Cell Differentiation , Lysosomes/metabolism , Signal Transduction , Alleles , Animals , Cell Self Renewal , Female , GTP Phosphohydrolases/metabolism , Genome , Humans , Male , Mice , Mouse Embryonic Stem Cells/cytology , Mouse Embryonic Stem Cells/metabolism , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Phosphorylation , Point Mutation/genetics , Protein Binding , Transcription, Genetic
11.
Cell Syst ; 7(1): 63-76.e12, 2018 07 25.
Article in English | MEDLINE | ID: mdl-30031774

ABSTRACT

Pluripotency is accompanied by the erasure of parental epigenetic memory, with naïve pluripotent cells exhibiting global DNA hypomethylation both in vitro and in vivo. Exit from pluripotency and priming for differentiation into somatic lineages is associated with genome-wide de novo DNA methylation. We show that during this phase, co-expression of enzymes required for DNA methylation turnover, DNMT3s and TETs, promotes cell-to-cell variability in this epigenetic mark. Using a combination of single-cell sequencing and quantitative biophysical modeling, we show that this variability is associated with coherent, genome-scale oscillations in DNA methylation with an amplitude dependent on CpG density. Analysis of parallel single-cell transcriptional and epigenetic profiling provides evidence for oscillatory dynamics both in vitro and in vivo. These observations provide insights into the emergence of epigenetic heterogeneity during early embryo development, indicating that dynamic changes in DNA methylation might influence early cell fate decisions.


Subject(s)
DNA Methylation/physiology , Gene Expression Regulation, Developmental/genetics , Pluripotent Stem Cells/metabolism , Animals , Cell Differentiation , Cellular Reprogramming , CpG Islands/genetics , DNA (Cytosine-5-)-Methyltransferases/metabolism , DNA Methylation/genetics , Embryo, Mammalian/cytology , Epigenesis, Genetic/genetics , Epigenomics , Genome , Genomic Imprinting , Germ Cells/metabolism , Male , Mice , Mice, Inbred C57BL , Mouse Embryonic Stem Cells/physiology , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/physiology
12.
Methods Mol Biol ; 1712: 87-95, 2018.
Article in English | MEDLINE | ID: mdl-29224070

ABSTRACT

DNA methylation is an epigenetic mark implicated in the regulation of key biological processes. Using high-throughput sequencing technologies and bisulfite-based approaches, it is possible to obtain comprehensive genome-wide maps of the mammalian DNA methylation landscape with a single-nucleotide resolution and absolute quantification. However, these methods were only applicable to bulk populations of cells. Here, we present a protocol to perform whole-genome bisulfite sequencing on single cells (scBS-Seq) using a post-bisulfite adapter tagging approach. In this method, bisulfite treatment is performed prior to library generation in order to both convert unmethylated cytosines and fragment DNA to an appropriate size. Then DNA fragments are pre-amplified with concomitant integration of the sequencing adapters, and libraries are subsequently amplified and indexed by PCR. Using scBS-Seq we can accurately measure DNA methylation at up to 50% of individual CpG sites and 70% of CpG islands.


Subject(s)
DNA Methylation/genetics , Genome-Wide Association Study/methods , Single-Cell Analysis/methods , Sulfites/chemistry , Animals , Cytosine/chemistry , DNA/chemistry , Epigenomics , Gene Library , High-Throughput Nucleotide Sequencing , Humans , Mice , Sequence Analysis, DNA , Whole Genome Sequencing
13.
Methods Mol Biol ; 1708: 161-169, 2018.
Article in English | MEDLINE | ID: mdl-29224144

ABSTRACT

The epigenetic mark 5-methylcytosine confers heritable regulation of gene expression that can be dynamically modulated during transitions in cell fate. With the development of high-throughput sequencing technologies, it is now possible to obtain comprehensive genome-wide maps of the mammalian DNA methylation landscape, but the application of these techniques to limited material remains challenging. Here, we present an optimized protocol to perform whole-genome bisulfite sequencing on low inputs (100-5000 somatic cells) using a post-bisulfite adapter tagging approach. In this strategy, bisulfite treatment is performed prior to library generation in order to both convert unmethylated cytosines and fragment DNA to an appropriate size. Then sequencing adapters are added by complementary strand synthesis using random tetramer priming, and libraries are subsequently amplified by PCR.


Subject(s)
5-Methylcytosine/analysis , High-Throughput Nucleotide Sequencing/methods , Whole Genome Sequencing/methods , Animals , CpG Islands , DNA Methylation , Epigenesis, Genetic , Gene Library , Mice , Sample Size , Sulfites
14.
Epigenetics Chromatin ; 10: 25, 2017.
Article in English | MEDLINE | ID: mdl-28507606

ABSTRACT

BACKGROUND: Gametogenesis in mammals entails profound re-patterning of the epigenome. In the female germline, DNA methylation is acquired late in oogenesis from an essentially unmethylated baseline and is established largely as a consequence of transcription events. Molecular and functional studies have shown that imprinted genes become methylated at different times during oocyte growth; however, little is known about the kinetics of methylation gain genome wide and the reasons for asynchrony in methylation at imprinted loci. RESULTS: Given the predominant role of transcription, we sought to investigate whether transcription timing is rate limiting for de novo methylation and determines the asynchrony of methylation events. Therefore, we generated genome-wide methylation and transcriptome maps of size-selected, growing oocytes to capture the onset and progression of methylation. We find that most sequence elements, including most classes of transposable elements, acquire methylation at similar rates overall. However, methylation of CpG islands (CGIs) is delayed compared with the genome average and there are reproducible differences amongst CGIs in onset of methylation. Although more highly transcribed genes acquire methylation earlier, the major transitions in the oocyte transcriptome occur well before the de novo methylation phase, indicating that transcription is generally not rate limiting in conferring permissiveness to DNA methylation. Instead, CGI methylation timing negatively correlates with enrichment for histone 3 lysine 4 (H3K4) methylation and dependence on the H3K4 demethylases KDM1A and KDM1B, implicating chromatin remodelling as a major determinant of methylation timing. We also identified differential enrichment of transcription factor binding motifs in CGIs acquiring methylation early or late in oocyte growth. By combining these parameters into multiple regression models, we were able to account for about a fifth of the variation in methylation timing of CGIs. Finally, we show that establishment of non-CpG methylation, which is prevalent in fully grown oocytes, and methylation over non-transcribed regions, are later events in oogenesis. CONCLUSIONS: These results do not support a major role for transcriptional transitions in the time of onset of DNA methylation in the oocyte, but suggest a model in which sequences least dependent on chromatin remodelling are the earliest to become permissive for methylation.


Subject(s)
DNA (Cytosine-5-)-Methyltransferases/genetics , DNA Methylation/genetics , Oocytes/growth & development , Oogenesis/genetics , Transcription, Genetic , Animals , Chromatin/genetics , Chromatin Assembly and Disassembly , CpG Islands/genetics , Female , Genomic Imprinting/genetics , Germ Cells , Histones/genetics , Mice , Oocytes/metabolism , Transcriptome/genetics
15.
Nat Protoc ; 12(3): 534-547, 2017 03.
Article in English | MEDLINE | ID: mdl-28182018

ABSTRACT

DNA methylation (DNAme) is an important epigenetic mark in diverse species. Our current understanding of DNAme is based on measurements from bulk cell samples, which obscures intercellular differences and prevents analyses of rare cell types. Thus, the ability to measure DNAme in single cells has the potential to make important contributions to the understanding of several key biological processes, such as embryonic development, disease progression and aging. We have recently reported a method for generating genome-wide DNAme maps from single cells, using single-cell bisulfite sequencing (scBS-seq), allowing the quantitative measurement of DNAme at up to 50% of CpG dinucleotides throughout the mouse genome. Here we present a detailed protocol for scBS-seq that includes our most recent developments to optimize recovery of CpGs, mapping efficiency and success rate; reduce hands-on time; and increase sample throughput with the option of using an automated liquid handler. We provide step-by-step instructions for each stage of the method, comprising cell lysis and bisulfite (BS) conversion, preamplification and adaptor tagging, library amplification, sequencing and, lastly, alignment and methylation calling. An individual with relevant molecular biology expertise can complete library preparation within 3 d. Subsequent computational steps require 1-3 d for someone with bioinformatics expertise.


Subject(s)
DNA Methylation/drug effects , Genomics/methods , Sequence Analysis, DNA/methods , Single-Cell Analysis/methods , Sulfites/pharmacology , Animals , Base Sequence , CpG Islands/genetics , Mice
16.
Sci Rep ; 6: 31640, 2016 08 19.
Article in English | MEDLINE | ID: mdl-27538787

ABSTRACT

Inflammatory bowel disease (IBD) arises by unknown environmental triggers in genetically susceptible individuals. Epigenetic regulation of gene expression may integrate internal and external influences and may thereby modulate disease susceptibility. Epigenetic modification may also affect the germ-line and in certain contexts can be inherited to offspring. This study investigates epigenetic alterations consequent to experimental murine colitis induced by dextran sodium sulphate (DSS), and their paternal transmission to offspring. Genome-wide methylome- and transcriptome-profiling of intestinal epithelial cells (IECs) and sperm cells of males of the F0 generation, which received either DSS and consequently developed colitis (F0(DSS)), or non-supplemented tap water (F0(Ctrl)) and hence remained healthy, and of their F1 offspring was performed using reduced representation bisulfite sequencing (RRBS) and RNA-sequencing (RNA-Seq), respectively. Offspring of F0(DSS) males exhibited aberrant methylation and expression patterns of multiple genes, including Igf1r and Nr4a2, which are involved in energy metabolism. Importantly, DSS colitis in F0(DSS) mice was associated with decreased body weight at baseline of their F1 offspring, and these F1 mice exhibited increased susceptibility to DSS-induced colitis compared to offspring from F0(Ctrl) males. This study hence demonstrates epigenetic transmissibility of metabolic and inflammatory traits resulting from experimental colitis.


Subject(s)
Colitis/genetics , DNA Methylation , Epigenesis, Genetic , Genetic Predisposition to Disease , Inflammatory Bowel Diseases/genetics , Animals , Colitis/chemically induced , Disease Models, Animal , Female , Genome-Wide Association Study , Inflammatory Bowel Diseases/chemically induced , Male , Mice
17.
Genome Biol ; 17: 72, 2016 Apr 18.
Article in English | MEDLINE | ID: mdl-27091476

ABSTRACT

Emerging single-cell epigenomic methods are being developed with the exciting potential to transform our knowledge of gene regulation. Here we review available techniques and future possibilities, arguing that the full potential of single-cell epigenetic studies will be realized through parallel profiling of genomic, transcriptional, and epigenetic information.


Subject(s)
Epigenomics/methods , Single-Cell Analysis/methods , Animals , DNA Methylation , Gene Expression Regulation , Humans
18.
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
20.
Genes Dev ; 29(23): 2449-62, 2015 Dec 01.
Article in English | MEDLINE | ID: mdl-26584620

ABSTRACT

Erasure and subsequent reinstatement of DNA methylation in the germline, especially at imprinted CpG islands (CGIs), is crucial to embryogenesis in mammals. The mechanisms underlying DNA methylation establishment remain poorly understood, but a number of post-translational modifications of histones are implicated in antagonizing or recruiting the de novo DNA methylation complex. In mouse oogenesis, DNA methylation establishment occurs on a largely unmethylated genome and in nondividing cells, making it a highly informative model for examining how histone modifications can shape the DNA methylome. Using a chromatin immunoprecipitation (ChIP) and genome-wide sequencing (ChIP-seq) protocol optimized for low cell numbers and novel techniques for isolating primary and growing oocytes, profiles were generated for histone modifications implicated in promoting or inhibiting DNA methylation. CGIs destined for DNA methylation show reduced protective H3K4 dimethylation (H3K4me2) and trimethylation (H3K4me3) in both primary and growing oocytes, while permissive H3K36me3 increases specifically at these CGIs in growing oocytes. Methylome profiling of oocytes deficient in H3K4 demethylase KDM1A or KDM1B indicated that removal of H3K4 methylation is necessary for proper methylation establishment at CGIs. This work represents the first systematic study performing ChIP-seq in oocytes and shows that histone remodeling in the mammalian oocyte helps direct de novo DNA methylation events.


Subject(s)
DNA Methylation , Histone Code , Oocytes/enzymology , Oogenesis/physiology , Animals , Chromatin Immunoprecipitation , CpG Islands , Flow Cytometry , Histone Demethylases/genetics , Histones/metabolism , Mice , Oxidoreductases, N-Demethylating/genetics , Sequence Analysis, DNA
SELECTION OF CITATIONS
SEARCH DETAIL
...