Abstract
In mammals, the canonical somatic DNA methylation landscape is established upon specification of the embryo proper and subsequently disrupted within many cancer types1-4. However, the underlying mechanisms that direct this genome-scale transformation remain elusive, with no clear model for its systematic acquisition or potential developmental utility5,6. Here we analyzed global remethylation from the mouse preimplantation embryo into the early epiblast and extraembryonic ectoderm. We show that these two states acquire highly divergent genomic distributions with substantial disruption of bimodal, CpG density-dependent methylation in the placental progenitor7,8. The extraembryonic epigenome includes specific de novo methylation at hundreds of embryonically-protected CpG island promoters particularly those that are associated with key developmental regulators and orthologously methylated across most human cancer types9. Our data suggest that the evolutionary innovation of extraembryonic tissues may have required cooption of DNA methylation-based suppression as an alternative to the embryonically utilized Polycomb group proteins, which coordinate germlayer formation in response to extraembryonic cues10. Moreover, we establish that this decision is made deterministically downstream of promiscuously utilized, and frequently oncogenic, signaling pathways via a novel combination of epigenetic cofactors. Methylation of developmental gene promoters during tumorigenesis may therefore reflect the misappropriation of an innate trajectory and the spontaneous reacquisition of a latent, developmentally-encoded epigenetic landscape.
To compare how epigenetic landscapes subsequently evolve during early mammalian development, we generated whole genome bisulfite sequencing (WGBS) and RNA-seq datasets from mouse precompacted 8-cell stage embryos, Inner Cell Mass (ICM) and Trophectoderm from E3.5 blastocysts, as well as Epiblast and Extraembryonic Ectoderm (E×E) from E6.5 conceptuses, the latest stage where these major developmental progenitors remain largely homogeneous and undifferentiated (Fig. 1a, Extended Data Fig. 1, Supplementary Tables 1 and 2). Holistically, our time series captures the expected transition through the indistinguishably hypomethylated, but transcriptionally distinct, blastocyst-stage tissues, followed by a considerable departure at implantation, where ∼80% of the genome becomes differentially methylated (Extended Data Fig. 2a). Specifically, the extraembryonic lineage lacks canonical bimodality: most CpGs are moderately less methylated than in epiblast, while 1.36% are specifically methylated (Extended Data Fig. 2b). E×E-specific hypo or hypermethylation segregate into distinct genomic compartments by CpG density and location, with de novo methylation preferentially enriched for CGIs near transcription start sites (TSSs) and 5′ exons (Fig. 1c, Extended Data Fig. 2c-f). Once established, these alternative landscapes are largely preserved across embryonic tissues and placenta, respectively11,12 (Extended Data Fig. 2g).
Intriguingly, E×E-methylated CGIs (E×E Hyper CGIs) frequently overlap with PRC2-regulated genes, including master transcription factors that direct germlayer and body-axis formation (Fig. 1e, Extended Data Fig. 3a,b, Supplementary Table 1). Although the majority are not yet expressed in the epiblast, E×E-specific promoter methylation is generally associated with repression, including of many pluripotency-specific regulators, as well as concurrent loss of chromatin accessibility (Fig. 1e, Extended Data Fig. 3, 4). The lower global methylation in E×E also results in a more abrupt relationship between promoter-methylation and gene-repression that is apparent at levels as low as 0.1 (Extended Data Fig. 4c). DNA methylation surrounding these promoters is largely dispersive, with flanking regions less methylated in E×E than Epiblast, but a maximal increase specifically at the TSS (Fig. 1d). Moreover, while de novo CGI methylation only reaches ∼0.25, methylated CpGs are distributed across 80% of the unique sequencing reads that fall within them, with a median per read methylation status of 0.25 (Extended Data Fig. 4d). The consistency between per molecule and aggregate methylation measurements can only be explained by population-wide recruitment of de novo methyltransferases, followed by stochastic gains at individual CpGs in phase, similar to a variety of cancer systems13,14. Importantly, the higher CpG density of E×E-targeted regions invariably leads to a higher local methylation density, even though the per-CpG methylation status is intermediate (Fig. 1e).
Suppression overlaps with WNT pathway effectors that are induced in the proximal epiblast to promote primitive streak formation (Fig. 2a). However, E×E expresses alternative Wnt proteins, suppresses Fgf loci by de novo methylation, and specifically expresses receptors for epiblast-secreted factors (Fig. 2b, Extended Data Fig. 5a). The extraembryonic landscape may proceed deterministically from these two major signaling pathways, which are promiscuously utilized in many downstream developmental processes and frequently misregulated in cancers. To investigate this hypothesis, we selected the ICM as a model because it is indistinguishably hypomethylated from TE and can be cultured independently of FGF, whereas extraembryonic development rapidly attenuates15. ICMs were cultured in four conditions using combinations of FGF4, the Mitogen-Activated Protein Kinase Kinase (MAPKK or MEK) inhibitor PD0325901 (PD), and the GSK3β inhibitor, WNT agonist CHIR99021 (CHIR) (Fig. 2c, Extended Data Fig. 5b). Isolated outgrowths were dually assayed by a combined RNA-seq and reduced representation bisulfite sequencing (RRBS) approach (Extended Data Fig. 6, Methods). Those cultured in FGF4+CHIR progressively diverged into two separate, morphologically distinguishable interior and exterior tissues that were independently isolated.
In combination, PD and CHIR comprise the “2i” condition, an FGF-impeded, WNT-activated state that maintains preimplantation-like global hypomethylation16. Alternatively, exogenous FGF is sufficient to drive genome and CGI methylation to nonphysiologically high levels (Fig. 2d). Surprisingly, when coupled with FGF, WNT agonism effectively blocks genome remethylation but redirects CGI-level methylation to a broader subset of extraembryonic targets (Fig. 2e, f). Targeting is specific to the FGF+CHIR outgrowth exterior, which establishes an asymmetric Fgfr2 and Fgf4 expression pattern with the interior, similar to what occurs in vivo (Supplementary Tables 3, 4). The specific overlap between in vitro methylated CGIs and E×E appears to reflect progressive restriction of potential targets over early development: those shared across conditions have early developmental functions and are often expressed in the ICM and 2i condition, those methylated in E×E and FGF+CHIR, but not in FGF alone, generally encompass neuroectodermal regulators, and E×E-exclusive islands are often endodermal and induced by dual FGF and WNT activity (Extended Data Fig. 5c). Seemingly, E×E-like global hypomethylation and CGI methylation can be recapitulated in vitro by WNT and FGF, but target-specificity can be modulated to include multiple discrete developmental programs.
We next sought to investigate the configuration of epigenetic regulators that specifically E×Ecute this transition. While Dnmt1 and Dnmt3b are expressed in both tissues, Dnmt3l and Dnmt3a isoform 2 (Dnmt3a2) are reciprocally expressed in either the E×E or epiblast and regulated by de novo methylation in the alternate (Extended Data Fig. 7a-d). A truncated, non-catalytic isoform of the H3K36 demethylase Kdm2b is expressed over preimplantation and within E×E, while a longer Jumonji (JMJ) demethylase domain-containing isoform is specifically induced in epiblast17 (Extended Data Fig. 7e). Otherwise, epigenetic regulator expression is relatively stable between the two tissues at this time, such that their specific integration could explain the assembly of such profoundly different landscapes. To compare their capacity to direct both global and CGI methylation, we acutely disrupted Dnmt1, 3a, 3b, and 3l, the essential PRC2 component Eed, and Kdm2b by CRISPR/Cas9 injection into zygotes (Supplementary Tables 5 and 6, Methods). We find that Dnmt1, 3b and 3l ablation substantially disrupt the E×E methylome, including at CGI targets, but show no specificity for these regions or corresponding changes to expression (Fig. 3a, b, Extended Data Fig. 7f). The near complete loss of methylation in Dnmt1-null E×E compared to epiblast indicates diminished de novo activity, and greater reliance on epigenetic maintenance, despite prolonged Dnmt3l expression (Fig. 3a, b). Alternatively, Eed-null E×E disrupts CGI methylation without affecting global levels, suggesting that PRC2 specifically coordinates repression upstream of DNMT3B as part of a novel developmental pathway (Fig. 3c, d, Extended Data Fig. 7g). Consistently, Eed-null E×E fails to suppress associated genes, which are induced to levels similar to those of sample-matched epiblast (Extended Data Fig. 7h).
Our data indicate a point in early development where sensitivity to promiscuously utilized growth factors instructs a novel epigenome that is not observed during ontogeny. However, de novo CGI methylation is also a general feature of tissue culture, cancer cell lines, and primary tumors, suggesting that somatic cells remain vulnerable5,18 (Fig. 4a, Extended Data Figs. 8-10). To investigate a possible link with the subsequent reemergence of this landscape in cancer, we mapped orthologous CGIs to directly compare extraembryonically-methylated CGIs across patient-matched DNA methylation profiles from The Cancer Genome Atlas (TCGA) project, an age-matched Chronic Lymphocytic Leukemia (CLL) cohort, as well as data from ENCODE and the Roadmap Epigenomics Project14,19-21. Of the 16 tumor types with sufficient normal biopsied samples, 15 significantly methylate E×E Hyper CGIs (Fig. 4a, b). The signal is surprisingly robust and segregates tumor and normal tissue when measured as a feature across patients or when examining CGI-level changes (Fig. 4b, c, Extended Data Fig. 8). 84% of E×E Hyper CGIs are methylated in at least one cancer type, and they are more frequently shared as conserved, pan-cancer targets (Fig. 4d). We find some direct and indirect evidence that CGI methylation can be influenced by FGF sensing. For example, matched mutational and methylation analyses of the entire TCGA data set (n=10,629 tumors) shows a 5.3 percentage point increase in the average methylation of E×E Hyper CGIs when any FGF pathway member is mutated (Extended Data Fig. 10). Similarly, statistical assessment of the connectivity between our E×E Hyper CGIs and the ten most mutated pathways in cancer reveals a striking enrichment for FGFR signaling in disease (Enrichment Z-score=3.88, Extended Data Fig. 10). Over the more expansive, but less internally controlled, ENCODE and ROADMAP data, cancers and immortalized cell lines clearly separate from primary tissues by E×E Hyper CGI methylation status (Fig. 4e, Supplementary Table 7). Intriguingly, mature adaptive immune cells and endodermal lineages are generally more susceptible to low level methylation within these regions, suggesting a preexisting heterogeneity even in normal populations.
We present the developmental acquisition of a novel epigenetic landscape that partitions extraembryonic tissues within the embryo and resembles a frequent, global departure in genome regulation in human cancers. This landscape cooccurs with the establishment of the first major signaling axes, can be partially directed from hypomethylated ICM in vitro, and appears to be determined by disparate regulation of the DNMTs and associated cofactors. Notably, de novo methylation of CGIs in the E×E requires PRC2, which indicates either a transient, biochemical interaction with the DNMTs or an upstream, primary silencing role. The coordination of this alternative, and presumably more permanent, repressive mechanism warrants further investigation and shares notable parallels to the somatic transition to cancer. Most obviously, FGF sensing passes through RAS/MAPK/ERK signaling, which has extensive oncogenic potential and putative roles in cancer methylome establishment22-24. Similarly, the E×E displays attenuated de novo methylation activity directed wholly by DNMT3B, broadly resembling the high frequency of somatic DNMT3A mutations in Acute Myeloid Leukemia (AML) and Myelodysplastic Syndrome (MDS) or DNMT3B-directed CGI methylation during colorectal transformation25-28. Transgenic mouse cancer models confirm conserved E×E Hyper CGI methylation in similar contexts (Extended Data Fig. 10). The extraembryonic landscape depends on extrinsic cues with numerous downstream developmental functions, which may provide a latent opportunity for spontaneous state transition without genetic perturbation in later development. If so, the likelihood for such a transition may relate to how closely a given regulatory network resembles the one governing extraembryonic specification. Whether or not additional morphological and molecular features of placental development that appear analogous to cancer hallmarks29,30 – such as immunosuppression, tissue invasion, and angiogenesis – proceed as part or downstream of this primary epigenetic switch remains unexplored, but would provide a parsimonious developmental foundation to their systematic emergence during transformation.
Online Methods
Sample isolation and library preparation
Preparation of preimplantation and postimplantation samples was performed as described in Ref 31. Briefly, B6D2F1 hybrid females between 5-8 weeks old (Charles River) were serially primed with 5 IU Pregnant Mare Gonadotropin (Sigma) followed by 5 IU Human Chorionic Gonadotrophin (HCG, Millipore) after 46 hours, and subsequently mated with B6D2F1 male mice ≤6 months old. For preimplantation time points, zygotes from mated females were isolated from the oviduct the following morning (E0.5) and cultured in KSOM media (Millipore) droplets under mineral oil until E2.25. The 8 cell sample was collected by careful monitoring of 4 cell embryos from ∼E2 onwards and emergent 8 cell embryos were swapped into KSOM supplemented with 1 μg/ml aphidicolin (Sigma) to ensure synchronization and minimal entry into the fourth replication cycle. 8 cell embryos were collected within 4 hours of the first apparent embryo of this stage. Prior to collection, embryos were serially transferred through Acid Tyrode's solution (Sigma) to remove the Zona Pellucida and carefully pipetted with a drawn glass capillary through 0.25% Trypsin-EDTA (Life Technologies) to remove maternal polar bodies. E3.5 blastocysts were also treated with Acid Tyrode's solution to remove the Zona and the ICM and TE of matched samples were dissected using standard micromanipulation equipment (Eppendorf) and a Hamilton Thorne XYClone laser with 300 μs pulsing at 100% intensity. Isolation of postimplantation tissues was performed as described32. The decidua of mated female mice were isolated on the morning of E6.5 and the conceptus removed. Then, under a stereomicroscope, the embryo was carefully bisected along the extraembryonic/embryonic axis, removing the ectoplacental cone from the extraembryonic ectoderm when apparent. After separation, Epiblast and Extraembryonic Ectoderm (E×E) were incubated for 15 min at 4°C in 0.5% Trypsin, 2.5% Pancreatin dissolved in PBS and allowed to rest for 5-10 minutes in KSOM at room temperature. Finally, visceral endoderm was removed by drawing the embryo through a narrow, flame drawn glass capillary and only samples with no apparent contamination were collected. On average, matched E×E and Epiblast or ICM and TE samples from 5-10 embryos or from 20 or more 8 cell embryos were collected per assay.
DNA for Whole Genome Bisulfite Sequencing was isolated according to Ref 33 and libraries were prepared using the Accel-NGS™ Bisulfite DNA library kit (Swift Biosciences) according to the manufacturers protocol. Final libraries were generated from 10-12 PCR cycles. RNA was purified using the RNAeasy Micro Kit (Qiagen) and RNA-seq libraries generated using the SMRT-seq v4 Ultralow Input Kit (Clontech) according to the manufacturer's protocol with 10-11 LD PCR cycles. Libraries were generated from 150 pg of the subsequent cDNA using the Nextera XT DNA library preparation kit (Illumina) and 12 PCR cycles. ATAC-seq libraries were generated according to Ref 34 using a 10 μl reaction and incubation with the TN5 transposase mixture (Nextera DNA library preparation kit, Illumina) for 45 minutes. Reaction was stopped according to the protocol described in Ref 35 and purified using silane beads (Thermo Fisher). Tagmented DNA was amplified for 12-14 cycles to generate the library. WGBS libraries were sequenced as a pool using the HiSeq × ten platform (Illumina), while RNA-seq and ATAC-seq data were sequenced using the HiSeq 2500 (Illumina).
Outgrowth experiments
To generate controlled outgrowth data, ICM were immunosurgically isolated from BDF1×129S1/SvImJ strain blastocysts at 96hpf as described31. Briefly, oocytes were isolated by hormone priming from B6D2F1 females 12-14 hours after administration of hCG and fertilized by Intracytoplasmic Sperm Injection using piezo actuated injection of 129S1/SvIMJ strain sperm36. At 96 hrs post-fertilization, blastocysts were stripped of their zona pellucida by brief incubation in Acid Tyrode's solution and incubated for 30 minutes in 1:10 diluted whole mouse antisera (Sigma) in CO2 equilibrated KSOM, followed by destruction of the trophectoderm by culture in 1:10 diluted Guinea Pig Complement Sera (Sigma). After 15 minutes at 37°C, the ICM separates from the complement-lysed TE and could be cleanly isolated by brief pulsing through a narrow glass capillary. ICM were isolated in batches of ∼12 per drop. Once isolated, ICM were then plated into basal N2/B27 media supplemented with 1000 U/mL LIF (made in house) and one of the following conditions; ‘2i’ supplemented with 1 μM PD0325901 and 3 μM CHIR99021(Reagents Direct)37; ‘PD’ supplemented with 1 uM PD0325901 and 10 ng/mL BMP4 to promote outgrowth expansion (Peprotech)38; ‘FGF+CHIR’ supplemented with 25ng/mL mouse recombinant FGF4 (R and D) and 3 μM CHIR99021; and ‘FGF’ supplemented with 25ng/mL FGF4 only. FGF4 was selected because it is the most highly expressed FGF family member in the preimplantation embryo and we sought to direct specific remethylation changes as is observed in vivo. ICM were plated in gelatin treated tissue culture dishes plated with irradiated CF-1 strain embryonic fibroblasts to promote attachment. The primary outgrowth from the ICM, characterized as a centrally expanding, three-dimensional mass, was isolated after 4 days of culture. In all cases but the 2i condition, an outer layer of differentiated cells was apparent and removed using an identical strategy to removal of the extraembryonic endoderm from E6.5 samples described above. However, under the FGF+CHIR condition, the ‘outer layer’ was often of the same size or larger than the internal outgrowth and only became defined during the latter portion of culture (see Extended Data Fig. 5b). As such, we collected both interior and exterior portions as they could clearly be distinguished as mutually ICM-derived. After incubation and either isolation or removal of external cells, outgrowths were serially washed through several KSOM drops under mineral oil before being snap frozen in minimal volume for RRBS and RNA-seq profiling.
Generation of KO embryos by CRISPR/Cas9 and zygotic injection
Zygotic injection was performed essentially as described39. To improve the efficiency with which null alleles were generated, three separate guide RNA sequences were designed per target, prioritizing highly scored protospacer sequences with no high scoring off target sites using the CHOPCHOP web tool40 and at as 5′ most a coding exon as possible given these constraints. Protospacer sequences were input into the following oligonucleotide primer pair and used to amplify off of the pX300 plasmid (Addgene): Forward primer, AGTCAGTTAATACGACTCACTATAGN19GTTTTAGAGCTAGAAATAGCAAG; Reverse primer, AAAAAAAGCACCGACTCGGTGCCAC. Protospacer sequences that did not begin with a G to initiate T7 transcription were inserted and an additional 5′ G was added. 200 ng of gel purified, T7 promoter containing sgRNA templates were used to generate gRNAs by in vitro transcription using the MEGAshortscript™ T7 transcription kit (Thermo Fisher), followed by purification with phenol:chloroform and ethanol precipitation. Translation competent spCas9 RNA was in vitro transcribed off of a similarly designed, T7 promoter driven template amplified off of the pX300 plasmid using the mMessage mMachine™ T7 Ultra kit (Thermo Fisher) and purified using the RNA Clean and Concentrator Kit (Zymo Research). RNA was resuspended in an injection buffer comprised of 5 mM Tris-HCl, 0.1 mM EDTA, pH=7.4. Zygotes were isolated from hormone primed B6D2F1females mated with B6D2F1 males as described above. Shortly after the formation of visible pronuclei (Pronuclear Stage 3), zygotes were cytoplasmically injected with 100 ng/ul of all three targeted sgRNAs pooled 1:1:1 and 200 ng/ul Cas9 mRNA. At E3.5, cavitated blastocyts were transferred in clutches of 10-15 into one uterine horn of pseudopregnant CD-1 strain mice (Charles River) that had been mated with vasectomized male Swiss-Weber strain mice (Taconic) two days prior. To account for the ∼1 day offset in developmental progression that results from uterine transfer, appropriately E6.5 stage conceptuses were isolated four days after uterine transfer and epiblast and extraembryonic ectoderm tissue were isolated as described above prior to snap freezing in minimal tissue. Each replicate consisted of ≥4 embryos and all experimental series include replicates generated from at least two rounds of zygotic injection. Care was taken to ensure epiblast and extraembryonic ectoderm tissue from matched embryos were included for each replicate set and RRBS data where both fractions did not cover >1 million CpGs at ≥5× coverage each were excluded from further analysis. Disruption of the target allele was confirmed by PCR amplification from the primary cDNA using primers that flank all three protospacer sequences to capture multiple simultaneous perturbations in phase.
Dual RRBS and RNA-seq profiling
Genomic DNA and mRNA purifications from low input samples were performed as described by Maculay et al. with modifications41. Briefly, the cells were mixed with 15 μl of RLT plus buffer (QIAGen) containing 1 U/μl of RNase inhibitor (SUPERase·In, ThermoFisher Scientific), 1% β-mercaptoethanol (Sigma-Aldrich), and were then transferred to one well in a 96-well DNA LoBind plate (Eppendorf). After adding 10 μl of M-280 streptavidin bead-conjugated RT primer to each sample, the reaction was incubated at 72°C for 3 min in a thermocycler followed by incubation at room temperature for 25 min with gentle rotation. The genomic DNA and mRNA were separated in a DynaMag™-96 Side Magnet (ThermoFisher Scientific). The bead-tagged mRNA was subjected to reverse transcription as described previously41 and the genomic DNA in the supernatant was transferred to a fresh 96-well DNA LoBind plate. After reverse transcription, the cDNA was PCR amplified and RNA-seq library was generated according to the Smart-seq 2 protocol42. IndE×Ed RNA-seq libraries were pooled and sequenced in an Illumina Hiseq2500 sequencer.
Genomic DNA was isolated utilizing 1× Agencourt AMPure beads (Beckman Coulter) and was eluted to 15 μl of low TE buffer. RRBS library was generated as reported previously with modifications43. We utilized the CutSmart buffer (New England Biolabs) for all three enzymatic reactions including MspI digestion, end-repair/A-tailing and T4 DNA ligation. To minimize DNA loss, DNA purification step was eliminated after each enzymatic reaction. Briefly, the genomic DNA was digested by 16 units of MspI (New England Biolabs) for 80 min at 37°C, and followed by heat inactivation at 65°C for 15 min. The digested DNA fragments were end-repaired and A-tailed by adding 4 units of Klenow exo- (New England Biolabs), 0.03 mM dCTP, 0.03 mM dGTP and 0.3 mM dATP; ant the reaction was carried out at 30°C for 25 min; 37°C for 25 min followed by incubation at 70 °C for 10 min to inactive the enzyme. We then ligated the A-tailed DNA fragments with indE×Ed adapters overnight at 16 °C by adding 2,000 units of T4 DNA ligase and 0.75 mM ATP and 7 nM of the adapters. The T4 ligase was heat-inactivated at 65 °C for 15 min before pooling libraries together. To remove adapter dimers, the library pool was cleaned up using 1.8X AMPure beads and the adapter-tagged DNA fragments were eluted to 30 μl of low TE buffer. The bisulfite conversion of the adapter-tagged DNA fragments ware conducted using a QIAGen EpiTect Fast Bisulfite Conversion Kit following the manufacturer's instructions with a minor modification. We extended the bisulfite conversion time from 2 cycles of 10 min to 2 cycles of 20 min to achieve bisulfite conversion rates >99%. The bisulfite converted DNA fragments were PCR amplified according to the following thermocycler settings: 98 °C for 45 s, 6 cycles of 98 °C for 20 s, 58 °C for 30 s, 72 °C for 1 min and then 8-10 cycles of 98 °C for 20 s, 65 °C for 30 s, 72 °C for 1 min followed by a final extension cycle of 5 min at 72 °C. The PCR amplified library DNA was cleaned up using 1.3X AMPure beads and the RRBS libraries were paired-end sequenced for 2×100 cycles. Only instances where the matched pool of Epiblast and E×E from a given replicate both had over 1 million CpGs covered at ≥5× were included for downstream analysis.
For each sample, 10 μl of M-280 streptavidin beads (ThermoFisher Scientific) were prepared per the manufacturer's recommendations. Specifically, after washing with Solution A (0.1 N NaOH, 0.05 M NaCl) and B (0.1 M NaCl) sequentially, the beads were resuspended in 10 μl of 2× Washing and Binding buffer (10 mM Tris-HCl, 1 mM EDTA, 2 M NaCl) and then mixed with an equal volume of 2 μM of RT primer41. The mixture was incubated for 15 min at room temperature with gentle rotation. The bead-bound RT primer was collected in a magnate and was subsequently resuspended in 10 μl of binding buffer (10 mM Tris-HCl (PH 8.0), 167 mM NaCl, 0.05% Tween-20).
Estimating methylation levels
The methylation level of each sampled cytosine was estimated as the number of reads reporting a C, divided by the total number of reads reporting a C or T. Single CpG methylation levels were limited to those CpGs that had at least fivefold coverage. For 100 bp tiles, reads for all the CpGs that were covered more than fivefold within the tile were pooled and used to estimate the methylation level as described for single CpGs. The CpG density for a given single CpG is the number of CpGs 50 bp up- and downstream of that CpG. The CpG density for a 100 bp tile is the number of CpGs in the tile. The methylation level reported for a sample is the average methylation by pooling all reads across replicates.
Genomic features
LINE, LTR and SINE annotations were downloaded from the UCSC (University of California, Santa Cruz) browser (mm9) RepeatMasker tracks. CGI annotations were downloaded from the UCSC browser (mm9) CpG Islands (CGI) track. Gene annotations (Exon, 5′ Exon, Intron) were downloaded from the UCSC browser (mm9) RefSeq track. Promoters (TSSs) are defined as +/– 2 kb of the RefSeq annotation. Corresponding human annotations were downloaded from the UCSC browser for hg19. In each case, the methylation level of an individual feature is estimated by averaging methylation for all CpGs within the feature that are covered greater than fivefold. Assignment of CGIs to a given TSS (CGI promoters) included annotated CGIs that fell within this boundary. Methylation was estimated for “core TSS” sequences defined as –/+ 1 kb of the RefSeq annotation and only included CpGs measured at ≥5× in both samples (WGBS) or pooled samples (RRBS). For Fig. 2b and Extended Data Fig. 3f, and 5c, promoters for all isoforms are included and the maximally different alternative TSS was reported. Within the Supplementary Tables, the methylation levels of all annotated TSSs were calculated and reported in this manner, with the mean TMP estimate for the gene reported for all associated TSSs.
Identification of differentially methylated loci and regions
For WGBS data, identification of differentially methylated loci was performed by DSS, which use biological replicates and information from CpG sites across the genome to stabilize the estimation of the dispersion parameters44. Only CpGs that were covered at least fivefold across all samples were considered for a given comparison. An FDR cutoff of 5% was used to identify differentially methylated CpGs. A CGI was called as differentially methylated if it was covered by at least 5 CpGs and 80% of them were significantly hyper/hypo methylated. For TCGA Illumina Infinium HumanMethylation450K BeadChip data, given that most cancer types have more than 20 tumor and normal samples, Wilcoxon rank-sum test was used to identify differentially methylated CpGs, with a FDR cutoff of 5%. All statistical tests throughout this study are two sided. A CGI was called as differentially methylated if 80% of covered CpGs were significantly hyper/hypo methylated. For RRBS data, a simple cutoff of 10% difference in CGI-level methylation was used to call differential methylation.
Gene expression analysis
Alignment was performed using TopHat2 against mouse genome assembly mm9 with default settings. Isoform-level expression was quantified by kallisto, which performs pseudoalignment of reads against cDNA sequence of transcripts. Gene-level expression was estimated as sum of expression of associated isoforms. Refseq mRNA sequences were downloaded from UCSC genome browser. Expression levels were reported as Transcripts Per Million (TPM).
Pathway Enrichment
Pathway enrichment was performed by hypergeometric test using the GSEA online tool. P-value was adjusted for multiple hypothesis testing according to Benjamini and Hochberg, with 5% as a cutoff.
Connectivity analysis
We used GRAIL (Gene Relationships Among Implicated Loci, Ref 45) to test whether a query gene is functionally related a set of seed genes. GRAIL utilizes text-mining to quantify the relatedness between two genes in the genome, by which a global gene-network is built. It has been demonstrated that genes who function in the same pathway tend to distribute in a coherent sub-network. In this study, we built a sub-network using E×E Hyper CGI-associated genes, which were significantly enriched in several pathways. To predict whether a query gene is functionally related to the E×E Hyper subnetwork, we project this gene to the global network, and test whether connection of this gene to the subnetwork is random or statistically significant.
ATAC-seq data processing
Reads were aligned to mouse genome mm9 using BWA with default parameters. Duplicates were removed by function MarkDuplicates from Picard tool kit. Reads with low mapping quality (< 10) or in mitochondrial chromosome were removed. NucleoATAC was used to generate Insert density, which was normalized by the total number insertions in each sample46.
Orthology mapping between human and mouse
Mouse mm9 CGIs were mapped to human hg19 segments using liftOver with chain file mm9ToHg19.over.chain. Then human orthologous CGIs were defined as the nearest CGIs to the mapped segments.
Extended Data
Supplementary Material
Supplementary Table 1. De novo methylation of CGIs during extraembryonic development, Methylation status of CGIs in Epiblast and Extraembryonic Ectoderm (E×E), including designation of differential methylation status in E×E as described in the Methods (hyper, hypermethylated; hypo, hypomethylated; NC, no change; ND, insufficient measurements). Assignment to nearest gene and distance to the TSS are included.
Supplementary Table 2. Promoter methylation and associated transcriptional dynamics during implantation are influenced by CGI methylation status, Methylation values for gene promoters (classified as the region +/− 1 kb of an annotated TSS), Log2 normalized TPM (Transcripts per Million) across late preimplantation and early postimplantation samples. Promoter methylation is reported if at least 5 CpGs are covered ≥5×. The ‘Symbol’ column identifies all annotated genes for a given promoter and the reported expression value is either the TPM of the associated gene or the mean TPM if multiple genes begin at the same TSS. ‘CpGs’ indicates the number of CpGs that exist within the promoter boundary.
Supplementary Table 3. CGI methylation status for ICM outgrowths under defined conditions, CGI methylation status as measured by RRBS for ICM explanted under conditions of modulated FGF and WNT signaling. CGIs are assigned to their nearest TSS and those existing within +/– 2 kb were given the additional assignment of TSS-associated. DMR status indicates differential methylation between Epiblast and E×E from WGBS data, and PRC2 regulatory status is taken from Ref 67. We observe three discrete scenarios where CGIs are preferentially methylated: within the E×E, in the external portion of FGF+CHIR stimulated ICM outgrowths, and in FGF stimulated outgrowths. A CGI whose methylation status deviates by ≥0.1 from epiblast is scored as ‘dynamic’ and used to generate the heatmap in Fig. 2f.
Supplementary Table 4. Promoter methylation status and transcriptional dynamics for ICM outgrowths under defined conditions, Promoter methylation and associated gene expression data of ICM outgrowth conditions as measured by dual RRBS and RNA-seq. Promoter methylation is reported if at least 5 CpGs are covered ≥5×. The ‘Symbol’ column identifies all annotated genes for a given promoter and the reported expression value is either the TPM of the associated gene or the mean TPM if multiple genes begin at the same TSS.
Supplementary Table 5. CGIs methylation status for epigenetic regulator deficient E6.5 embryos, CGI methylation status as measured by RRBS for samples isolated from CRISPR/Cas9 injected embryos. CGIs are assigned to their nearest TSS and those existing within +/– 2 kb were given the additional assignment of TSS-associated. DMR status indicates differential methylation between Epiblast and E×E from WGBS data, and PRC2 regulatory status is taken from Ref 67. A CGI whose methylation status deviates by ≥0.1 from its wild type tissue is scored as ‘dynamic’ and is highlighted in Fig. 3d.
Supplementary Table 6. Promoter methylation status and transcriptional dynamics for epigenetic regulator deficient E6.5 embryos, Promoter methylation and associated gene expression data of CRISPR/Cas9 targeted embryos as measured by dual RRBS and RNA-seq. Promoter methylation is reported if at least 5 CpGs are covered ≥5×. The ‘Symbol’ column identifies all annotated genes for a given promoter and the reported expression value is either the TPM of the associated gene or the mean TPM if multiple genes begin at the same TSS. In general, E×E Hyper CGIs are preferentially induced in both the Epiblast and E×E fraction of Eed targeted E6.5 embryos and de novo methylation of these regions in E×E is specifically blocked.
Supplementary Table 7. Methylation status of E×E hypermethylated CGIs within human tissues, cancers, and cell lines, Mean and median methylation status of the 489 orthologously mapped CGIs that are called as E×E-hypermethylated in mouse across 107 ENCODE and Roadmap Initiative samples. Note, the lymphoblastoid cell line GM12878 is not characterized as cancer cell line within Encode but was generated using the Epstein-Barr Virus and scored as such in this study. Information includes designation as cancer versus normal as well as other assignments included in Extended Data Figure 9.
Acknowledgments
We thank members of the Meissner and Michor labs for thoughtful discussions and advice, in particular R. Karnik for assistance in data processing and alignment, as well as B.E. Bernstein and R.P. Koche for their expertise. FM and JS gratefully acknowledge support from the Dana-Farber Cancer Institute Physical Sciences-Oncology Center (NIH U54CA193461). The work was funded by the New York Stem Cell Foundation, Broad-ISF Partnership for Cell Circuit Research, Starr Foundation, NIH grants (1P50HG006193, P01GM099117, R01DA036898) and the Max Planck Society. AM is a New York Stem Cell Foundation Robertson Investigator.
Footnotes
Author contributions: Z.D.S, J.S., F.M., and A.M. designed and conceived the study and prepared the manuscript. Z.D.S performed all experiments and assisted in data analysis as performed by J.S. J.D. made the ATAC-Seq, D.C. made RNA-Seq libraries, and H.G. made the dual RRBS and RNA-seq libraries with supervision from A.G. and alignment by K.C. F.M. and A.M. jointly supervised the work.
Competing financial interest: There is NO Competing Interest.
Data accession: All data sets have been deposited in GEO and are accessible under GSE84236. Additional data include: Roadmap and ENCODE samples from RnBeads Methylome Resource (http://rnbeads.mpi-inf.mpg.de/methylomes.php), mouse adult tissues from GSE42836, and CLL and normal B lymphocytes from GSE58889.
References
- 1.Smith ZD, Meissner A. DNA methylation: roles in mammalian development. Nat Rev Genet. 2013;14:204–220. doi: 10.1038/nrg3354. [DOI] [PubMed] [Google Scholar]
- 2.Ohm JE, et al. A stem cell-like chromatin pattern may predispose tumor suppressor genes to DNA hypermethylation and heritable silencing. Nat Genet. 2007;39:237–242. doi: 10.1038/ng1972. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Schlesinger Y, et al. Polycomb-mediated methylation on Lys27 of histone H3 pre-marks genes for de novo methylation in cancer. Nat Genet. 2007;39:232–236. doi: 10.1038/ng1950. [DOI] [PubMed] [Google Scholar]
- 4.Widschwendter M, et al. Epigenetic stem cell signature in cancer. Nat Genet. 2007;39:157–158. doi: 10.1038/ng1941. [DOI] [PubMed] [Google Scholar]
- 5.Feinberg AP, Ohlsson R, Henikoff S. The epigenetic progenitor origin of human cancer. Nat Rev Genet. 2006;7:21–33. doi: 10.1038/nrg1748. [DOI] [PubMed] [Google Scholar]
- 6.Flavahan WA, Gaskell E, Bernstein BE. Epigenetic plasticity and the hallmarks of cancer. Science. 2017;357 doi: 10.1126/science.aal2380. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Schroeder DI, et al. The human placenta methylome. Proc Natl Acad Sci U S A. 2013;110:6037–6042. doi: 10.1073/pnas.1215145110. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Branco MR, et al. Maternal DNA Methylation Regulates Early Trophoblast Development. Dev Cell. 2016;36:152–163. doi: 10.1016/j.devcel.2015.12.027. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Deaton AM, Bird A. CpG islands and the regulation of transcription. Genes Dev. 2011;25:1010–1022. doi: 10.1101/gad.2037511. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Arnold SJ, Robertson EJ. Making a commitment: cell lineage allocation and axis patterning in the early mouse embryo. Nat Rev Mol Cell Biol. 2009;10:91–103. doi: 10.1038/nrm2618. [DOI] [PubMed] [Google Scholar]
- 11.Hon GC, et al. Epigenetic memory at embryonic enhancers identified in DNA methylation maps from adult mouse tissues. Nat Genet. 2013;45:1198–1206. doi: 10.1038/ng.2746. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Ziller MJ, et al. Charting a dynamic DNA methylation landscape of the human genome. Nature. 2013;500:477–481. doi: 10.1038/nature12433. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Landan G, et al. Epigenetic polymorphism and the stochastic formation of differentially methylated regions in normal and cancerous tissues. Nat Genet. 2012;44:1207–1214. doi: 10.1038/ng.2442. [DOI] [PubMed] [Google Scholar]
- 14.Landau DA, et al. Locally disordered methylation forms the basis of intratumor methylome variation in chronic lymphocytic leukemia. Cancer Cell. 2014;26:813–825. doi: 10.1016/j.ccell.2014.10.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Arman E, Haffner-Krausz R, Chen Y, Heath JK, Lonai P. Targeted disruption of fibroblast growth factor (FGF) receptor 2 suggests a role for FGF signaling in pregastrulation mammalian development. Proc Natl Acad Sci U S A. 1998;95:5082–5087. doi: 10.1073/pnas.95.9.5082. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Leitch HG, et al. Naive pluripotency is associated with global DNA hypomethylation. Nat Struct Mol Biol. 2013;20:311–316. doi: 10.1038/nsmb.2510. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Boulard M, Edwards JR, Bestor TH. Abnormal X chromosome inactivation and sex-specific gene dysregulation after ablation of FBXL10. Epigenetics Chromatin. 2016;9:22. doi: 10.1186/s13072-016-0069-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Meissner A, et al. Genome-scale DNA methylation maps of pluripotent and differentiated cells. Nature. 2008;454:766–770. doi: 10.1038/nature07107. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Consortium EP. An integrated encyclopedia of DNA elements in the human genome. Nature. 2012;489:57–74. doi: 10.1038/nature11247. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Hoadley KA, et al. Multiplatform analysis of 12 cancer types reveals molecular classification within and across tissues of origin. Cell. 2014;158:929–944. doi: 10.1016/j.cell.2014.06.049. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Roadmap Epigenomics, C. et al. Integrative analysis of 111 reference human epigenomes. Nature. 2015;518:317–330. doi: 10.1038/nature14248. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.MacLeod AR, Rouleau J, Szyf M. Regulation of DNA methylation by the Ras signaling pathway. J Biol Chem. 1995;270:11327–11337. doi: 10.1074/jbc.270.19.11327. [DOI] [PubMed] [Google Scholar]
- 23.Lu CW, et al. Ras-MAPK signaling promotes trophectoderm formation from embryonic stem cells and mouse embryos. Nat Genet. 2008;40:921–926. doi: 10.1038/ng.173. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Serra RW, Fang M, Park SM, Hutchinson L, Green MRA. KRAS-directed transcriptional silencing pathway that mediates the CpG island methylator phenotype. Elife. 2014;3:e02313. doi: 10.7554/eLife.02313. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Rhee I, et al. DNMT1 and DNMT3b cooperate to silence genes in human cancer cells. Nature. 2002;416:552–556. doi: 10.1038/416552a. [DOI] [PubMed] [Google Scholar]
- 26.Lin H, et al. Suppression of intestinal neoplasia by deletion of Dnmt3b. Mol Cell Biol. 2006;26:2976–2983. doi: 10.1128/MCB.26.8.2976-2983.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Ley TJ, et al. DNMT3A mutations in acute myeloid leukemia. N Engl J Med. 2010;363:2424–2433. doi: 10.1056/NEJMoa1005143. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Walter MJ, et al. Recurrent DNMT3A mutations in patients with myelodysplastic syndromes. Leukemia. 2011;25:1153–1158. doi: 10.1038/leu.2011.44. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Novakovic B, Saffery R. Placental pseudo-malignancy from a DNA methylation perspective: unanswered questions and future directions. Front Genet. 2013;4:285. doi: 10.3389/fgene.2013.00285. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell. 2011;144:646–674. doi: 10.1016/j.cell.2011.02.013. [DOI] [PubMed] [Google Scholar]
- 31.Smith ZD, et al. DNA methylation dynamics of the human preimplantation embryo. Nature. 2014;511:611–615. doi: 10.1038/nature13581. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Chenoweth JG, Tesar PJ. Isolation and maintenance of mouse epiblast stem cells. Methods Mol Biol. 2010;636:25–44. doi: 10.1007/978-1-60761-691-7_2. [DOI] [PubMed] [Google Scholar]
- 33.Smith ZD, et al. A unique regulatory phase of DNA methylation in the early mammalian embryo. Nature. 2012;484:339–344. doi: 10.1038/nature10960. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Buenrostro JD, Giresi PG, Zaba LC, Chang HY, Greenleaf WJ. Transposition of native chromatin for fast and sensitive epigenomic profiling of open chromatin, DNA-binding proteins and nucleosome position. Nat Methods. 2013;10:1213–1218. doi: 10.1038/nmeth.2688. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Lara-Astiaso D, et al. Immunogenetics. Chromatin state dynamics during blood formation. Science. 2014;345:943–949. doi: 10.1126/science.1256271. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Yoshida N, Perry AC. Piezo-actuated mouse intracytoplasmic sperm injection (ICSI) Nat Protoc. 2007;2:296–304. doi: 10.1038/nprot.2007.7. [DOI] [PubMed] [Google Scholar]
- 37.Ying QL, et al. The ground state of embryonic stem cell self-renewal. Nature. 2008;453:519–523. doi: 10.1038/nature06968. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Ying QL, Nichols J, Chambers I, Smith A. BMP induction of Id proteins suppresses differentiation and sustains embryonic stem cell self-renewal in collaboration with STAT3. Cell. 2003;115:281–292. doi: 10.1016/s0092-8674(03)00847-x. [DOI] [PubMed] [Google Scholar]
- 39.Wang H, et al. One-step generation of mice carrying mutations in multiple genes by CRISPR/Cas-mediated genome engineering. Cell. 2013;153:910–918. doi: 10.1016/j.cell.2013.04.025. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Labun K, Montague TG, Gagnon JA, Thyme SB, Valen E. CHOPCHOP v2: a web tool for the next generation of CRISPR genome engineering. Nucleic Acids Res. 2016;44:W272–276. doi: 10.1093/nar/gkw398. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Macaulay IC, et al. G&T-seq: parallel sequencing of single-cell genomes and transcriptomes. Nat Methods. 2015;12:519–522. doi: 10.1038/nmeth.3370. [DOI] [PubMed] [Google Scholar]
- 42.Picelli S, et al. Full-length RNA-seq from single cells using Smart-seq2. Nat Protoc. 2014;9:171–181. doi: 10.1038/nprot.2014.006. [DOI] [PubMed] [Google Scholar]
- 43.Gu H, et al. Preparation of reduced representation bisulfite sequencing libraries for genome-scale DNA methylation profiling. Nat Protoc. 2011;6:468–481. doi: 10.1038/nprot.2010.190. [DOI] [PubMed] [Google Scholar]
- 44.Wu H, et al. Detection of differentially methylated regions from whole-genome bisulfite sequencing data without replicates. Nucleic Acids Res. 2015;43:e141. doi: 10.1093/nar/gkv715. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Raychaudhuri S, et al. Identifying relationships among genomic disease regions: predicting genes at pathogenic SNP associations and rare deletions. PLoS Genet. 2009;5:e1000534. doi: 10.1371/journal.pgen.1000534. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Schep AN, et al. Structured nucleosome fingerprints enable high-resolution mapping of chromatin architecture within regulatory regions. Genome Res. 2015;25:1757–1770. doi: 10.1101/gr.192294.115. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Ciruna BG, Rossant J. Expression of the T-box gene Eomesodermin during early mouse development. Mech Dev. 1999;81:199–203. doi: 10.1016/s0925-4773(98)00243-3. [DOI] [PubMed] [Google Scholar]
- 48.Ralston A, Rossant J. Cdx2 acts downstream of cell polarization to cell-autonomously promote trophectoderm fate in the early mouse embryo. Dev Biol. 2008;313:614–629. doi: 10.1016/j.ydbio.2007.10.054. [DOI] [PubMed] [Google Scholar]
- 49.Savory JG, et al. Cdx2 regulation of posterior development through non-Hox targets. Development. 2009;136:4099–4110. doi: 10.1242/dev.041582. [DOI] [PubMed] [Google Scholar]
- 50.Donnison M, et al. Loss of the extraembryonic ectoderm in Elf5 mutants leads to defects in embryonic patterning. Development. 2005;132:2299–2308. doi: 10.1242/dev.01819. [DOI] [PubMed] [Google Scholar]
- 51.Goldin SN, Papaioannou VE. Paracrine action of FGF4 during periimplantation development maintains trophectoderm and primitive endoderm. Genesis. 2003;36:40–47. doi: 10.1002/gene.10192. [DOI] [PubMed] [Google Scholar]
- 52.Kang M, Piliszek A, Artus J, Hadjantonakis AK. FGF4 is required for lineage restriction and salt-and-pepper distribution of primitive endoderm factors but not their initial expression in the mouse. Development. 2013;140:267–279. doi: 10.1242/dev.084996. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Nichols J, Silva J, Roode M, Smith A. Suppression of Erk signalling promotes ground state pluripotency in the mouse embryo. Development. 2009;136:3215–3222. doi: 10.1242/dev.038893. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Auclair G, Guibert S, Bender A, Weber M. Ontogeny of CpG island methylation and specificity of DNMT3 methyltransferases during embryonic development in the mouse. Genome Biol. 2014;15:545. doi: 10.1186/s13059-014-0545-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Smallwood SA, et al. Dynamic CpG island methylation landscape in oocytes and preimplantation embryos. Nat Genet. 2011;43:811–814. doi: 10.1038/ng.864. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Ooi SK, et al. DNMT3L connects unmethylated lysine 4 of histone H3 to de novo methylation of DNA. Nature. 2007;448:714–717. doi: 10.1038/nature05987. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.He J, et al. Kdm2b maintains murine embryonic stem cell status by recruiting PRC1 complex to CpG islands of developmental genes. Nat Cell Biol. 2013;15:373–384. doi: 10.1038/ncb2702. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Wu X, Johansen JV, Helin K. Fbxl10/Kdm2b recruits polycomb repressive complex 1 to CpG islands and regulates H2A ubiquitylation. Mol Cell. 2013;49:1134–1146. doi: 10.1016/j.molcel.2013.01.016. [DOI] [PubMed] [Google Scholar]
- 59.Blackledge NP, et al. Variant PRC1 complex-dependent H2A ubiquitylation drives PRC2 recruitment and polycomb domain formation. Cell. 2014;157:1445–1459. doi: 10.1016/j.cell.2014.05.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Boulard M, Edwards JR, Bestor TH. FBXL10 protects Polycomb-bound genes from hypermethylation. Nat Genet. 2015;47:479–485. doi: 10.1038/ng.3272. [DOI] [PubMed] [Google Scholar]
- 61.Irizarry RA, et al. The human colon cancer methylome shows similar hypo- and hypermethylation at conserved tissue-specific CpG island shores. Nat Genet. 2009;41:178–186. doi: 10.1038/ng.298. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Steine EJ, et al. Genes methylated by DNA methyltransferase 3b are similar in mouse intestine and human colon cancer. J Clin Invest. 2011;121:1748–1752. doi: 10.1172/JCI43169. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Schulze I, et al. Increased DNA methylation of Dnmt3b targets impairs leukemogenesis. Blood. 2016;127:1575–1586. doi: 10.1182/blood-2015-07-655928. [DOI] [PubMed] [Google Scholar]
- 64.Yang L, et al. DNMT3A Loss Drives Enhancer Hypomethylation in FLT3-ITD-Associated Leukemias. Cancer Cell. 2016;29:922–934. doi: 10.1016/j.ccell.2016.05.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Mayle A, et al. Dnmt3a loss predisposes murine hematopoietic stem cells to malignant transformation. Blood. 2015;125:629–638. doi: 10.1182/blood-2014-08-594648. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Haney SL, et al. Promoter Hypomethylation and Expression Is Conserved in Mouse Chronic Lymphocytic Leukemia Induced by Decreased or Inactivated Dnmt3a. Cell Rep. 2016;15:1190–1201. doi: 10.1016/j.celrep.2016.04.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Ben-Porath I, et al. An embryonic stem cell-like gene expression signature in poorly differentiated aggressive human tumors. Nat Genet. 2008;40:499–507. doi: 10.1038/ng.127. [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Supplementary Materials
Supplementary Table 1. De novo methylation of CGIs during extraembryonic development, Methylation status of CGIs in Epiblast and Extraembryonic Ectoderm (E×E), including designation of differential methylation status in E×E as described in the Methods (hyper, hypermethylated; hypo, hypomethylated; NC, no change; ND, insufficient measurements). Assignment to nearest gene and distance to the TSS are included.
Supplementary Table 2. Promoter methylation and associated transcriptional dynamics during implantation are influenced by CGI methylation status, Methylation values for gene promoters (classified as the region +/− 1 kb of an annotated TSS), Log2 normalized TPM (Transcripts per Million) across late preimplantation and early postimplantation samples. Promoter methylation is reported if at least 5 CpGs are covered ≥5×. The ‘Symbol’ column identifies all annotated genes for a given promoter and the reported expression value is either the TPM of the associated gene or the mean TPM if multiple genes begin at the same TSS. ‘CpGs’ indicates the number of CpGs that exist within the promoter boundary.
Supplementary Table 3. CGI methylation status for ICM outgrowths under defined conditions, CGI methylation status as measured by RRBS for ICM explanted under conditions of modulated FGF and WNT signaling. CGIs are assigned to their nearest TSS and those existing within +/– 2 kb were given the additional assignment of TSS-associated. DMR status indicates differential methylation between Epiblast and E×E from WGBS data, and PRC2 regulatory status is taken from Ref 67. We observe three discrete scenarios where CGIs are preferentially methylated: within the E×E, in the external portion of FGF+CHIR stimulated ICM outgrowths, and in FGF stimulated outgrowths. A CGI whose methylation status deviates by ≥0.1 from epiblast is scored as ‘dynamic’ and used to generate the heatmap in Fig. 2f.
Supplementary Table 4. Promoter methylation status and transcriptional dynamics for ICM outgrowths under defined conditions, Promoter methylation and associated gene expression data of ICM outgrowth conditions as measured by dual RRBS and RNA-seq. Promoter methylation is reported if at least 5 CpGs are covered ≥5×. The ‘Symbol’ column identifies all annotated genes for a given promoter and the reported expression value is either the TPM of the associated gene or the mean TPM if multiple genes begin at the same TSS.
Supplementary Table 5. CGIs methylation status for epigenetic regulator deficient E6.5 embryos, CGI methylation status as measured by RRBS for samples isolated from CRISPR/Cas9 injected embryos. CGIs are assigned to their nearest TSS and those existing within +/– 2 kb were given the additional assignment of TSS-associated. DMR status indicates differential methylation between Epiblast and E×E from WGBS data, and PRC2 regulatory status is taken from Ref 67. A CGI whose methylation status deviates by ≥0.1 from its wild type tissue is scored as ‘dynamic’ and is highlighted in Fig. 3d.
Supplementary Table 6. Promoter methylation status and transcriptional dynamics for epigenetic regulator deficient E6.5 embryos, Promoter methylation and associated gene expression data of CRISPR/Cas9 targeted embryos as measured by dual RRBS and RNA-seq. Promoter methylation is reported if at least 5 CpGs are covered ≥5×. The ‘Symbol’ column identifies all annotated genes for a given promoter and the reported expression value is either the TPM of the associated gene or the mean TPM if multiple genes begin at the same TSS. In general, E×E Hyper CGIs are preferentially induced in both the Epiblast and E×E fraction of Eed targeted E6.5 embryos and de novo methylation of these regions in E×E is specifically blocked.
Supplementary Table 7. Methylation status of E×E hypermethylated CGIs within human tissues, cancers, and cell lines, Mean and median methylation status of the 489 orthologously mapped CGIs that are called as E×E-hypermethylated in mouse across 107 ENCODE and Roadmap Initiative samples. Note, the lymphoblastoid cell line GM12878 is not characterized as cancer cell line within Encode but was generated using the Epstein-Barr Virus and scored as such in this study. Information includes designation as cancer versus normal as well as other assignments included in Extended Data Figure 9.