scholarly journals TBIO-08. BASE-RESOLUTION METHYLOMES OF GLIOMAS BEARING HISTONE H3.3 MUTATIONS REVEAL A G34 MUTANT-SPECIFIC SIGNATURE SHARED WITH BONE TUMORS

2020 ◽  
Vol 22 (Supplement_3) ◽  
pp. iii468-iii468
Author(s):  
Yuhei Sangatsuda ◽  
Fumihito Miura ◽  
Hiromitsu Araki ◽  
Masahiro Mizuguchi ◽  
Nobuhiro Hata ◽  
...  

Abstract BACKGROUND Two recurrent mutations, K27M and G34R/V, in H3F3A, encoding non-canonical histone H3.3, are reported in pediatric and young adult gliomas, whereas G34W mutation was prevalent in bone tumors. In contrast to K27 mutation, it remains elusive how G34 mutations affect the epigenome. Here we performed whole-genome bisulfite sequencing of four G34R-mutated gliomas and the G34V-mutated glioma cell line KNS-42. Similarly, we analyzed seven and three gliomas harboring K27M and no mutations in H3F3A, respectively. These data were compared with those on bone tumors. RESULTS G34R-mutated gliomas exhibited lower global methylation levels, similar CpG island (CGI) methylation levels, and compromised hypermethylation of telomere-proximal CGIs compared with those bearing K27M and no mutations. Hypermethylated regions specific to G34R-mutated gliomas were enriched for CGIs, including those of OLIG1, OLIG2, and canonical histone genes in the HIST1 cluster. These CGIs were hypermethylated in osteosarcomas with, but not without, the G34W mutation. In KNS-42 cells, CGIs with G34V-mutated histone H3.3 exhibited higher methylation levels than those with wild-type histone H3.3. This effect was also observed in the G34R-mutated glioma samples. CONCLUSIONS Gliomas bearing G34R/V mutations display characteristic methylomic alterations, some of which are shared by osteosarcomas with the G34W mutation. Deposition of G34 variants may lead to elevated methylation of otherwise hypomethylated, histone H3.3-bearing CGIs.

2020 ◽  
Vol 10 (1) ◽  
Author(s):  
Yuhei Sangatsuda ◽  
Fumihito Miura ◽  
Hiromitsu Araki ◽  
Masahiro Mizoguchi ◽  
Nobuhiro Hata ◽  
...  

Abstract Two recurrent mutations, K27M and G34R/V, in H3F3A, encoding non-canonical histone H3.3, are reported in pediatric and young adult gliomas, whereas G34W mutation is prevalent in bone tumors. In contrast to K27M mutation, it remains elusive how G34 mutations affect the epigenome. Here we performed whole-genome bisulfite sequencing of four G34R-mutated gliomas and the G34V-mutated glioma cell line KNS-42 for comparison with gliomas harboring K27M and no mutations in H3F3A and with G34W-mutated bone tumors. G34R-mutated gliomas exhibited lower global methylation levels, similar CpG island (CGI) methylation levels, and compromised hypermethylation of telomere-proximal CGIs, compared to the other two glioma subgroups. Hypermethylated regions specific to G34R-mutated gliomas were enriched for CGIs, including those of OLIG1, OLIG2, and canonical histone genes in the HIST1 cluster. They were notably hypermethylated in osteosarcomas with, but not without, G34W mutation. Independent component analysis revealed that G34 mutation-specific components shared a significant similarity between glioma and osteosarcoma, suggesting that G34 mutations exert characteristic methylomic effects regardless of the tumor tissue-of-origin. CRISPR/Cas9-mediated disruption of G34V-allele in KNS-42 cells led to demethylation of a subset of CGIs hypermethylated in G34R-mutated gliomas. These findings will provide a basis for elucidating epigenomic roles of G34 oncohistone in tumorigenesis.


Leukemia ◽  
2021 ◽  
Author(s):  
Elisabeth R. Wilson ◽  
Nichole M. Helton ◽  
Sharon E. Heath ◽  
Robert S. Fulton ◽  
Jacqueline E. Payton ◽  
...  

AbstractRecurrent mutations in IDH1 or IDH2 in acute myeloid leukemia (AML) are associated with increased DNA methylation, but the genome-wide patterns of this hypermethylation phenotype have not been comprehensively studied in AML samples. We analyzed whole-genome bisulfite sequencing data from 15 primary AML samples with IDH1 or IDH2 mutations, which identified ~4000 focal regions that were uniquely hypermethylated in IDHmut samples vs. normal CD34+ cells and other AMLs. These regions had modest hypermethylation in AMLs with biallelic TET2 mutations, and levels of 5-hydroxymethylation that were diminished in IDH and TET-mutant samples, indicating that this hypermethylation results from inhibition of TET-mediated demethylation. Focal hypermethylation in IDHmut AMLs occurred at regions with low methylation in CD34+ cells, implying that DNA methylation and demethylation are active at these loci. AML samples containing IDH and DNMT3AR882 mutations were significantly less hypermethylated, suggesting that IDHmut-associated hypermethylation is mediated by DNMT3A. IDHmut-specific hypermethylation was highly enriched for enhancers that form direct interactions with genes involved in normal hematopoiesis and AML, including MYC and ETV6. These results suggest that focal hypermethylation in IDH-mutant AML occurs by altering the balance between DNA methylation and demethylation, and that disruption of these pathways at enhancers may contribute to AML pathogenesis.


2017 ◽  
Author(s):  
Nelly Olova ◽  
Felix Krueger ◽  
Simon Andrews ◽  
David Oxley ◽  
Rebecca V. Berrens ◽  
...  

AbstractBackgroundWhole-genome bisulfite sequencing (WGBS) is becoming an increasingly accessible technique, used widely for both fundamental and disease-oriented research. Library preparation methods benefit from a variety of available kits, polymerases and bisulfite conversion protocols. Although some steps in the procedure, such as PCR amplification, are known to introduce biases, a systematic evaluation of biases in WGBS strategies is missing.ResultsWe perform a comparative analysis of several commonly used pre-and post-bisulfite WGBS library preparation protocols for their performance and quality of sequencing outputs. Our results show that bisulfite conversion per se is the main trigger of pronounced sequencing biases, and PCR amplification builds on these underlying artefacts. The majority of standard library preparation methods yield a significantly biased sequence output and overestimate global methylation. Importantly, both absolute and relative methylation levels at specific genomic regions vary substantially between methods, with clear implications for DNA methylation studies.ConclusionsWe show that amplification-free library preparation is the least biased approach for WGBS. In protocols with amplification, the choice of BS conversion protocol or polymerase can significantly minimize artefacts. To aid with the quality assessment of existing WGBS datasets, we have integrated a bias diagnostic tool in the Bismark package and offer several approaches for consideration during the preparation and analysis of WGBS datasets.


Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 433-433
Author(s):  
Shamika Ketkar-Kulkarni ◽  
Angela Maria Verdoni ◽  
Nichole Helton ◽  
Christopher B Cole ◽  
Celia Venezia ◽  
...  

Abstract We previously identified recurrent mutations in the de novo DNA methyltransferase DNMT3A in patients with acute myeloid leukemia (AML). The most common DNMT3A mutation in AML (R882H) creates a dominant negative protein that reduces DNA methylation activity by ~80% in AML cells, and causes canonical patterns of DNA hypomethylation in the AML genome (Russler-Germain et al, Cancer Cell 2014). Approaches to restore DNMT3A activity in these AML genomes may be therapeutically relevant, but only if remethylation can return these genomes to their native methylation state. To begin to address whether DNA methylation can be restored in hematopoietic cells that are deficient for Dnmt3a, we performed an Òadd-backÓ experiment using a transgenic mouse model system. First, to characterize the genome-wide effect of Dnmt3a loss on DNA methylation in hematopoietic cells, we carried out whole-genome bisulfite sequencing (WGBS) on the total bone marrow cells of wild type (WT) mice, vs. marrow derived from littermates homozygous for a germline Dnmt3a null mutation (Dnmt3a-/-, Okano et al Cell, 1999), which have overtly normal hematopoiesis. Total bone marrow cells from Dnmt3a-/- mice have a canonical pattern of DNA hypomethylation at specific CpG residues and regions in the genome; many of the CpGs are virtually unmethylated in specific regions, suggesting that the normal Dnmt3a-dependent methylation ÒmarkÓ was added in stem/progenitor cells, and then maintained in all lineages. To define the timing and specificity of remethylation in Dnmt3a-/- mice, we crossed heterozygotes from this line with transgenic mice containing a tetracycline-inducible human wild type DNMT3A cDNA (DNMT3A Tg mice), and a second transgenic mouse containing the rtTA coactivator, expressed from the Rosa26 locus. When DNMT3A Tg+, rtTA+ mice are fed Doxycycline (Dox) chow for one week, WT human DNMT3A is expressed in the marrow at a level ~4 times higher that of endogenous murine Dnmt3a. Since Dnmt3a-/- mice die of severe runting at about three weeks of age, we harvested the marrow cells from Dnmt3a-/-, DNMT3A Tg+, rtTA+mice at 2 weeks of age, and transplanted them into lethally irradiated C57Bl/6 recipients. The marrow was allowed to engraft for four weeks. Half of the mice were then given Dox chow, and half were untreated. Whole bone marrow was isolated from pairs of mice (+ vs. -Dox), DNA was purified, and then subjected to whole genome bisulfite sequencing (WGBS). WGBS produced methylation data on >93% of the CpGs in the mouse reference sequence with a median coverage of 10-12x per sample. Differential methylation analysis was performed on 2 kb tiled windows across the whole genome, revealing 108, 797 differentially methylated regions (DMRs) that were hypomethylated (Table 1). Dnmt3a-/-, DNMT3A Tg+, rtTA+bone marrow from mice without Dox (i.e. no DNMT3A was expressed) demonstrated no evidence for remethylation at any time after transplant. However, if mice were treated with Dox for only 2 weeks, 59% of these DMRs were remethylated, increasing to 70% at 4 weeks, and 83% by week 9. Data from weeks 12 and 24 are pending. Patterns of remethylation for a subset of 560 differentially methylated CpGs (DMCpGs) are shown in Figure 1. These CpGs all had methylation values of >= 90% in WT mouse bone marrow cells, and <=10% in Dnmt3a-/- derived marrow. The methylation patterns for these CpGs is shown as a function of time, after Òadd-backÓ of WT DNMT3A (induced by Dox chow, upper panel). Individual CpGs are tracked by color-coded lines, which show the % methylation at each time point (red = > 66% methylated at a time point, yellow = 33%-66%, and green = < 33%). Most CpGs are remethylated after only 2 weeks of DNMT3A expression, but some are delayed, and become remethylated later (yellow at week 2, changing to red at 4 or 9 weeks). A small subset of these CpGs remethylate very slowly, if at all (green). Without the induction of DNMT3A (-Dox, lower panel), little or no remethylation is detected. Inspection of specific regions of the genome showed that regions that are normally unmethylated in the bone marrow cells of WT mice are rarely methylated by adding back human DNMT3A in this system (data not shown), showing that remethylation is highly specific. Although the mechanisms involved in specifying remethylation patterns are not yet clear, these data may have important implications for therapeutically restoring DNMT3A activity in AML patients with DNMT3A mutations that reduce its activity. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. SCI-47-SCI-47
Author(s):  
Andrew P. Feinberg

Abstract Since the discovery of altered DNA methylation in cancer in 1982, most studies of cancer epigenetics have focused on epimutations which could serve as surrogates of mutation. In the past decade, we and our collaborators have been leading efforts to develop whole-genome approaches to epigenetic analysis of human disease, that include novel approaches to array-based analysis and whole-genome bisulfite sequencing. This has led to the first whole-genome bisulfite sequencing methylation map of the cancer genome. Surprising results have been the discovery of CpG island shores and large hypomethylated blocks corresponding to nuclear lamina/heterochromatin regions, and accounting for the vast majority of epigenetic alterations in cancer. These results point to the possibility that at least solid tumors represent epigenetically a single process with a common molecular characteristic; namely increased epigenetic plasticity that allows selection of the tumor cells at the expense of the host. This view can explain most of the hallmarks of cancer, as well as offering a strategy for powerful new approaches to risk detection and treatment. This whole-genome epigenetic approach has also revealed that the classical lineage structure of hematopoiesis is very well reflected in DNA methylation, but that there are very surprising dynamic changes in this process, with waves of both increasing and decreasing methylation changes. This work has also revealed a molecular basis for phenotypic memory in induced pluripotent stem cells, as well as a strong connection between reprogramming differentially methylated regions, or R-DMRs, and tissue-specific differentially methylated regions, or T-DMRs. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 608-608 ◽  
Author(s):  
David H. Spencer ◽  
Bilal Al-Khalil ◽  
David Russler-Germain ◽  
Tamara Lamprecht ◽  
Nicole Havey ◽  
...  

Abstract Mutations in the de novo DNA methyltransferase DNMT3A are found in ~25% of patients with acute myeloid leukemia (AML) and most commonly affect codon 882 within the catalytic domain of the protein. We have previously shown that this mutation has dominant negative activity in vitro and is associated with hypomethylation at specific CpG dinucleotides in primary AML samples using array-based methylation data. However, the genome-wide extent and patterns of DNA methylation associated with this hypomethylation are currently unknown. In addition, it is unclear if the methylation differences caused by this mutation result in RNA expression changes at specific targets across the genome, or whether they are associated with altered chromatin structure. To explore the genome-wide consequences of the DNMT3A R882H mutation on DNA methylation and chromatin structure, we carried out whole-genome bisulfite sequencing (WGBS) and transposase-mediated chromatin accessibility profiling (ATAC-seq) on 3 primary normal karyotype AML samples with the DNMT3A R882H mutation and 4 matched AML samples without a DNMT3A mutation. All 7 had the NPMc mutation but lacked mutations in other genes involved in DNA methylation, including IDH1, IDH2, and TET2. WGBS produced methylation data on >93% of the CpGs in the human reference sequence with a median coverage of 7-13x. The overall mean methylation was not statistically different in the samples with R882H mutations, although there was a small but statistically significant difference in the methylation at CpGs in CpG islands (DNMT3A R882H mean: 18.1%, DNMT3A wild-type mean: 21.4%; P=0.02). Differential methylation analysis was performed on ~5 million CpG clusters (median of 5 CpGs per cluster; median cluster size of 202 bp) and identified 95,845 differentially methylated clusters with a mean difference >25% and a q-value < 0.01, the majority of which (88,512; 93%) were hypomethylated in the DNMT3A R882H samples. Using more strict criteria (>50% mean difference) and merging differentially methylated clusters within 50 bp, we identified 2,782 differentially methylated regions (DMRs) with a mean size of 255 bp (median of 11 CpGs), of which 97% were hypomethylated. These DMRs were distributed across the genome and were statistically associated with CpG dense regions, including annotated CpG islands and shores (islands: 1,104 of 2,782; 29.9%; shores: 1,118 of 2,782; 30.3%; P<10-10), and gene promoters (816 of 2,782; 23.7%; P< 10-12). Analysis of chromatin accessibility data from 6 samples (3 DNMT3A R882H and 3 DNMT3A wild-type) showed that a subset of the DNMT3A R882H-associated hypomethylated DMRs (366 of 2,704; 13.5%) were located within 100 bp of an ATAC-seq peak unique to DNMT3A R882H AML samples. Further analysis of all DMRs showed ATAC-seq signal enrichment in the R882H samples specifically at hypomethylated loci (Figure 1). Similar enrichment was not observed in the DNMT3A wild-type AMLs at hypomethylated DMRs (N=78), suggesting that hypomethylation caused by the DNMT3A R882H mutation is specifically associated with changes in chromatin structure. Initial analysis of existing PolyA+ RNA-seq data for these AMLs did not reveal canonical expression changes in annotated genes located near the DMRs, implying that methylation and other epigenetic changes might affect distant genes or previously unannotated RNA species that were not present in our dataset. Efforts to sequence all RNA species present in these samples are therefore underway. In summary, we have conducted an initial analysis of genome-wide, CpG-resolution DNA methylation data from primary AML samples with the DNMT3A R882H mutation. This mutation is associated with a genome-wide, focal hypomethylation phenotype that occurs at small, CpG-dense loci across the genome. We also found that many hypomethylated loci are associated with changes in chromatin structure. These findings represent the first evidence that the methylation changes caused by this mutation can have functional consequences on the epigenetic state of specific loci in AML cells, and set the stage for defining the specific events that are responsible for AML pathogenesis in patients who have this mutation. Figure 1 WGBS (bottom tracks) and chromatin accessibility (ATAC-seq, top tracks) from 3 primary AML samples with the DNMT3A R882H mutation (in red) and 3 with no DNMT3A mutation (in blue) at a hypomethylated locus within the HS3ST3B1 gene. Figure 1. WGBS (bottom tracks) and chromatin accessibility (ATAC-seq, top tracks) from 3 primary AML samples with the DNMT3A R882H mutation (in red) and 3 with no DNMT3A mutation (in blue) at a hypomethylated locus within the HS3ST3B1 gene. Disclosures No relevant conflicts of interest to declare.


2021 ◽  
Vol 12 (1) ◽  
Author(s):  
Laura Santini ◽  
Florian Halbritter ◽  
Fabian Titz-Teixeira ◽  
Toru Suzuki ◽  
Maki Asami ◽  
...  

AbstractIn mammalian genomes, differentially methylated regions (DMRs) and histone marks including trimethylation of histone 3 lysine 27 (H3K27me3) at imprinted genes are asymmetrically inherited to control parentally-biased gene expression. However, neither parent-of-origin-specific transcription nor imprints have been comprehensively mapped at the blastocyst stage of preimplantation development. Here, we address this by integrating transcriptomic and epigenomic approaches in mouse preimplantation embryos. We find that seventy-one genes exhibit previously unreported parent-of-origin-specific expression in blastocysts (nBiX: novel blastocyst-imprinted expressed). Uniparental expression of nBiX genes disappears soon after implantation. Micro-whole-genome bisulfite sequencing (µWGBS) of individual uniparental blastocysts detects 859 DMRs. We further find that 16% of nBiX genes are associated with a DMR, whereas most are associated with parentally-biased H3K27me3, suggesting a role for Polycomb-mediated imprinting in blastocysts. nBiX genes are clustered: five clusters contained at least one published imprinted gene, and five clusters exclusively contained nBiX genes. These data suggest that early development undergoes a complex program of stage-specific imprinting involving different tiers of regulation.


Cancers ◽  
2021 ◽  
Vol 13 (1) ◽  
pp. 158
Author(s):  
Valentina Condelli ◽  
Giovanni Calice ◽  
Alessandra Cassano ◽  
Michele Basso ◽  
Maria Grazia Rodriquenz ◽  
...  

Epigenetics is involved in tumor progression and drug resistance in human colorectal carcinoma (CRC). This study addressed the hypothesis that the DNA methylation profiling may predict the clinical behavior of metastatic CRCs (mCRCs). The global methylation profile of two human mCRC subgroups with significantly different outcome was analyzed and compared with gene expression and methylation data from The Cancer Genome Atlas COlon ADenocarcinoma (TCGA COAD) and the NCBI GENE expression Omnibus repository (GEO) GSE48684 mCRCs datasets to identify a prognostic signature of functionally methylated genes. A novel epigenetic signature of eight hypermethylated genes was characterized that was able to identify mCRCs with poor prognosis, which had a CpG-island methylator phenotype (CIMP)-high and microsatellite instability (MSI)-like phenotype. Interestingly, methylation events were enriched in genes located on the q-arm of chromosomes 13 and 20, two chromosomal regions with gain/loss alterations associated with adenoma-to-carcinoma progression. Finally, the expression of the eight-genes signature and MSI-enriching genes was confirmed in oxaliplatin- and irinotecan-resistant CRC cell lines. These data reveal that the hypermethylation of specific genes may provide prognostic information that is able to identify a subgroup of mCRCs with poor prognosis.


2020 ◽  
Vol 22 (Supplement_3) ◽  
pp. iii348-iii348
Author(s):  
Tina Huang ◽  
Andrea Piunti ◽  
Elizabeth Bartom ◽  
Jin Qi ◽  
Rintaro Hashizume ◽  
...  

Abstract BACKGROUND Histone H3.3 mutation (H3F3A) occurs in 50% of cortical pediatric high-grade gliomas. This mutation replaces glycine 34 with arginine or valine (G34R/V), impairing SETD2 activity (H3K36-specific trimethyltransferase), resulting in reduced H3K36me on H3G34V nucleosomes relative to wild-type. This contributes to genomic instability and drives distinct gene expressions associated with tumorigenesis. However, it is not known if this differential H3K36me3 enrichment is due to H3G34V mutant protein alone. Therefore, we set to elucidate the effect of H3G34V on genomic H3K36me3 enrichment in vitro. METHODS Doxycycline-inducible short hairpin RNA (shRNA) against H3F3A was delivered via lentivirus to established H3G34V mutant pediatric glioma cell line KNS42, and H3G34V introduced into H3.3 wild type normal human astrocytes (NHA). Transfections were confirmed by western blot, fluorescent imaging, and flow cytometry, with resulting H3.3WT and H3K36me3 expression determined by western blot. H3.3WT, H3K36me3, and H3G34V ChIP-Seq was performed to evaluate genomic enrichment. RESULTS Complete knockdown of H3G34V was achieved with DOX-induced shRNA, with no change in total H3.3, suggesting disproportionate allelic frequency of genes encoding H3.3 (H3F3A and H3F3B). Modest increase in H3K36me3 occurred after H3F3A-knockdown from KNS42, suggesting H3G34V alone impacts observed H3K36me3 levels. Distinct H3K36me3 genomic enrichment was observed with H3G34V knock-in. CONCLUSIONS We demonstrate that DOX-inducible knockdown of H3F3A in an H3G34V mutant pediatric glioma cells and H3G34V mutation transduction in wild-type astrocytes affects H3K36me3 expression. Further evaluation by ChIP-Seq analysis for restoration of wild-type genomic H3K36me3 enrichment patterns with H3G34V knockdown, and mutant H3K36me3 patterns with H3G34V transduction, is currently underway.


Sign in / Sign up

Export Citation Format

Share Document