scholarly journals Comparative analysis of genome-wide DNA methylation identifies patterns that associate with conserved transcriptional programs in osteosarcoma

2020 ◽  
Author(s):  
Lauren J. Mills ◽  
Milcah C. Scott ◽  
Pankti Shah ◽  
Anne R. Cunanan ◽  
Archana Deshpande ◽  
...  

AbstractOsteosarcoma is an aggressive tumor of the bone that primarily affects young adults and adolescents. Osteosarcoma is characterized by genomic chaos and heterogeneity. While inactivation of tumor suppressor p53 TP53 is nearly universal other high frequency mutations or structural variations have not been identified. Despite this genomic heterogeneity, key conserved transcriptional programs associated with survival have been identified across human, canine and induced murine osteosarcoma. The epigenomic landscape, including DNA methylation, plays a key role in establishing transcriptional programs in all cell types. The role of epigenetic dysregulation has been studied in a variety of cancers but has yet to be explored at scale in osteosarcoma. Here we examined genome-wide DNA methylation patterns in 24 human and 44 canine osteosarcoma samples identifying groups of highly correlated DNA methylation marks in human and canine osteosarcoma samples. We also link specific DNA methylation patterns to key transcriptional programs in both human and canine osteosarcoma. Building on previous work, we built a DNA methylation-based measure for the presence and abundance of various immune cell types in osteosarcoma. Finally, we determined that the underlying state of the tumor, and not changes in cell composition, were the main driver of differences in DNA methylation across the human and canine samples.SignificanceThis is the first large scale study of DNA methylation in osteosarcoma and lays the ground work for the exploration of DNA methylation programs that help establish conserved transcriptional programs in the context of different genomic landscapes.

2021 ◽  
Vol 4 (1) ◽  
Author(s):  
Todd R. Robeck ◽  
Zhe Fei ◽  
Ake T. Lu ◽  
Amin Haghani ◽  
Eve Jourdain ◽  
...  

AbstractThe development of a precise blood or skin tissue DNA Epigenetic Aging Clock for Odontocete (OEAC) would solve current age estimation inaccuracies for wild odontocetes. Therefore, we determined genome-wide DNA methylation profiles using a custom array (HorvathMammalMethyl40) across skin and blood samples (n = 446) from known age animals representing nine odontocete species within 4 phylogenetic families to identify age associated CG dinucleotides (CpGs). The top CpGs were used to create a cross-validated OEAC clock which was highly correlated for individuals (r = 0.94) and for unique species (median r = 0.93). Finally, we applied the OEAC for estimating the age and sex of 22 wild Norwegian killer whales. DNA methylation patterns of age associated CpGs are highly conserved across odontocetes. These similarities allowed us to develop an odontocete epigenetic aging clock (OEAC) which can be used for species conservation efforts by provide a mechanism for estimating the age of free ranging odontocetes from either blood or skin samples.


EBioMedicine ◽  
2019 ◽  
Vol 43 ◽  
pp. 411-423 ◽  
Author(s):  
Ewoud Ewing ◽  
Lara Kular ◽  
Sunjay J. Fernandes ◽  
Nestoras Karathanasis ◽  
Vincenzo Lagani ◽  
...  

2021 ◽  
Vol 3 (Supplement_2) ◽  
pp. ii14-ii14
Author(s):  
Grayson Herrgott ◽  
Ruicong She ◽  
Thais Sabedot ◽  
Michael Wells ◽  
Karam Asmaro ◽  
...  

Abstract Background Tumor-infiltrating immune cell compositions have been previously correlated to encouragement or inhibition of tumor growth. This association highlights immune-landscape profiling through non-invasive methods as a crucial step in approaches to treatment of patients with meningioma (MNG), a prevalent primary intracranial tumor. Genome-wide DNA methylation patterns can aid in definition and assessment of cell compositions in liquid biopsy serum specimens, and allow for development of machine-learning models with predictive capabilities. Methods We profiled the cfDNA methylome (EPIC array) in liquid biopsy specimens from patients with MNG (n = 63) and nontumor controls (n = 6). We conducted both unsupervised epigenome-wide and supervised analyses of the meningioma methylome. Estimation of immune cell composition was conducted using Python-based methodology, where a reference methylome atlas of chosen cell types (B-cells, CD4- and CD8T-cells, neutrophils, natural killer cells, monocytes, cortical neuron, vascular endothelial cells, and healthy meninge) was used to deconvolute the MNG samples. Recurrence risk was estimated using an existing methylation-based Random-Forest classifier previously reported and validated, adapted to our serum-based cohort through employment of translatable meningioma subgroup-specific methylation markers (differentially methylated probes). Results We identified four distinct genome-wide methylation subgroups (k-clusters) of MNG which presented differential tumor micro-environments across all cell types investigated. Application of the DNA methylation-based Random-Forest classifier allowed for categorization of primary MNG serum samples into estimated recurrence-risk subgroups. Significantly contrasting micro-environments for the subgroups were observed across several cell-types, with those MNG more likely to recur displaying depletion in cell types reported to improve anti-tumoral response in many tumors (e.g. T-Cells). Conclusions DNA methylation based deconvolution allowed for detection of contrasting tumor microenvironment compositions across MNG methylation subtypes and recurrence-risk estimation subgroups. These results suggest that microenvironment profiling can be informative of probable tumor behavior and prognostic outcomes, helping guide therapeutic approaches towards treatment of patients with MNG.


2019 ◽  
Author(s):  
Paul J. Hop ◽  
René Luijk ◽  
Lucia Daxinger ◽  
Maarten van Iterson ◽  
Koen F. Dekkers ◽  
...  

SUMMARYDNA methylation is a key epigenetic modification in human development and disease, yet there is limited understanding of its highly coordinated regulation. Here, we identified 818 genes that influence DNA methylation patterns in blood using large-scale population genomics data. By employing genetic instruments as causal anchors, we identified directed associations between gene expression and distant DNA methylation levels, whilst ensuring specificity of the associations by correcting for linkage disequilibrium and pleiotropy among neighboring genes. We found that DNA methylation patterns are commonly shaped by transcription factors that consistently increase or decrease DNA methylation levels. However, we also observed genes encoding proteins without DNA binding activity with widespread effects on DNA methylation (e.g. NFKBIE, CDCA7(L) and NLRC5) and we suggest plausible mechanisms underlying these findings. Many of the reported genes were unknown to influence DNA methylation, resulting in a comprehensive resource providing insights in the principles underlying epigenetic regulation.


2012 ◽  
Vol 22 (8) ◽  
pp. 1419-1425 ◽  
Author(s):  
W. Qu ◽  
S.-i. Hashimoto ◽  
A. Shimada ◽  
Y. Nakatani ◽  
K. Ichikawa ◽  
...  

Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 391-391
Author(s):  
Amber Hogart ◽  
Subramanian S. Ajay ◽  
Hatice Ozel Abaan ◽  
Stacie M. Anderson ◽  
Elliott H. Margulies ◽  
...  

Abstract Abstract 391 DNA methylation is a reversible epigenetic modification that is required for proper mammalian development and is proposed to contribute to the pathogenesis of hematologic diseases including leukemia and bone marrow failure syndromes. Elucidating the pathways and genes regulated by DNA methylation during hematopoiesis may reveal new therapeutic targets for disease. Because the phenotype and activity of hematopoietic stem cells (HSC) and hematopoietic progenitor cells of many different lineages have been defined by both in vitro and in vivo assays, hematopoiesis is an excellent model for investigating epigenomic changes during differentiation. HSCs have the ability to self-renew and to generate blood cells of all lineages, which allows them to repopulate recipients after stem cell transplantation. The common myeloid progenitor (CMP) gives rise to all myeloid cell types including neutrophils, monocytes, platelets, and red blood cells, but cannot self renew or repopulate. In contrast to the multipotent HSC and CMP, erythroblasts (ERY) are terminally committed cells that become mature enucleated red blood cells. These three cell types represent unique stages of lineage commitment with distinct transcriptional programs, and potentially unique epigenomic signatures. In contrast to human HSC, which are defined by the absence of several cell surface markers, mouse HSC have the cell surface phenotype of lineage marker negative (Lin-) c-kit+ Sca-1+ and can be positively selected. For this reason we chose the mouse model for genome-wide methylation profiling. Murine HSC and CMP (Lin- c-kit+ Sca-1-) cells were enriched from adult mouse bone marrow with flow cytometry. Erythroblasts (CD71+/Ter119+) were positively selected from E13.5 mouse fetal livers. Genomic DNA isolated from each enriched cell population was sheared to 200-300 bp fragments. MBD2, one of five endogenous mammalian methyl CpG binding domain proteins, binds methylated DNA sequences with broad affinity. Methylated DNA fragments were enriched from the genomic DNA using a tagged, recombinant MBD2 pulldown kit (Active Motif). After pulldown, enrichment of known methylated sequences regulating the imprints of Snrpn and Rasgrf was validated by qPCR. Two biological replicates of HSC, CMP, and ERY methylated sequences and negative control supernatant fractions were submitted for high-throughput sequencing with the Illumina Genome Analyzer platform. Raw sequence data containing 32-46 × 106 reads of 36-50 base pairs were obtained for each sample. The Eland program was used to map 41-59% of reads to unique sequences in the mouse genome. Model-based Analysis of ChIP-Seq (MACS) was used to estimate the mean and variance of the sequence tag distribution across the genome and define peaks below the significance threshold of p<10-5. The number of methylation peaks decreased as cells differentiated, with 64,000 peaks identified in HSC (24,000 unique), 41,000 peaks in CMP (2000 unique), and 23,000 peaks in ERY (1000 unique). Approximately 20,000 peaks were common between all cell types with 57% of these peaks residing in RefSeq genes, 8% in regions adjacent to RefSeq genes (<10 kb), and 35% of methylation peaks in intergenic regions. Comparison of HSC expression data from Akashi et al (Blood 101: 383, 2003) to our HSC genic methylation peaks revealed that 2/3 of HSC genic peaks are within transcriptionally silent genes while 1/3 of HSC genic peaks are within expressed genes. Although DNA methylation is often associated with gene silencing, the important developmental gene Gata2 contains methylation peaks in HSC and CMP, cells that express Gata2, that are absent in ERY, where Gata2 is repressed. A Gata1-Fog1-Mbd2 complex has been described by Rodriguez et al (EMBO 24: 2354, 2005), therefore providing a link between DNA methylation and proteins known to bind at the Gata2 locus. Grass et al (Mol. Cell. Biol. 26:7056, 2006) determined that Gata2 is regulated by long-range interactions of GATA protein complexes, and consistent with this observation, distinct methylation patterns are observed up to 100 kb upstream of the Gata2 gene. Our genome-wide analysis supports an association of methylation with gene silencing but also suggests that DNA methylation is a dynamic epigenetic mark that influences hematopoietic differentiation. The changes in DNA methylation we observe around Gata2 may also contribute to long-range chromatin organization. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2011 ◽  
Vol 117 (2) ◽  
pp. 553-562 ◽  
Author(s):  
Brian A. Walker ◽  
Christopher P. Wardell ◽  
Laura Chiecchio ◽  
Emma M. Smith ◽  
Kevin D. Boyd ◽  
...  

Abstract We used genome-wide methylation microarrays to analyze differences in CpG methylation patterns in cells relevant to the pathogenesis of myeloma plasma cells (B cells, normal plasma cells, monoclonal gammopathy of undetermined significance [MGUS], presentation myeloma, and plasma cell leukemia). We show that methylation patterns in these cell types are capable of distinguishing nonmalignant from malignant cells and the main reason for this difference is hypomethylation of the genome at the transition from MGUS to presentation myeloma. In addition, gene-specific hypermethylation was evident at the myeloma stage. Differential methylation was also evident at the transition from myeloma to plasma cell leukemia with remethylation of the genome, particularly of genes involved in cell–cell signaling and cell adhesion, which may contribute to independence from the bone marrow microenvironment. There was a high degree of methylation variability within presentation myeloma samples, which was associated with cytogenetic differences between samples. More specifically, we found methylation subgroups were defined by translocations and hyperdiploidy, with t(4;14) myeloma having the greatest impact on DNA methylation. Two groups of hyperdiploid samples were identified, on the basis of unsupervised clustering, which had an impact on overall survival. Overall, DNA methylation changes significantly during disease progression and between cytogenetic subgroups.


2018 ◽  
Author(s):  
AJ Price ◽  
L Collado-Torres ◽  
NA Ivanov ◽  
W Xia ◽  
EE Burke ◽  
...  

AbstractWe have characterized the landscape of DNA methylation (DNAm) across the first two decades of human neocortical development in NeuN+ neurons using whole-genome bisulfite sequencing and compared them to non-neurons (primarily glia) and prenatal homogenate cortex. We show that DNAm changes more dramatically during the first five years of postnatal life than during the entire remaining period. We further refined global patterns of increasingly divergent neuronal CpG and CpH methylation (mCpG and mCpH) into six developmental trajectories and found that in contrast to genome-wide patterns, neighboring mCpG and mCpH levels within these regions were highly correlated. We then integrated paired RNA-seq data and identified direct regulation of hundreds of transcripts and their splicing events exclusively by mCpH levels, independently from mCpG levels, across this period. We finally explored the relationship between DNAm patterns and development of brain-related phenotypes and found enriched heritability for many phenotypes within DNAm features we identify.


2020 ◽  
Author(s):  
Izaskun Mallona ◽  
Ioana Mariuca Ilie ◽  
Massimiliano Manzo ◽  
Amedeo Caflisch ◽  
Tuncay Baubec

AbstractMammalian de novo DNA methyltransferases (DNMT) are responsible for the establishment of cell-type-specific DNA methylation in healthy and diseased tissues. Through genome-wide analysis of de novo methylation activity in murine stem cells we uncover that DNMT3A prefers to methylate CpGs followed by cytosines or thymines, while DNMT3B predominantly methylates CpGs followed by guanines or adenines. These signatures are further observed at non-CpG sites, resembling methylation context observed in specialised cell types, including neurons and oocytes. We further show that these preferences are not mediated by the differential recruitment of the two de novo DNMTs to the genome but are resulting from structural differences in their catalytic domains. Molecular dynamics simulations suggest that, in case of DNMT3A, the preference is due to favourable polar interactions between the flexible Arg836 side chain and the guanine that base-pairs with the cytosine following the CpG. This context-dependent de novo DNA methylation provides additional insights into the complex regulation of methylation patterns in different cell types.


2020 ◽  
Vol 10 (1) ◽  
Author(s):  
Wardah Mahmood ◽  
Lars Erichsen ◽  
Pauline Ott ◽  
Wolfgang A. Schulz ◽  
Johannes C. Fischer ◽  
...  

AbstractLINE-1 hypomethylation of cell-free DNA has been described as an epigenetic biomarker of human aging. However, in the past, insufficient differentiation between cellular and cell-free DNA may have confounded analyses of genome-wide methylation levels in aging cells. Here we present a new methodological strategy to properly and unambiguously extract DNA methylation patterns of repetitive, as well as single genetic loci from pure cell-free DNA from peripheral blood. Since this nucleic acid fraction originates mainly in apoptotic, senescent and cancerous cells, this approach allows efficient analysis of aged and cancerous cell-specific DNA methylation patterns for diagnostic and prognostic purposes. Using this methodology, we observe a significant age-associated erosion of LINE-1 methylation in cfDNA suggesting that the threshold of hypomethylation sufficient for relevant LINE-1 activation and consequential harmful retrotransposition might be reached at higher age. We speculate that this process might contribute to making aging the main risk factor for many cancers.


Sign in / Sign up

Export Citation Format

Share Document