PATH-06. DNA METHYLATION PATTERNS AND IMMUNE MICROENVIRONMENT IN CYSTICGBM

2021 ◽  
Vol 23 (Supplement_6) ◽  
pp. vi115-vi116
Author(s):  
Zhihua Chen ◽  
Denis O’Meally ◽  
David Frankhouser ◽  
Mari Shahmanyan ◽  
Biao Tang ◽  
...  

Abstract As a rare subtype of glioblastomas (GBM), genetic features in cystic GBM (cGBM) are still largely unknown. We have previously identified a series of active T cell subsets and positive cytokines/chemocytokines in the cystic fluid of cGBMs, including IFN-gamma, IL-2, IL-6, IL-8, TNF-alpha, and MIP-1alpha/beta. This study aimed to evaluate the prognosis of cGBM through DNA methylation patterns and immune microenvironment transcriptional signatures. IvyGAP, EGA and fresh samples from a prospective Chinese cohort were collected to investigate genomic signatures, immune microenvironment and survival outcomes in patients with cGBMs. Gd T1-weighted, T2-weighted, T2-Flair, DWI or ADC images were used to classify cGBM. 143 cases with MGMT/IDH record were used to compare survival time and genomic analysis. Among the three cohorts, Kaplan-Meier analysis showed cystic features have benefit to overall survival (OS). Moreover, cyst feature is the forth factor (HR=0.65, [95% CI 0.42, 1.01]) of survival after IDH, MGMT and Stupp therapeutic regimen. Interestingly, in IDH-wild/MGMT-unmethyled subtype, cGBM patients have longer OS vs noncystic GBM (noncGBM) patients (HR = 0.57, [95% CI 0.33, 0.99]). Compared to noncGBM, cGBM patients have hypomethylation state both in whole gene region and cpG islands. Ivygap database showed between cystic and noncystic group, in sub-structures such as CT (cellular tumor region), CTpan (pseudopalisading cells around necrosis) and CTpnz (perinecrotic zone) there are differential gene expressions and different enrichment pathways. GSEA analysis showed within cGBM group, many gene sets associated with immune function activation such as TRL1/2/3/7 and IFN pathways. These results suggest that cysts of GBM may be associated with hypomethylation status an activated immune microenvironment which is associated with longer survival and may define a unique subgroup of GBM with intrinsically different biology and prognosis.

Author(s):  
Chao Wang ◽  
Min Shi ◽  
Lei Zhang ◽  
Jun Ji ◽  
Ruyan Xie ◽  
...  

Abstract Objective To investigate the molecular characteristics in tumor immune microenvironment that affect long-term survival of patients with pancreatic adenocarcinoma (PAAD). Methods The tumor related genetic features of a female PAAD patient (over 13-year survival) who suffered from multiple recurrences and metastases, and six operations over one decade were investigated deeply. Genomic features and immune microenvironment signatures of her primary lesion as well as six metastatic tumors at different time-points were characterized. Results High-frequency clonal neoantigenic mutations identified in these specimens revealed the significant associations between clonal neoantigens with her prognosis after each surgery. Meanwhile, the TCGA and ICGC databases were employed to analyse the function of KRAS G12V in pancreatic cancer. Conclusions The genomic analysis of clonal neoantigens combined with tumor immune microenvironment could promote the understandings of personalized prognostic evaluation and the stratification of resected PAAD individuals with better outcome.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 2498-2498
Author(s):  
Claudia Gebhard ◽  
Mohammed Sadeh ◽  
Dagmar Glatz ◽  
Lucia Schwarzfischer ◽  
Rainer Spang ◽  
...  

Abstract Abstract 2498 CpG islands show frequent and often disease-specific epigenetic alterations during malignant transformation, however, the underlying mechanisms are poorly understood. We used methyl-CpG immunoprecipitation (MCIp) to generate comparative DNA methylation profiles of 30 patients with acute myeloid leukemia for human CpG islands across the genome. DNA methylation profiles across 23.000 CpG islands revealed highly heterogeneous methylation patterns in AML with over 6000 CpG islands showing aberrant de novo methylation in AML. Based on these profiles we selected a subset of 380 CpG islands (covering 15.000 individual CpGs) for detailed fine-mapping analyses of aberrant DNA methylation in 185 patients with AML (50% normal karyotype). We found that a proportion of patients (5/185) displayed a concerted hypermethylation at almost all studied loci, representing the rare CpG island methylator phenotype (CIMP) in AML. Meta analysis of methylation profiling and published ChIP sequencing data separated CpG islands in two groups. A highly correlated subgroup of CpG island regions was strongly associated with histone H3 lysine 27 trimethylation in human hematopoietic progenitor cells, suggesting that disease-related de novo DNA methylation at these CpG islands is linked with polycomb group protein (PcG)-mediated repression. The group of mainly non-PcG target CpG islands showed heterogeneous methylation patterns across patients and unsupervised hierarchical clustering revealed a correlation of methylation profiles with genetic disease markers, including oncofusion proteins as well as CEBPA- and NPM1-mutations. Our study suggests that both epigenetic as well as genetic aberrations may underlay AML-related changes in CpG island DNA methylation states. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 211-211
Author(s):  
Amber Hogart ◽  
Jens Lichtenberg ◽  
Subramanian Ajay ◽  
Elliott Margulies ◽  
David M. Bodine

Abstract Abstract 211 The hematopoietic system is ideal for the study of epigenetic changes in primary cells because hematopoietic cells representing distinct stages of hematopoiesis can be enriched and isolated by differences in surface marker expression. DNA methylation is an essential epigenetic mark that is required for normal development. Conditional knockout of the DNA methyltransferase enzymes in the mouse hematopoietic compartment have revealed that methylation is critical for long-term renewal and lineage differentiation of hematopoietic stem cells (Broske et al 2009, Trowbridge el al 2009). To better understand the role of DNA methylation in self-renewal and differentiation of hematopoietic cells, we characterized genome-wide DNA methylation in primary cells representing three distinct stages of hematopoiesis. We isolated mouse hematopoietic stem cells (HSC; Lin- Sca-1+ c-kit+), common myeloid progenitor cells (CMP; Lin- Sca-1- c-kit+), and erythroblasts (ERY; CD71+ Ter119+). Methyl Binding Domain Protein 2 (MBD2) is an endogenous reader of DNA methylation that recognizes DNA with a high concentration of methylated CpG residues. Recombinant MBD2 enrichment of DNA followed by massively-parallel sequencing was used to map and compare genome-wide DNA methylation patterns in HSC, CMP and ERY. Two biological replicates were sequenced for each cell type with total read counts ranging from 32,309,435–46,763,977. Model-based analysis of ChIP Seq (MACS) with a significance cutoff of p<10−5 was used to determine statistically significant peaks of methylation in each replicate. Globally, the number of methylation peaks was highest in HSC (85,797peaks), lower in CMP (50,638 peaks), and lowest in ERY (27,839 peaks). Comparison of the peaks in HSC, CMP and ERY revealed that only 2% of the peaks in CMP or ERY are absent in HSC indicating that the vast majority of methylation in HSC is lost during differentiation. Comparison of methylation with genomic features revealed that CpG islands associated with promoters are hypomethylated, while many non-promoter CpG islands are methylated. Furthermore, methylation of non-promoter associated CpG islands occurs infrequently in cell-type specific peaks but is more abundant in common methylation peaks. When the DNA methylation patterns were compared to mRNA expression, we found that as expected, proximal promoter sequences of expressed genes were hypomethylated in all three cell types, while methylation in the gene body positively correlated with gene expression in HSC and CMP. Utilizing de novo motif discovery we found a subset of transcription factor consensus binding motifs that were overrepresented in methylated sequences. Motifs for several ETS transcription factors, including GABPalpha and ELF1 were found to be overrepresented in cell-type specific as well as common methylated regions. Other transcription factor consensus sites, such as the NFAT factors involved in T-cell activation, were specifically overrepresented in the methylated promoter regions of CMP and ERY. Comparison of our methylation data with the occupancy of hematopoietic transcription factors in the HPC7 cell line, which is similar to CMP (Wilson et al 2010), revealed a significant anti-correlation between DNA methylation and the binding of Fli1, Lmo2, Lyl1, Runx1, and Scl. Our genome-wide survey provides new insights into the role of DNA methylation in hematopoiesis. Firstly, the methylation of CpG islands is associated with the most primitive hematopoietic cells and is unlikely to drive hematopoietic differentiation. We feel that the elevated genome-wide DNA methylation in HSC compared to CMP and ERY, combined with the positive association between gene body methylation and gene expression demonstrates that DNA methylation is a mark of cellular plasticity in HSC. Finally, the finding that transcription factor binding sites are over represented in the methylated sequences of the genome leads us to conclude that DNA methylation modulates key hematopoietic transcription factor programs that regulate hematopoiesis. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 653-653 ◽  
Author(s):  
Ying Qu ◽  
Andreas Lennartsson ◽  
Verena I. Gaidzik ◽  
Stefan Deneberg ◽  
Sofia Bengtzén ◽  
...  

Abstract Abstract 653 DNA methylation is involved in multiple biologic processes including normal cell differentiation and tumorigenesis. In AML, methylation patterns have been shown to differ significantly from normal hematopoietic cells. Most studies of DNA methylation in AML have previously focused on CpG islands within the promoter of genes, representing only a very small proportion of the DNA methylome. In this study, we performed genome-wide methylation analysis of 62 AML patients with CN-AML and CD34 positive cells from healthy controls by Illumina HumanMethylation450K Array covering 450.000 CpG sites in CpG islands as well as genomic regions far from CpG islands. Differentially methylated CpG sites (DMS) between CN-AML and normal hematopoietic cells were calculated and the most significant enrichment of DMS was found in regions more than 4kb from CpG Islands, in the so called open sea where hypomethylation was the dominant form of aberrant methylation. In contrast, CpG islands were not enriched for DMS and DMS in CpG islands were dominated by hypermethylation. DMS successively further away from CpG islands in CpG island shores (up to 2kb from CpG Island) and shelves (from 2kb to 4kb from Island) showed increasing degree of hypomethylation in AML cells. Among regions defined by their relation to gene structures, CpG dinucleotide located in theoretic enhancers were found to be the most enriched for DMS (Chi χ2<0.0001) with the majority of DMS showing decreased methylation compared to CD34 normal controls. To address the relation to gene expression, GEP (gene expression profiling) by microarray was carried out on 32 of the CN-AML patients. Totally, 339723 CpG sites covering 18879 genes were addressed on both platforms. CpG methylation in CpG islands showed the most pronounced anti-correlation (spearman ρ =-0.4145) with gene expression level, followed by CpG island shores (mean spearman rho for both sides' shore ρ=-0.2350). As transcription factors (TFs) have shown to be crucial for AML development, we especially studied differential methylation of an unbiased selection of 1638 TFs. The most enriched differential methylation between CN-AML and normal CD34 positive cells were found in TFs known to be involved in hematopoiesis and with Wilms tumor protein-1 (WT1), activator protein 1 (AP-1) and runt-related transcription factor 1 (RUNX1) being the most differentially methylated TFs. The differential methylation in WT 1 and RUNX1 was located in intragenic regions which were confirmed by pyro-sequencing. AML cases were characterized with respect to mutations in FLT3, NPM1, IDH1, IDH2 and DNMT3A. Correlation analysis between genome wide methylation patterns and mutational status showed statistically significant hypomethylation of CpG Island (p<0.0001) and to a lesser extent CpG island shores (p<0.001) and the presence of DNMT3A mutations. This links DNMT3A mutations for the first time to a hypomethylated phenotype. Further analyses correlating methylation patterns to other clinical data such as clinical outcome are ongoing. In conclusion, our study revealed that non-CpG island regions and in particular enhancers are the most aberrantly methylated genomic regions in AML and that WT 1 and RUNX1 are the most differentially methylated TFs. Furthermore, our data suggests a hypomethylated phenotype in DNMT3A mutated AML. Disclosures: No relevant conflicts of interest to declare.


2015 ◽  
Vol 33 (7_suppl) ◽  
pp. 432-432
Author(s):  
Gabriel Malouf ◽  
Jianping Zhang ◽  
Nizar M. Tannir ◽  
Erika Thompson ◽  
Jean-Philippe Spano ◽  
...  

432 Background: Long non-coding RNAs (lncRNA) play a key role in regulating cell physiology through different mechanisms including the recruitment of histone-modifying enzymes. Recently, lncRNA classification of clear-cell renal cell carcinomas (cc-RCC) established four subtypes with different clinico-pathological features. However, the interplay between those lncRNA and DNA methylation patterns remains unknown. Methods: We performed a genomic analysis of GENCODE lncRNAs in cc-RCC using The Cancer Genome Atlas (TCGA) molecular RNAseq profiles of 471 primary tumors. Furthermore, we reannotated data of DNA methylation combining 27K (n=173) and 450K (n=298) Infinium arrays. We described global correlations between lncRNA expression and DNA methylation and established lncRNA methylation subtypes based on distinct signatures. In addition, we identified lncRNA regulated by DNA methylation and associated with poor outcome. Results: We identified 2,138 lncRNAs which contain promoters located in CpG islands (CGI). Out of those, 59 (2.8%) lncRNAs showed DNA methylation in both normal and cancer as compared to 1,487 (69.5%) lncRNA which were unmethylated in both of them. Importantly, 592 (27.7%) lncRNAs gained DNA methylation in ccRCC and this was associated with the repression of the expression of 70 of them. Unsupervised clustering of lncRNA using the most variable DNA methylation probes revealed three robust subtypes associated with distinct outcome. Finally, by integrating cancer subtypes data with clinical information, we identified several lncRNAs which promoter methylation was associated with patient outcome. Conclusions: Our study represents the first integrative analysis of lncRNA and DNA methylation in ccRCC and provides new insights in the role of epigenetic alterations in kidney cancer. Furthermore, we identified a subset of lncRNA regulated epigenetically which may represent potential therapeutic targets.


2020 ◽  
Vol 14 (5) ◽  
pp. 933-950 ◽  
Author(s):  
Shamik Mitra ◽  
Martin Lauss ◽  
Rita Cabrita ◽  
Jiyeon Choi ◽  
Tongwu Zhang ◽  
...  

2018 ◽  
Vol 4 (2) ◽  
pp. 100040 ◽  
Author(s):  
Anna Wierczeiko ◽  
David Fournier ◽  
Hristo Todorov ◽  
Susanne Klingenberg ◽  
Kristina Endres ◽  
...  

Aging is a multi-factorial process, where epigenetic factors play one of the major roles in declines of gene expression and organic function. DNA methylation at CpG islands of promoters can directly change the expression of the neighbouring gene mostly through inhibition. Furthermore, it is known that DNA methylation patterns change during aging In our study, we investigated gene regulation through DNA methylation of genes up- and downregulated in long-lived people compared to a younger cohort. Our data revealed that comparatively highly methylated genes were associated with high expression in long-lived people (e.g. over 85). Genes with lower levels of methylation were associated with low expression. These findings might contradict the general model used to associate methylation status with expression. Indeed, we found that methylation in the promoter regions of all investigated genes is rather constant across different age groups, meaning that the disparity between methylation and expression only happens in older people. A potential explanation could be the impact of other epigenetic mechanisms, possibly related to stress.


Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 1646-1646
Author(s):  
Constance Regina Baer ◽  
Niroshan Nadarajah ◽  
Yasunobu Nagata ◽  
Tamara Alpermann ◽  
Genta Nagae ◽  
...  

Abstract Introduction: Myelodysplastic syndromes (MDS) are characterized by recurrent mutations, which contribute to the classification of patients into prognostic subgroups. Besides genetic lesions, MDS cells show aberrant DNA methylation, which is used as a therapeutic target for pharmacological DNA demethylation. Genetic and epigenetic aberrations are tightly linked: The most frequently mutated genes in MDS are key players in epigenetic pathways, including DNMT3A and TET2, which directly regulate DNA methylation. Additionally, other epigenetic master regulators (EZH2 and ASXL1) are recurrently mutated. Recent data in myeloid malignancies also suggested that cases with mutations in splicing factors (e.g. SRSF2) are characterized by a specific DNA methylation signature. Aim: To describe the role of recurrent mutations in MDS as prognostic markers and elucidate the mutation-specific DNA methylation pattern. Patients and Methods: By next-generation sequencing 786 bone marrow samples were analyzed for mutations in 14 genes. Patients (488 males, 298 females) had a median age of 73 (23- 91) years. The cohort was balanced according to WHO classification subtypes, karyotype risk groups and IPSS-R risk groups. For DNA methylation analysis, 196 samples were analyzed on the 450 K platform (Illumina). Differences between groups were calculated for individual probes by t-test with IMA script; differences with an adjusted p-value <0.01 were considered significant. Results: We recently suggested four prognostic subgroups in MDS (Haferlach et al.; Leukemia; 2014), defined by combining the mutation status of 14 genes (including the aforementioned ASXL1, EZH2 and TET2) with clinical parameters. First, we re-analyzed the data set with an extended follow-up time (median 45 vs. 32 months) and confirmed separation of the subgroups regarding overall survival (OS, p<0.001) in 786 patients. Of our initial cohort, we now selected 196 cases for a comparison of DNA methylation patterns in patients with and without different mutations. In 70/196 cases (36%) with at least one TET2 mutation we identified 2% aberrant DNA methylation. In line with the function of TET2 in DNA demethylation, 90% of aberrant DNA methylation was hypermethylation. Importantly, hypermethylation almost exclusively located outside of promoters (only 3% of fraction expected by array design), and was overrepresented by twofold in regions with enhancer function. Only 6% of hypermethylated CpGs were located in CpG Islands (expected background: 31%). In contrast, in 31/196 (16%) DNMT3A mutated cases, only 0.02% abnormal DNA methylation was observed and 94% was hypomethylation, as expected by the role of DNMT3A to regulated DNA de novo methylation. In 42/196 (21%) of ASXL1 mutated cases, a methylation difference of 2% was identified with 67% hypermethylation, whereas in 13/196 (7%) EZH2 mutated cases only minute recurrent DNA methylation differences were observed (0.01%). Next, we analyzed the effect of mutated splicing factors on DNA methylation. In both 67/196 (34%) SF3B1 and 39/196 (20%) SRSF2 mutated cases, we identified strong DNA methylation differences (>3%), however with opposite direction (SF3B1: 99% hypomethylation, SRSF2: 91% hypermethylation). Finally, in 58/196 (30%) patients with an aberrant karyotype we could identify that 1% of probes showed DNA methylation changes, which was almost exclusively hypermethylation. Conclusion: 1) We confirmed the prognostic capacity of our previously suggested scoring model including the mutation status of 14 genes in MDS. 2) The mutation differences resulted in a mutation specific epigenetic signature regarding the degree, direction and localization of aberrant DNA methylation. 3) For the most frequently mutated gene in MDS, TET2, DNAhypermethylation is found outside of the classically analyzed promoter regions and CpG Islands, and enriched in enhancer regions. 4) MDS is treated with drugs that alter DNA methylation. The understanding of mutation specific DNA methylation patterns would allow to choose the right genomic loci to monitor the DNA methylation reduction under treatment. This would be a key step toward a deeper understanding of drug function or response. Disclosures Baer: MLL Munich Leukemia Laboratory: Employment. Nadarajah:MLL Munich Leukemia Laboratory: Employment. Alpermann:MLL Munich Leukemia Laboratory: Employment. Haferlach:MLL Munich Leukemia Laboratory: Employment, Equity Ownership. Kern:MLL Munich Leukemia Laboratory: Employment, Equity Ownership. Haferlach:MLL Munich Leukemia Laboratory: Employment, Equity Ownership.


2019 ◽  
Vol 3 (Supplement_1) ◽  
Author(s):  
Fayth Miles ◽  
Andrew Mashchak ◽  
Gary Fraser

Abstract Objectives It is unclear how diets differing in content of animal products affect DNA methylation patterns. We sought to determine if DNA methylation patterns differed between vegans and non-vegetarians in the Adventist Health Study-2 (AHS-2) cohort. Methods DNA methylation was profiled using the Infinium Human Methylation 450 K BeadChip in white blood cells of 137 participants in the AHS-2 cohort classified as vegan (57) or non-vegetarian (80) based on validated diet history data. Linear regression models were generated to test associations between diet pattern and methylation, where the response variable represented methylation intensity for individual CpG sites. This was repeated for sites separated into gene regions or in relation to CpG islands. CpG methylation was also averaged across each gene. The permutation-identified null distribution, false discovery rate of Storey et al was used to adjust for multiple testing. Results A total of 53,809 individual CpG sites were estimated to be differentially methylated (non-null). Of these, with this small sample, we could individually identify only up to 5.5% (differing by gene region) at FDR <0.05. A vegan diet was associated almost exclusively with hypomethylation of individual CpG sites. Significant CpG sites numbered 2504 within CpG islands, 749 and 13 CpG sites 0–200 base pairs or 201–1500 base pairs upstream of the transcription start site (TSS200 and TSS1500), respectively, and 51 and 458 sites falling within the 5’ UTR and first exon, respectively. The greatest difference in methylation was observed for sites mapping to TSS200 and CpG islands, where methylation was 9.2% lower in vegans. No differences were found when CpG methylation was averaged to obtain a cumulative value for all sites (no FDR criterion) within each gene region. A total of 328 genes (averaging all methylation regions) were significantly hypomethylated in vegans relative to non-vegetarians at FDR <0.05. The greatest number of genes were hypomethylated when considering methylation of only CpG islands, followed by the TSS200, first exon, and TSS1500 regions. Conclusions Our findings suggest substantial differences in methylation of CpG sites and genes, particularly in regulatory regions, between vegans and non-vegetarians, with a preponderance of hypomethylation among vegans. Funding Sources National Institute of Health, National Cancer Institute, World Cancer Research Fund (UK).


2019 ◽  
Vol 40 (11) ◽  
pp. 1332-1340
Author(s):  
Venkatrao Vantaku ◽  
Chandra Sekhar Amara ◽  
Danthasinghe Waduge Badrajee Piyarathna ◽  
Sri Ramya Donepudi ◽  
Chandrashekar R Ambati ◽  
...  

Abstract Racial/ethnic disparities have a significant impact on bladder cancer outcomes with African American patients demonstrating inferior survival over European-American patients. We hypothesized that epigenetic difference in methylation of tumor DNA is an underlying cause of this survival health disparity. We analyzed bladder tumors from African American and European-American patients using reduced representation bisulfite sequencing (RRBS) to annotate differentially methylated DNA regions. Liquid chromatography–mass spectrometry (LC-MS/MS) based metabolomics and flux studies were performed to examine metabolic pathways that showed significant association to the discovered DNA methylation patterns. RRBS analysis showed frequent hypermethylated CpG islands in African American patients. Further analysis showed that these hypermethylated CpG islands in patients are commonly located in the promoter regions of xenobiotic enzymes that are involved in bladder cancer progression. On follow-up, LC-MS/MS revealed accumulation of glucuronic acid, S-adenosylhomocysteine, and a decrease in S-adenosylmethionine, corroborating findings from the RRBS and mRNA expression analysis indicating increased glucuronidation and methylation capacities in African American patients. Flux analysis experiments with 13C-labeled glucose in cultured African American bladder cancer cells confirmed these findings. Collectively, our studies revealed robust differences in methylation-related metabolism and expression of enzymes regulating xenobiotic metabolism in African American patients indicate that race/ethnic differences in tumor biology may exist in bladder cancer.


Sign in / Sign up

Export Citation Format

Share Document