scholarly journals Differential DNA Methylation of Gene Promoters in Small B-Cell Lymphomas

2005 ◽  
Vol 124 (3) ◽  
pp. 430-439 ◽  
Author(s):  
Juyuan Guo ◽  
Matthias Burger ◽  
Inko Nimmrich ◽  
Sabine Maier ◽  
Evelyne Becker ◽  
...  
Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 831-831
Author(s):  
Megan Ryan ◽  
Leandro Cerchietti ◽  
Maria E. Figueroa ◽  
John Greally ◽  
Ari Melnick

Abstract DNA methyltransferase inhibitor drugs (MTIs) such as decitabine can overcome gene silencing due to aberrant hypermethylation of gene promoters. Presumably, this effect is responsible for the therapeutic activity of MTIs as clinically demonstrated in myelodysplasias (MDS) and leukemias. Other tumors such as diffuse large B-cell lymphomas (DLBCLs) can also present with aberrant promoter hypermethylation. However, it is currently difficult to prospectively identify patients likely to respond to MTIs, since specific methylation markers or signatures have not yet been identified. We predicted that decitabine would have anti-lymphoma activity in a subset of DLBCLs, and that these cases would exhibit specific methylation signatures predictive of response to these drugs. To determine whether this is the case we first exposed a panel of 7 DLBCL cell lines (Ly1, Ly7, Ly10, SU-DHL6, Farage, Pfeiffer and Toledo) to increasing concentrations of decitabine (0.5, 1, 2.5, 5, 10, 50 and 100 μM) administered after synchronization by 12 hr serum starvation. Viability was assessed after 48 hr of culture by MTS-based assay and Trypan blue exclusion. The IC25 and IC50 were calculated for all cell lines by constructing dose-response curves. The IC25 was used to discriminate sensitive (6.3 ± 1.2 μM) vs. resistant (49.4 ± 5 μM, p < 0.01) cell lines. Interestingly, there was no correlation between MTI sensitivity and DLBCL subtype as defined by recent gene expression profiling classification efforts (i.e. GCB vs. ABC, or BCR vs. OxPhos). To identify the methylation signatures of these DLBCL cells we used a method that we developed for genome-wide DNA methylation quantification called HELP (HpaII tiny fragment Enrichment by LM-PCR). HELP is based on comparative Msp1 and HpaII digestion of genomic DNA, followed by size specific amplification and co-hybridization to custom high-density oligonucleotide arrays designed to provide uniform data collection over 25,000 promoters. HELP compares favorably to other high throughput methods in that it is highly reproducible (R > 0.98) and has an extremely robust signal-to-noise ratio. DNA was collected from the DLBCL cells for HELP prior to drug treatment. Most significantly we found that unsupervised (i.e. unbiased) clustering of DNA methylation profiles could readily segregate decitabine resistant vs. sensitive DLBCL cell lines. Correspondence analysis clearly identified a methylation signature consisting of 133 differentially methylated genes that distinguishes between decitabine sensitive and resistant cells. Most of these appeared to be functionally relevant including such genes as Caspase-9, RARB, JUNB, and ELK1. Biological assays to determine the contribution of these genes to the phenotype are underway. Taken together, our data suggest that MTIs might be effective in a cohort of DLBCL cases that exhibit the specific methylation signature that we have identified. Prospective evaluation of the predictive value of this signature may allow optimal selection of patients for clinical trials with these agents.


Author(s):  
Daniel Xia ◽  
Alberto Jose Leon ◽  
Jiong Yan ◽  
Anjali Silva ◽  
Mehran Bakhtiari ◽  
...  

2010 ◽  
Vol 207 (9) ◽  
pp. 1939-1950 ◽  
Author(s):  
Anne Y. Lai ◽  
Mehrnaz Fatemi ◽  
Archana Dhasarathy ◽  
Christine Malone ◽  
Steve E. Sobol ◽  
...  

Aberrant DNA methylation commonly occurs in cancer cells where it has been implicated in the epigenetic silencing of tumor suppressor genes. Additional roles for DNA methylation, such as transcriptional activation, have been predicted but have yet to be clearly demonstrated. The BCL6 oncogene is implicated in the pathogenesis of germinal center–derived B cell lymphomas. We demonstrate that the intragenic CpG islands within the first intron of the human BCL6 locus were hypermethylated in lymphoma cells that expressed high amounts of BCL6 messenger RNA (mRNA). Inhibition of DNA methyltransferases decreased BCL6 mRNA abundance, suggesting a role for these methylated CpGs in positively regulating BCL6 transcription. The enhancer-blocking transcription factor CTCF bound to this intronic region in a methylation-sensitive manner. Depletion of CTCF by short hairpin RNA in neoplastic plasma cells that do not express BCL6 resulted in up-regulation of BCL6 transcription. These data indicate that BCL6 expression is maintained during lymphomagenesis in part through DNA methylation that prevents CTCF-mediated silencing.


2009 ◽  
Vol 43 (5) ◽  
pp. 420 ◽  
Author(s):  
Sung Sun Kim ◽  
Young Hyo Choi ◽  
Chang Woo Han ◽  
Yoo Duk Choi ◽  
Youngkyu Park ◽  
...  

PLoS Genetics ◽  
2013 ◽  
Vol 9 (1) ◽  
pp. e1003137 ◽  
Author(s):  
Subhajyoti De ◽  
Rita Shaknovich ◽  
Markus Riester ◽  
Olivier Elemento ◽  
Huimin Geng ◽  
...  

Blood ◽  
2009 ◽  
Vol 113 (11) ◽  
pp. 2478-2487 ◽  
Author(s):  
Matthew J. Frank ◽  
David W. Dawson ◽  
Steven J. Bensinger ◽  
Jason S. Hong ◽  
Wendy M. Knosp ◽  
...  

B-cell lymphoma is the most common immune system malignancy. TCL1 transgenic mice (TCL1-tg), in which TCL1 is ectopically expressed in mature lymphocytes, develop multiple B- and T-cell leukemia and lymphoma subtypes, supporting an oncogenic role for TCL1 that probably involves AKT and MAPK-ERK signaling pathway augmentation. Additional, largely unknown genetic and epigenetic alterations cooperate with TCL1 during lymphoma progression. We examined DNA methylation patterns in TCL1-tg B-cell tumors to discover tumor-associated epigenetic changes, and identified hypermethylation of sprouty2 (Spry2). Sprouty proteins are context-dependent negative or positive regulators of MAPK-ERK pathway signaling, but their role(s) in B-cell physiology or pathology are unknown. Here we show that repression of Spry2 expression in TCL1-tg mouse and human B-cell lymphomas and cell lines is associated with dense DNA hypermethylation and was reversed by inhibition of DNA methylation. Spry2 expression was induced in normal splenic B cells by CD40/B-cell receptor costimulation and regulated a negative feedback loop that repressed MAPK-ERK signaling and decreased B-cell viability. Conversely, loss of Spry2 function hyperactivated MAPK-ERK signaling and caused increased B-cell proliferation. Combined, these results implicate epigenetic silencing of Spry2 expression in B lymphoma progression and suggest it as a companion lesion to ectopic TCL1 expression in enhancing MAPK-ERK pathway signaling.


Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 1045-1045 ◽  
Author(s):  
Maria Del Pilar Dominguez ◽  
Matt Teater ◽  
David Redmond ◽  
Zhengming Cheng ◽  
Jayanta Chaudhuri ◽  
...  

Abstract Diffuse large B-cell lymphomas (DLBCLs) are aggressive tumors derived from germinal center B cells (GC) B cells. Previous work from our group established that inferior outcome in DLBCL is associated with higher degrees of intra-tumor and inter-tumor cytosine methylation heterogeneity, although the molecules driving this epigenetic perturbation are unknown. We investigated the contribution of activation-induced cytidine deaminase (AICDA) to cytosine methylation heterogeneity in DLBCLs. AICDA is highly expressed in GC B cells where it drives somatic hypermutation (SHM) of immunoglobulin (Ig) and non-Ig genes. AICDA is also expressed in DLBCLs and high level of AICDA in CHOP-treated DLBCL patients is associated with unfavorable prognosis. In addition to mutagenesis, AICDA can cause DNA demethylation through the removal of modified cytosine residues. Indeed, we previously reported that GC B cells manifest a marked AICDA-mediated DNA hypomethylation signature as well as a high degree of epigenetic heterogeneity as compared to other B-cell subsets. Thus, we hypothesized that AICDA could contribute to the progression of GC-derived lymphomas by facilitating epigenetic plasticity through the redistribution of cytosine methylation. We overexpressed AICDA in VavP-Bcl2 transgenic mice -which develop B cell lymphomas of GC origin- using an AICDA-expressing retroviral vector (VavP-Bcl2+Aicda) vs empty vector (VavP-Bcl2) as control and transplanted them into lethally irradiated recipient mice. We observed more aggressive lymphomas based on greater disruption of the splenic architecture and higher degree of B cell infiltration in organs such as lung, liver and kidney in VavP-Bcl2+Aicda mice (n=7) compared to VavP-Bcl2 mice (n=6). Notably, the overexpression of AICDA reduced significantly the lifespan of the mice (Log-rank test p=0.0289) with a median survival of 214 days for VavP-Bcl2+Aicda mice (n=10) and 293 days for VavP-Bcl2 animals (n=9). Neoplastic B cells from VavP-Bcl2+Aicda and VavP-Bcl2 micedisplayed similar SHM rate in Ig genes (JH4 and Sm) and AICDA off-targets (Bcl6, Cd83 and Pax5), suggesting that the more aggressive phenotype is not likely due to increased mutagenesis. We then profiled the DNA methylation landscape of these lymphomas using enhanced reduced representation bisulfite sequencing. We analyzed the DNA methylation level and heterogeneity of all represented CpGs by calculating the mean methylation difference and interquartile range (IQR) between VavP-Bcl2+Aicda and VavP-Bcl2 tumors. A principal component analysis of all CpGs, each one represented by its mean methylation and IQR differences across replicates, revealed methylation loss and increased intertumor heterogeneity within VavP-Bcl2+Aicda methylomes compared to VavP-Bcl2. AICDA-perturbed CpGs (n=49,750) in particular showed significant intratumor heterogeneity (p<1e-300) in AICDA overexpressing tumors. These altered CpGs were depleted in promoters and enriched in introns and intergenic regions. We observed a remarkably similar pattern of hypomethylation and increased methylation heterogeneity in Aicda+/+ GC B cells compared to Aicda-/- GC B cells (AICDA-perturbed CpGs n=64,323) and also in primary DLBCLs with high AID compared to low AID expression (AICDA-perturbed CpGs n=37,557), suggesting a conserved epigenetic function of AICDA in GC B cells and human and mouse GC-derived lymphomas. We performed RNA sequencing on BCL2-driven lymphomas and found that expressed genes containing AICDA-perturbed CpGs displayed significantly lower expression in VavP-Bcl2+Aicda compared to VavP-Bcl2 tumors (gene set enrichment analysis (GSEA) NES=-1.68, FDR<0.001). Pathway analysis revealed that these AICDA-epigenetic target genes were associated with MAPK- and KMT2D-signaling pathways, which are related to B cell activation and lymphomagenesis. Similarly to mouse tumors, genes enriched for AICDA-perturbed CpG in human lymphomas exhibited lower expression in AID high DLBCLs compared to AICDA low DLBCLs (GSEA NES=-1.65, FDR<0.001) and were associated with MAPK and KMT2D pathways. These results demonstrate that AICDA acts as a methylome modifier in GC-derived lymphomas, introducing epigenetic plasticity and leading to gene expression changes, thus contributing to clonal evolution and higher adaptability to an evolving environment. Disclosures Melnick: Janssen: Research Funding.


Blood ◽  
2021 ◽  
Vol 138 (Supplement 1) ◽  
pp. 3289-3289
Author(s):  
Karina Close ◽  
Hannah Armes ◽  
Emil Kumar ◽  
Findlay Bewicke-Copley ◽  
Adam Cribbs ◽  
...  

Abstract Introduction Germinal centre (GC) lymphomas Follicular Lymphoma (FL) and Diffuse Large B Cell Lymphoma (DLBCL) are strikingly dependent on mutations affecting chromatin regulators. The majority of FL tumours harbour multiple mutated chromatin-regulatory genes, including loss-of-function mutations in KMT2D (80%), CREBBP (60%) and gain-of-function mutations in EZH2 (25%), whilst these are also frequently mutated within the GC B-cell (GCB) subtype of DLBCL. The FDA approval of EZH2-inhibitor Tazemetostat for relapsed/refractory FL exemplifies the potential and the need to broaden our search for novel avenues targeting epigenetic reprogramming in GC lymphomas. Hence, in this study, we sought to identify novel epigenetic vulnerabilities and biological interactions in GC B cell lymphomas. Methods and Results: 25 compounds targeting 18 histone binding/modifying enzymes were tested across 12 GCB-DLBCL cell lines harbouring differing degrees of epigenetic dysregulation. The two most potent cytotoxic compounds (GSK-J4 and KDOBA67) both target the H3K27me3 demethylase, KDM6. The KDM6 family consists of KDM6A (UTX) and KDM6B (JMJD3), both of which demethylate H3K27me3/H3K27me2 to lower methylation states, and KDM6C (UTY) which has negligible enzymatic activity. Indeed, KDM6 inhibition (KDM6i) with GSK-J4 increased levels of the inhibitory chromatin mark H3K27me3 and simultaneously reduced H3K27me1/me2, while we observed no global changes in H3K4me3. The induction of apoptosis in the cell lines following either inhibition of the 'eraser' KDM6 (leading to increased H3K27me3) or the opposing 'writer' EZH2 (lowering H3K27me3) appears paradoxical at first glance, given the EZH2 gain-of-function mutations seen in FL/DLBCL and known efficacy of EZH2 inhibitors. However, RNA-sequencing analysis of GSK-J4-treated cells revealed no significant overlap with published EZH2 mutation or EZH2 inhibition signatures from GC lymphoma models, implying that KDM6i-regulated H3K27me3 peaks do not overlay gene promoters targeted by EZH2 in the GC reaction. The most significant gene regulation by KDM6i was a significant up-regulation of 6 members of the metallothionein (MT) family within 4-hours of treatment, which was sustained for 72-hours (MT1E, MT1F, MT1G, MT1H, MT1X and MT2A at 4 hours log2FoldChange 5.1-12.1, average p adj 3.4E-06). These metal-binding proteins cluster on chromosome 16 and their up-regulation correlated with up-regulation of the zinc transporter SLC30A1, suggesting a potential biological relationship between KDM6 enzymes and zinc homeostasis, with both isoforms containing a zinc-binding domain. Targeted validation across 6 DLBCL cell lines also showed MT up-regulation correlated with sensitivity to GSK-J4, whilst siRNA-mediated knockdown of KDM6A or KDM6B individually recapitulated the up-regulation of MT genes, albeit to a lower extent. ChIP-qPCR profiling at the MT loci for H3K27me3 and H3K4me3 revealed no significant changes at 4 or 24 hours; considering that expression of MT genes increase within 60 minutes of drug exposure, suggesting these changes may be induced by an indirect mechanism of KDM6A and/or KDM6B activity. Using ENCODE data of 4 cell lines included in our series (DOHH-2, OCI-LY-1, SU-DHL-6, KARPAS-422), we observe a link between GSK-J4 sensitivity, up-regulation of MT genes and significantly lower basal enrichment for H3K27ac at the MT loci. This distinction may reflect altered 3D chromatin conformation that render only sensitive cell lines permissive to regulation by KDM6i, with H3K27ac having recently been shown to modulate enhancer positioning and interactions in DLBCL cell lines. Conclusion: Screening epigenetic-targeting compounds in GC lymphomas has identified KDM6 as a particularly promising candidate target. Overall, we describe up-regulation of MT genes as one potential mechanism of action, which correlates with response in DLBCL cell lines and suggests that KDM6 members may regulate H3K27me3 in a manner different from EZH2. In our current model we propose, that in GC-lymphomas KDM6A may have tumour suppressive roles whilst KDM6B is required for cell survival. Given KDM6A is frequently lost in cancer and Kabuki syndrome, a congenital developmental disorder caused by germline mutations in KMT2D or KDM6A, our results highlight the importance of establishing what roles the KDM6 family play in lymphoma pathogenesis. Disclosures Fitzgibbon: Epizyme: Research Funding.


Sign in / Sign up

Export Citation Format

Share Document