Genome-Wide Promoter Methylation Profiling Of Splenic Marginal Zone Lymphoma (SMZL) Identifies Two Subgroups Of Patients With Distinct Genetic and Biologic Features and Different Outcomes

Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 77-77
Author(s):  
Alberto J. Arribas ◽  
Andrea Rinaldi ◽  
Ivo Kwee ◽  
Afua A. Mensah ◽  
Luciano Cascione ◽  
...  

Abstract SMZL usually has an indolent clinical course. Transformation to aggressive lymphoma occurs in only 4-5% of cases. Tools to identify patients facing a more aggressive clinical course and targeted therapies are needed. In the last years, we have characterized SMZL by DNA profiling (Blood 2011) and RNA/miRNA expression profiling (Mod Path 2013), and shown that somatic mutations of NOTCH2 and other genes involved in the pathway are frequent and mutually exclusive with mutations affecting NKFB pathway (JEM 2012). Here, we report genome-wide DNA promoter methylation profiling of 137 SMZL samples with its clinical and biologic implications. Methods A test series of 101 samples, including 3 SMZL cell lines (Karpas1718, VL51, SSK41) and 98 clinical SMZL cases from 10 Centers and, a validation set of 36 SMZL cases from 2 additional Centers were analyzed with the Illumina Infinium HumanMethylation27 arrays, as described (BJH 2013). Gene GEP was available for a subset of 11/101 cases. Methylation profiles were correlated with the presence of the copy number aberrations (CNAs) and somatic mutations recurrent in SMZL, IGHV mutational status, HCV infection and clinical data. Probes mapped outside CpG islands were discarded. Supervised analyses were performed using a t-test on the continuous beta values, followed by multiple test correction. For Fisher’s test, probes were classified into unmethylated (beta-values <0.3) or methylated (beta-value >0.3). Results Unsupervised clustering analysis of 101 SMZL samples identified 2 clusters. Cluster one (23/101, 23%) comprised the 3 cell lines and included cases characterized by a generalized higher degree of promoter methylation (High-M), and had a significantly poorer overall survival (OS) than the remaining 79/101 (77%) cases (Low-M) (P=0.048; HR 2.4; 95% CI, 1-5.8). To better understand the biology underlying this clustering, supervised analysis comparing High-M to Low-M cases was performed. By combining t- and Fisher's tests, 3200 gene probes showed differential methylation status between High-M and Low-M lymphomas. This probes signature was significantly enriched of promoters of promoter regions of PRC2-complex targets, genes involved in chromatin remodeling, and HOX and SOX family genes, and genes down-regulated by CDH1. Genes coding for subunits of the PRC2-complex (EZH2, EED, SUZ12) appeared unmethylated in their promoter regions and over-expressed by GEP in the High-M set, while EZH2-target genes and genes harboring the H3K27me3 mark had methylated promoters and down-regulated expression. A number of reported tumor-suppressor genes were highly methylated in High-M cases, including KLF4, DAPK1, CDKN2A/B, CDKN1C, CDH1, CDH2 and TIMP3. Known pro-survival lymphoma genes, such as SYK, MCL1, CARD11, BIRC5, BTK, BCL10, FOXP1, CD40, TACI and TCL1A/B, were un-methylated and over-expressed in High-M cases. The prognostic relevance of the High-M phenotype was confirmed in an independent validation set. An unsupervised clustering using the top-100 differentially methylated probes obtained by t-test in the test series identified two clusters with one (11/36, 30%) bearing methylation profile similar to High-M and a poorer OS (P=0.038). All the samples were then pooled to improve the statistical power of further analyses regarding the impact of methylation on the clinical course and on the association with biologic variables. High-M was significantly associated with poorer OS (P=0.008; HR 2.58; 95% CI, 1.2-5.4), transformation into aggressive lymphoma (33% vs 0.02%, P<0.001) and with 7q31-33 loss (38% vs 16%, P=0.026). In addition, High-M showed a higher frequency of NOTCH-pathway somatic mutations (39% vs 16%, P=0.063). HCV was more frequent in Low-M (20% vs 4%, P=0.058). In a Cox regression model adjusted for the variables significantly associated with outcome at univariate analysis [High-M, age>60, Interguppo Italiano Linfomi (IIL) SMZL score, HCV infection], High-M (HR 16; 95% CI, 1.3-201) and IIL SMZL score (HR 4.5; 95% CI, 1.3-15) retained their prognostic significance on OS (P<0.001). Conclusions Genome-wide promoter-methylation profiling in splenic MZL identified a subset of patients with poorer OS, characterized by a high promoter methylation and by specific genetic and biologic features. These data may provide the rational to explore epigenetic drugs in SMZL. *, AJA and AR equally contributed. Disclosures: No relevant conflicts of interest to declare.

Cancers ◽  
2019 ◽  
Vol 11 (10) ◽  
pp. 1466
Author(s):  
Su-Jin Jeong ◽  
Kang-Hoon Lee ◽  
A-Reum Nam ◽  
Je-Yoel Cho

Genome-wide methylation profiling is used in breast cancer (BC) studies, because DNA methylation is a crucial epigenetic regulator of gene expression, involved in many diseases including BC. We investigated genome-wide methylation profiles in both canine mammary tumor (CMT) tissues and peripheral blood mononuclear cells (PBMCs) using reduced representation bisulfite sequencing (RRBS) and found unique CMT-enriched methylation signatures. A total of 2.2–4.2 million cytosine–phosphate–guanine (CpG) sites were analyzed in both CMT tissues and PBMCs, which included 40,000 and 28,000 differentially methylated regions (DMRs) associated with 341 and 247 promoters of differentially methylated genes (DMGs) in CMT tissues and PBMCs, respectively. Genes related to apoptosis and ion transmembrane transport were hypermethylated, but cell proliferation and oncogene were hypomethylated in tumor tissues. Gene ontology analysis using DMGs in PBMCs revealed significant methylation changes in the subset of immune cells and host defense system-related genes, especially chemokine signaling pathway-related genes. Moreover, a number of CMT tissue-enriched DMRs were identified from the promoter regions of various microRNAs (miRNAs), including cfa-mir-96 and cfa-mir-149, which were reported as cancer-associated miRNAs in humans. We also identified novel miRNAs associated with CMT which can be candidates for new miRNAs associated with human BC. This study may provide new insight for a better understanding of aberrant methylation associated with both human BC and CMT, as well as possible targets for methylation-based BC diagnostic markers.


Genomics Data ◽  
2015 ◽  
Vol 3 ◽  
pp. 94-96 ◽  
Author(s):  
Aaron L. Statham ◽  
Phillippa C. Taberlay ◽  
Theresa K. Kelly ◽  
Peter A. Jones ◽  
Susan J. Clark

2021 ◽  
Vol 13 (1) ◽  
Author(s):  
Sarah Bazzocco ◽  
Higinio Dopeso ◽  
Águeda Martínez-Barriocanal ◽  
Estefanía Anguita ◽  
Rocío Nieto ◽  
...  

Abstract Background Cancer initiation and progression are driven by genetic and epigenetic changes. Although genome/exome sequencing has significantly contributed to the characterization of the genetic driver alterations, further investigation is required to systematically identify cancer driver genes regulated by promoter hypermethylation. Results Using genome-wide analysis of promoter methylation in 45 colorectal cancer cell lines, we found that higher overall methylation levels were associated with microsatellite instability (MSI), faster proliferation and absence of APC mutations. Because epigenetically silenced genes could represent important oncogenic drivers, we used mRNA expression profiling of colorectal cancer cell lines and primary tumors to identify a subset of 382 (3.9%) genes for which promoter methylation was negatively associated with gene expression. Remarkably, a significant enrichment in zinc finger proteins was observed, including the transcriptional repressor ZBTB18. Re-introduction of ZBTB18 in colon cancer cells significantly reduced proliferation in vitro and in a subcutaneous xenograft mouse model. Moreover, immunohistochemical analysis revealed that ZBTB18 is frequently lost or reduced in colorectal tumors, and reduced ZBTB18 expression was found to be associated with lymph node metastasis and shorter survival of patients with locally advanced colorectal cancer. Conclusions We identified a set of 382 genes putatively silenced by promoter methylation in colorectal cancer that could significantly contribute to the oncogenic process. Moreover, as a proof of concept, we demonstrate that the epigenetically silenced gene ZBTB18 has tumor suppressor activity and is a novel prognostic marker for patients with locally advanced colorectal cancer.


Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 1359-1359
Author(s):  
Davide Rossi ◽  
Andrea Rinaldi ◽  
Ivo Kwee ◽  
Luca Arcaini ◽  
Marco Lucioni ◽  
...  

Abstract Abstract 1359 RS represents the development of DLBCL in the context of chronic lymphocytic leukemia (CLL). The pathogenesis of RS is still largely unknown. We have previously shown that RS lacks many of the typical genetic lesions recurrently observed in de novo DLBCL (Rossi et al, Blood 2011). Here, we report genome-wide promoter methylation profiling of RS and compare it with the methylation profile of de novo DLBCL, and non-transformed CLL. Methods. The study included 21 RS, 10 clonally related CLL phases (seven paired with RS samples), 9 de novo non-GC DLBCL, 6 non-transformed CLL and 6 normal peripheral blood CD19+ B-cells. All RS were classified as DLBCL, showed a non-germinal center (GC) phenotype, and lacked EBV infection. Both RS and DLBCL DNA was extracted from diagnostic frozen lymph node biopsies. DNA samples underwent bisulfite treatment with the EZ DNA methylation kit and were hybridized on Illumina Infinium HumanMethylation27 arrays, that enables the interrogation of more than 27,000 CpG island over the entire human genome. Probes were discarded if they: i) mapped outside CpG islands; ii) presented a standard deviation <0.10 across all the samples; or iii) were always non-methylated, always fully methylated, or always partially methylated. For clustering and for unpaired t-tests, probes mapped on sex chromosomes were excluded. Supervised analysis of differential methylation between groups was performed using a t-test on the continuous beta values, followed by multiple test correction (MTC). A paired t-test was utilized for comparing the methylation profile between the RS phase and the matched CLL-phase. Probes showing a q-value <0.1 were defined differentially methylated. Results. By unsupervised clustering, RS samples separated from both de novo DLBCL, that instead clustered in a single group, and non-transformed CLL. There were over 3,000 probes differentially methylated between RS and de novo DLBCL. The 443 probes showing higher methylation in RS were significantly enriched of promoter regions of genes involved in TP53 signaling (P=6E-03) and cell cycle regulation (P=5E-02) pathways. The 2,687 probes showing higher methylation in de novo DLBCL were significantly enriched of promoter regions of genes involved in histone modification, including genes harboring the H3K27me3 mark (P < 1.0E–16), and of genes that are targets of the EED and SUZ12 Polycomb proteins (P < 1.0E–16). Analysis of paired CLL/RS sequential samples revealed an extensive overlap between the methylation profile of the CLL phase and the transformed RS phase, suggesting that changes in the methylation pattern are an early event in RS pathogenesis that occurred already at the time of the initial CLL clone. Two single probes in the promoter regions of the OSM and S1PR4/EDG6 genes were differentially methylated in CLL/RS sequential samples and, therefore, might have contributed to the acquisition of the aggressive clinico-pathologic phenotype of RS. OSM codes for oncostatin M, which inhibits cell proliferation and induces cell differentiation and apoptosis. S1PR4/EDG6 is a gene specifically expressed in the lymphoid tissue and involved in cell migration. Both OSM and S1PR4/EDG6 promoter regions were unmethylated in the chronic CLL phase, and became methylated in the paired aggressive RS phase, suggesting the acquisition of methylation during transformation. CLL that subsequently transformed to RS differed from CLL that never transformed to RS for 2 probes showing a higher and 2 a lower degree of methylation. Since changes in the methylation pattern are an early event in RS, this observation prompts the investigation of the methylation status of DLX5 and GBGT1, the two genes with promoter regions showing a higher degree of methylation, as biomarkers allowing the early identification of CLL that will transform to RS. Conclusions. RS clearly differ from de novo DLBCL in terms of methylation profile. The overrepresentation of epigenetic changes affecting genes of the TP53 pathway, along with the significantly higher prevalence of TP53 structural abnormalities, might explain the differences in chemosensitivity between RS and de novo DLBCL. Epigenetic changes are an early events in RS pathogenesis, suggesting that, similarly to what shown in transformed follicular lymphoma, lesions initiating transformation are acquired by a cell belonging to the initial tumor clone, rather being progressively accumulated during disease course. Disclosures: No relevant conflicts of interest to declare.


2020 ◽  
Vol 11 ◽  
Author(s):  
Shengyue Li ◽  
Lei Wang ◽  
Qiang Zhao ◽  
Zhihao Wang ◽  
Shuxian Lu ◽  
...  

As one of the most malicious cancers, pancreatic cancer is difficult to treat due to the lack of effective early diagnosis. Therefore, it is urgent to find reliable diagnostic and predictive markers for the early detection of pancreatic cancer. In recent years, the detection of circulating cell-free DNA (cfDNA) methylation in plasma has attracted global attention for non-invasive and early cancer diagnosis. Here, we carried out a genome-wide cfDNA methylation profiling study of pancreatic ductal adenocarcinoma (PDAC) patients by methylated DNA immunoprecipitation coupled with high-throughput sequencing (MeDIP-seq). Compared with healthy individuals, 775 differentially methylated regions (DMRs) located in promoter regions were identified in PDAC patients with 761 hypermethylated and 14 hypomethylated regions; meanwhile, 761 DMRs in CpG islands (CGIs) were identified in PDAC patients with 734 hypermethylated and 27 hypomethylated regions (p-value &lt; 0.0001). Then, 143 hypermethylated DMRs were further selected which were located in promoter regions and completely overlapped with CGIs. After performing the least absolute shrinkage and selection operator (LASSO) method, a total of eight markers were found to fairly distinguish PDAC patients from healthy individuals, including TRIM73, FAM150A, EPB41L3, SIX3, MIR663, MAPT, LOC100128977, and LOC100130148. In conclusion, this work identified a set of eight differentially methylated markers that may be potentially applied in non-invasive diagnosis of pancreatic cancer.


2020 ◽  
Author(s):  
Michael Wells ◽  
Karam P. Asmaro ◽  
Thais S. Sabedot ◽  
Tathiane M. Malta ◽  
Maritza S. Mosella ◽  
...  

AbstractGenome-wide DNA methylation aberrations are pervasive and associated with clinicopathological features across pituitary tumors (PT) subtypes. The feasibility to detect CpG methylation abnormalities in circulating cell-free DNA (cfDNA) has been reported in central nervous system tumors other than PT. Here, we aimed to profile and identify methylome-based signatures in the serum of patients harboring PT (n =13). Our analysis indicated that serum cfDNA methylome from patients with PT are distinct from the counterparts in patients with other tumors (gliomas, meningiomas, colorectal carcinomas, n =134) and nontumor conditions (n = 4). Furthermore, the serum methylome patterns across PT was associated with functional status and adenohypophyseal cell lineage PT subtypes, recapitulating epigenetic features reported in PT-tissue. A machine learning algorithm using serum PT-specific signatures generated a score that distinguished PT from non-PT conditions with 100% accuracy in our validation set. These preliminary results underpin the potential clinical application of a liquid biopsy-based DNA methylation profiling as a noninvasive approach to identify clinically relevant epigenetic markers that can be used in the management of PT.


Gut ◽  
2018 ◽  
Vol 68 (3) ◽  
pp. 389-399 ◽  
Author(s):  
Ming Yu ◽  
Sean K Maden ◽  
Matthew Stachler ◽  
Andrew M Kaz ◽  
Jessica Ayers ◽  
...  

ObjectiveTo identify and characterise DNA methylation subtypes in oesophageal adenocarcinoma (EAC) and its precursor Barrett’s oesophagus (BE).DesignWe performed genome-wide DNA methylation profiling on samples of non-dysplastic BE from cancer-free patients (n=59), EAC (n=23), normal squamous oesophagus (n=33) and normal fundus (n=9), and identified methylation subtypes using a recursively partitioned mixture model. We assessed genomic alterations for 9 BE and 22 EAC samples with massively parallel sequencing of 243 EAC-associated genes, and we conducted integrative analyses with transcriptome data to identify epigenetically repressed genes. We also carried out in vitro experiments treating EAC cell lines with 5-Aza-2'-Deoxycytidine (5-Aza-dC), short hairpin RNA knockdown and anticancer therapies.ResultsWe identified and validated four methylation subtypes of EAC and BE. The high methylator subtype (HM) of EAC had the greatest number of activating events in ERBB2 (p<0.05, Student’s t-test) and the highest global mutation load (p<0.05, Fisher’s exact test). PTPN13 was silenced by aberrant methylation in the HM subtype preferentially and in 57% of EACs overall. In EAC cell lines, 5-Aza-dC treatment restored PTPN13 expression and significantly decreased its promoter methylation in HM cell lines (p<0.05, Welch’s t-test). Inhibition of PTPN13 expression in the SK-GT-4 EAC cell line promoted proliferation, colony formation and migration, and increased phosphorylation in ERBB2/EGFR/Src kinase pathways. Finally, EAC cell lines showed subtype-specific responses to topotecan, SN-38 and palbociclib treatment.ConclusionsWe identified and characterised methylator subtypes in BE and EAC. We further demonstrated the biological and clinical relevance of EAC methylator subtypes, which may ultimately help guide clinical management of patients with EAC.


Sign in / Sign up

Export Citation Format

Share Document