scholarly journals Aberrant DNA methylation and epigenetic inactivation of Eph receptor tyrosine kinases and ephrin ligands in acute lymphoblastic leukemia

Blood ◽  
2010 ◽  
Vol 115 (12) ◽  
pp. 2412-2419 ◽  
Author(s):  
Shao-Qing Kuang ◽  
Hao Bai ◽  
Zhi-Hong Fang ◽  
Gonzalo Lopez ◽  
Hui Yang ◽  
...  

Eph receptors and their ephrin ligands are involved in normal hematopoietic development and tumorigenesis. Using methylated CpG island amplification/DNA promoter microarray, we identified several EPH receptor and EPHRIN genes as potential hypermethylation targets in acute lymphoblastic leukemia (ALL). We subsequently studied the DNA methylation status of the Eph/ephrin family by bisulfite pyrosequencing. Hypermethylation of EPHA2, -A4, -A5, -A6, -A7, -A10, EPHB1, -B2, -B3, -B4, EFNA1, -A3, -A5, and EFNB1 and -B2 genes was detected in leukemia cell lines and primary ALL bone marrow samples. Expression analysis of EPHB4, EFNB2, and EFNA5 genes demonstrated that DNA methylation was associated with gene silencing. We cloned the promoter region of EPHB4 and demonstrated that promoter hypermethylation can result in EPHB4 transcriptional silencing. Restoration of EPHB4 expression by lentiviral transduction resulted in reduced proliferation and apoptotic cell death in Raji cells in which EPHB4 is methylated and silenced. Finally, we demonstrated that phosphorylated Akt is down-regulated in Raji cells transduced with EPHB4. These results suggest that epigenetic silencing by hypermethylation of EPH/EPHRIN family genes contributes to ALL pathogenesis and that EPHB4 can function as a tumor suppressor in ALL.

Blood ◽  
2009 ◽  
Vol 114 (27) ◽  
pp. 5490-5498 ◽  
Author(s):  
Dominique J. P. M. Stumpel ◽  
Pauline Schneider ◽  
Eddy H. J. van Roon ◽  
Judith M. Boer ◽  
Paola de Lorenzo ◽  
...  

Abstract MLL-rearranged infant acute lymphoblastic leukemia (ALL) remains the most aggressive type of childhood leukemia, displaying a unique gene expression profile. Here we hypothesized that this characteristic gene expression signature may have been established by potentially reversible epigenetic modifications. To test this hypothesis, we used differential methylation hybridization to explore the DNA methylation patterns underlying MLL-rearranged ALL in infants. The obtained results were correlated with gene expression data to confirm gene silencing as a result of promoter hypermethylation. Distinct promoter CpG island methylation patterns separated different genetic subtypes of MLL-rearranged ALL in infants. MLL translocations t(4;11) and t(11;19) characterized extensively hypermethylated leukemias, whereas t(9;11)-positive infant ALL and infant ALL carrying wild-type MLL genes epigenetically resembled normal bone marrow. Furthermore, the degree of promoter hypermethylation among infant ALL patients carrying t(4;11) or t(11;19) appeared to influence relapse-free survival, with patients displaying accentuated methylation being at high relapse risk. Finally, we show that the demethylating agent zebularine reverses aberrant DNA methylation and effectively induces apoptosis in MLL-rearranged ALL cells. Collectively these data suggest that aberrant DNA methylation occurs in the majority of MLL-rearranged infant ALL cases and guides clinical outcome. Therefore, inhibition of aberrant DNA methylation may be an important novel therapeutic strategy for MLL-rearranged ALL in infants.


Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 847-847
Author(s):  
Tadayuki Akagi ◽  
Dong Yin ◽  
Norihiko Kawamata ◽  
Claus R. Bartram ◽  
Carl W. Miller ◽  
...  

Abstract Asparagine synthetase (ASY) is an enzyme expressed ubiquitously in mammalian cells. Expression of the gene, however, is repressed in acute lymphoblastic leukemia (ALL), and the cells are unable to synthesize sufficient asparagine. Depletion of asparagine by L-asparaginase leads to cell death in ALL; therefore, asparaginase has been used as treatment of ALL. Here, we show the methylation status of ASY gene in ALL clinical samples, and report a novel insertional polymorphism in the gene. The ASY gene has a CpG island located from −313 to +336 including 49 CpG sites. We analyzed the methylation status in the region by sodium bisulfite conversion of genomic DNA followed by PCR analysis. A total of 18 of 22 (82%) bone marrow samples of ALL at diagnosis were methylated in the CpG island. After chemotherapic induced-remission, all the bone marrow samples were unmethylated including the 18 patients whose ALL samples were methylated. Experiments by others using an ALL cell line showed that methylation of the ASY gene silenced the gene. In addition, we discovered either a 14 bp or 28 bp inserted sequence between +83 and +84 located in the 1st intron of the gene. Methylation status of the gene was independent of the insertion. To calculate the frequency of the insertion, 92 ALL samples were analyzed by PCR and sequencing. 69 samples (75%) did not contain the insertion in either allele, 22 samples (24%) contained the insertion in one allele, and only 1 sample (1%) contained the insertion in both alleles. To examine whether the insertion was specific for leukemic cells, we analyzed 71 non-ALL samples. 48 samples (68%) did not contain the insertion in either allele, 12 samples (27%) contained the insertion in one allele, and 4 samples (6%) contained the insertion in both allele. These results showed that the insertion is a germline polymorphism; we are exploring if this insertion changes expression of the gene. In summary, we found a novel insertional polymorphism in the ASY gene. Also, our data for the first time suggests that ALL cells cannot express ASY because the gene’s CpG island is methylated, thus explaining why ALL cells are sensitive to L-asparaginase. Notably, 18% of ALL samples were not methylated, and these patients might be expected to be resistant to L-asparaginase therapy suggesting that a simple test could be of therapeutic value.


Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 2242-2242
Author(s):  
Xiaoqing Li ◽  
Jun Liu ◽  
Rui Zhou ◽  
Yanli He ◽  
Shi Huang ◽  
...  

Abstract The regulation of human microRNA (miRNA) expression is still poorly understood and aberrant epigenetic regulation has recently been implicated in the down-regulation of tumor suppressor miRNAs. In this study, we investigated whether histone modifications would contribute to the dysregulation of miRNAs in lymphoblastic leukemia cells. Using a precursor B-cell acute lymphoblastic leukemia cell line, NALM-6 cells, we demonstrated by miRNA microarray analysis that a specific histone deacetylases inhibitor, trichostatin A (TSA), induced a differential alteration in cellular miRNA expression. A total of 10 miRNAs were down-regulated and 31 up-regulated significantly following TSA treatment. Among TSA-up-regulated miRNAs, miR-22 is an extronic miRNA and resides in the second exon of the non-coding transcript MGC14376. Up-regulation of both miR-22 and MGC14376 was found in NALM-6 cells treated with TSA but not 5-AZA-2’-deoxycytidine, a DNA demethylating agent. Luciferase reporter analysis identified three regions in the promoter of miR-22 and MGC14376 that differentially regulated its transcriptional activation. Although there is a CpG island within the promoter of miR-22 and MGC14376, no obvious methylation was detected at this region in NALM-6 cells. Conversely, H3K27 trimethylation (H3K27triM)-associated histone modification was identified in the first intron of MGC14376 gene and was involved in TSA-induced miR-22 expression. Thus, miR-22 silencing in NALM-6 cells involves H3K27triM-associated histone modification but is independent of DNA methylation, suggesting that methylation-independent H3K27triM histone modification may be an important mechanism for miRNA dysregulation in cancer cells.


Blood ◽  
2002 ◽  
Vol 99 (7) ◽  
pp. 2291-2296 ◽  
Author(s):  
Jose Roman-Gomez ◽  
Juan Antonio Castillejo ◽  
Antonio Jimenez ◽  
Maria Gracia Gonzalez ◽  
Fernanda Moreno ◽  
...  

The p21 is a downstream effector of p53/p73 and belongs to the CIP/KIP family of cyclin-dependent kinase inhibitors (CDKIs). It is, therefore, a potential tumor suppressor gene and probably plays an important role in tumor development. Moreover, reduced expression of p21 has been reported to have prognostic value in several human malignancies. In contrast with other CDKIs, mutational inactivation of p21 is infrequent, but gene inactivation by an alternative mechanism seems to be the general pathway. In this study, we analyzed the methylation status of the p21 promoter region using semiquantitative polymerase chain reaction in 124 patients with acute lymphoblastic leukemia (ALL). We observed p21 hypermethylation in bone marrow cells from 41% (51 of 124) of ALL patients. Hypermethylation within promoter strongly correlated with decreased p21 messenger RNA expression in tumoral cells. Clinical, molecular, and laboratory features and complete remission rate did not differ significantly between hypermethylated and normally methylated patients. Estimated disease-free survival (DFS) and overall survival at 7 and 9 years, respectively, were 59% and 65% for healthy patients and 6% and 8% for hypermethylated patients (P = .00001 andP = .006). Multivariate analysis of potential prognostic factors demonstrated that p21 methylation status was an independent prognostic factor in predicting DFS (P = .0001). Our results indicate that the p21 gene is subject to methylation regulation at the transcription level in ALL and seems to be an important factor in predicting the clinical outcome of these patients.


Blood ◽  
2020 ◽  
Vol 136 (Supplement 1) ◽  
pp. 28-29
Author(s):  
Juliette Roels ◽  
Morgan Thénoz ◽  
Bronisława Szarzyńska ◽  
Mattias Landfors ◽  
Stien De Coninck ◽  
...  

During the last decade, aberrant DNA methylation has been identified as a hallmark of human cancer and several studies have highlighted the potential of DNA methylation as a clinically or diagnostically relevant biomarker. In comparison to their normal healthy counterparts, cancer cells generally display DNA hypermethylation at specific CpG islands, but the actual mechanism that drives this so-called CpG island methylator phenotype (CIMP) remains poorly understood. To profile the DNA methylation landscape of human T-cell acute lymphoblastic leukemia (T-ALL), we analyzed 109 T-ALLs together with 10 stages of normal T cell development, which are considered the normal human counterparts of this disease, by 850 EPIC arrays. Here, we show that CpG islands are hypermethylated in all human T- ALLs compared to their normal counterparts. We designed a DNA methylation signature that can distinguish two types of T-ALL, with low or high levels of CpG island hypermethylation. This profile is dominated by CpGs in promoters of PRC2 target genes. T-ALLs with high levels of CpG island hypermethylation show low levels of H3K27me3 and vice versa, resulting in gene repression in both subtypes of T-ALL by different mechanisms. Furthermore, we found that aberrant CpG island hypermethylation shows a strong correlation with the epigenetic age of the leukemic T cells. By investigating the DNA methylation profile of two distinct mouse T-ALL models, the Lck-Cre Ptenfl/fland the CD2-Lmo2 transgenic mouse model, by Reduced Representation Bisulfite Sequencing, we could indeed recapitulate the DNA methylation features of the two human T-ALL subtypes in mice (Fig.1). The aggressive, fast-transforming Ptenfl/flmouse model displays low levels of CpG island hypermethylation, which correlated with human T-ALLs that have a shorter proliferative history and a worse prognosis. In contrast, murine CD2-Lmo2 T-ALLs have a longer disease latency and display a CpG island hypermethylation phenotype that is similar to human T-ALLs with a longer proliferative history. In CD2-Lmo2 mice, a pre-leukemic phase is present with self-renewing thymocytes. We elucidate that the CpG island methylation signature is gradually established in aging pre-leukemic thymocytes of 8, 16 and 24 weeks old CD2-Lmo2 mice (Fig.1). Of note, this hypermethylation phenotype is completely absent in age matched Lck-Cre Ptenfl/flmice that did not yet develop leukemia, suggesting that the proliferative history is responsible for aberrant CpG island DNA methylation observed in human T-ALL. Notably, this provides the first evidence that a pre-leukemic phase might be present in a large subset of human T-ALLs, and that epigenetic aberrations, either in the DNA methylation or histone methylation machinery are one of the first detectable alterations during T-ALL development. Finally, using patient derived xenografts (PDX), we show that DNA hypomethylation by the FDA-approved hypomethylating agent Decitabine is very effective in treating T-ALL. Gene expression profiling revealed that the anti-leukemic effect is exerted by down-regulation of the oncogenic MYC pathway. However, by profiling these PDX T-ALLs by EPIC arrays, we unexpectedly uncover that the age-related CpG island hypermethylation signature is completely resistant to Decitabine treatment. Altogether, our work demonstrates that DNA methylation reflects the epigenetic history of leukemic T cells and suggests that methylation-based subtypes of human T-ALL have followed a different trajectory towards T-cell transformation, possibly mediated by differences in the self-renewing capacity of the putative T-ALL cell-of-origin. Given that the concept of preleukemic thymocytes has only been reported in T-ALL mouse models so far, we here provide, for the first time, conceptual evidence that a pre-leukemic phase might also be involved in the pathogenesis of the human disease. Disclosures No relevant conflicts of interest to declare.


2020 ◽  
Vol 20 ◽  
Author(s):  
Ezzatollah Fathi ◽  
Raheleh Farahzadi ◽  
Soheila Montazersaheb ◽  
Yasin Bagheri

Background:: Epigenetic modification pattern is considered as a characteristic feature in blood malignancies. Modifications in the DNA methylation modulators are recurrent in lymphoma and leukemia, so that, the distinct methylation pattern defines different types of leukemia. Generally, the role of epigenetics is less understood and most investigations are focused on genetic abnormalities and cytogenic studies to develop novel treatments for patients with hematologic disorders. Recently, understanding the underlying mechanism of acute lymphoblastic leukemia (ALL), especially epigenetic altera-tions as a driving force in the development of ALL opens a new era of investigation for developing promising strategy, be-yond available conventional therapy. Objective:: This review will focus on a better understanding of the epigenetic mechanisms in cancer development and pro-gression, with an emphasis on epigenetic alterations in ALL including, DNA methylation, histone modification, and mi-croRNA alterations. Other topics that will be discussed include the use of epigenetic alterations as a promising therapeutic target in order to develop novel well-suited approaches against ALL. Conclusion:: According to the literature review, leukemogenesis of ALL is extensively influenced by epigenetic modifica-tions, particularly DNA hyper-methylation, histone modification, and miRNA alteration.


2021 ◽  
Vol 22 (3) ◽  
pp. 1388
Author(s):  
Natalia Maćkowska ◽  
Monika Drobna-Śledzińska ◽  
Michał Witt ◽  
Małgorzata Dawidowska

Distinct DNA methylation signatures, related to different prognosis, have been observed across many cancers, including T-cell acute lymphoblastic leukemia (T-ALL), an aggressive hematological neoplasm. By global methylation analysis, two major phenotypes might be observed in T-ALL: hypermethylation related to better outcome and hypomethylation, which is a candidate marker of poor prognosis. Moreover, DNA methylation holds more than a clinical meaning. It reflects the replicative history of leukemic cells and most likely different mechanisms underlying leukemia development in these T-ALL subtypes. The elucidation of the mechanisms and aberrations specific to (epi-)genomic subtypes might pave the way towards predictive diagnostics and precision medicine in T-ALL. We present the current state of knowledge on the role of DNA methylation in T-ALL. We describe the involvement of DNA methylation in normal hematopoiesis and T-cell development, focusing on epigenetic aberrations contributing to this leukemia. We further review the research investigating distinct methylation phenotypes in T-ALL, related to different outcomes, pointing to the most recent research aimed to unravel the biological mechanisms behind differential methylation. We highlight how technological advancements facilitated broadening the perspective of the investigation into DNA methylation and how this has changed our understanding of the roles of this epigenetic modification in T-ALL.


Sign in / Sign up

Export Citation Format

Share Document