Elsevier

Leukemia Research

Volume 31, Issue 5, May 2007, Pages 611-621
Leukemia Research

Analysis of histone modification around the CpG island region of the p15 gene in acute myeloblastic leukemia

https://doi.org/10.1016/j.leukres.2006.09.023Get rights and content

Abstract

Seven of 11 patients with acute myeloblastic leukemia (AML) had allele(s) in which more than half of 27 CpG sites in the p15 gene were methylated. The p15 CpG island region was surrounded with both the acetylated histone H3 (AcH3) and dimethylated histone H3-lysine 9 (MeH3K9) in bone marrow cells of AML patients, whereas with AcH3 alone in normal marrow cells. The p15 CpG islands of DNA immunoprecipitated with anti-AcH3 antibody and anti-MeH3K9 antibody were not always unmethylated and methylated, respectively, in the patients. These results suggest perturbed modifications of histone H3 around the p15 CpG island region in AML.

Introduction

DNA methylation and histone modifications are the major epigenetic mechanisms for gene expression. Hypermethylation of DNA in the promoter CpG islands of tumor suppressor genes that causes transcriptional silencing is thought to play an important role in carcinogenesis [1], [2], [3]. Histone proteins assemble into nucleosomes, which function as both DNA packaging units and transcriptional regulators. The amino-terminal tails of histones protrude from the nucleosome and are subject to chemical modifications including acetylation, methylation and phosphorylation [4]. Acetylated histones are associated with transcriptionally active regions of euchromatin, whereas hypoacetylated histones are associated with transcriptionally inert regions of heterochromatin [5], [6], [7], [8]. Histone H3 methylated at lysine 9 is enriched in deacetylated, transcriptionally silent heterochromatic regions, whereas histone H3 acetylated at lysine 9 is enriched in euchromatic domains and correlates with active gene expressions [9], [10], [11]. Recent reports have identified a mechanistic pathway of epigenetic silencing by cytosine methylation, histone hypoacetylation and chromatin condensation suggesting that these mechanisms act together to inactivate gene transcription [5], [12].

The p15 (also known as INK4b or MTS2) gene is a candidate tumor suppressor gene with structural and functional similarity to the p16 gene. Both p15 and p16 specifically inhibit cyclin-dependent kinase (CDK)4 and CDK6 [13], [14], which are key regulators of the progression of eukaryotic cells through the G1 phase of the cell cycle. Cameron et al. [15] reported that the degree of methylation varies in primary acute leukemia, whereas the entire CpG island region of p15 in normal lymphocytes is largely devoid of methylation. We have shown a dynamic change in methylation at the p15 CpG island and the inverse expression of p15 during normal myeloid development under stimulation with granulocyte-macrophage colony-stimulating factor alone (GM-CSF) or in combination with stem cell factor (SCF) [16]. More recently, we reported that GM-CSF can induce de novo methylation of the p15 gene, using histone deacetylase as well as DNA methyltransferase [17].

However, histone modifications regulating expression of the genes which encode CDK inhibitors in normal peripheral blood (PB)/bone marrow (BM) cells and acute myeloblastic leukemia (AML) remain unclear. We attempted, for the first time, to elucidate histone modifications around the CpG island region of the p15 gene in AML cells, using the chromatin immunoprecipitation (ChIP) assay and methylation-specific PCR with bisulfite genomic sequencing.

Section snippets

Cell preparation

BM cells were aspirated in heparinized plastic syringes from 11 children with AML after obtaining the informed consent of each patient and/or the parents. Mononuclear cells (MNCs) were separated by density centrifugation over Ficoll-Paque (Amersham Biosciences, Uppsala, Sweden), and were frozen with liquid nitrogen until the experiment. Fifty-two to 97% of the isolated cells were leukemic blasts. The patients were classified into FAB subtypes on the basis of morphologic and cytochemical

Histone modifications around the CpG island of the p15 gene in leukemic cell lines

To elucidate DNA methylation and histone modifications around the CpG island of the p15 gene in leukemic cells, we performed bisulfite genomic sequencing and ChIP assays on leukemic cell lines (HL60 cells and Molt4 cells). The results are presented in Fig. 1. The CpG island region of p15 is completely unmethylated in HL60 cells, whereas it is almost fully methylated in Molt4 cells, being consistent with the previous results [16], [20]. Nucleoprotein complexes were sonicated to reduce the size

Discussion

Frequent p15 hypermethylation was demonstrated in children and adults with AML, whereas p16 methylation has rarely been observed [21]. When at least half of 27 CpG sites in the p15 gene were methylated in an individual allele, the allele was defined as methylated. Given this criterion, 7 (63.6%) of 11 patients with AML had allele(s) with methylated p15 CpG islands. These results are consistent with those reported previously [21], [22], [23]. There have been controversies regarding a

Acknowledgments

This work was supported by Grants-in-Aid Nos. 15790521, 11670753, and 09041178 from the Ministry of Education of Japan.

References (25)

  • S.J. Nielsen et al.

    Rb targets histone H3 methylation and HP1 to promoters

    Nature

    (2001)
  • K. Noma et al.

    Transitions in distinct histone H3 methylation patterns at the heterochromatin domain boundaries

    Science

    (2001)
  • Cited by (11)

    • Effect of BIX-01294 on proliferation, apoptosis and histone methylation of acute T lymphoblastic leukemia cells

      2017, Leukemia Research
      Citation Excerpt :

      It has been shown that H3K9 methylation plays an important role in heterochromatin formation, gene imprinting, X chromosome inactivation and transcriptional regulation [3]. Recent studies have found that imbalance of H3K9 methylation by aberrant expression of G9a importantly contributes to the development of tumor [4]. BIX-01294 is a synthetic G9a-selective inhibitor.

    • Signatures of polycomb repression and reduced H3K4 trimethylation are associated with p15INK4b DNA methylation in AML

      2010, Blood
      Citation Excerpt :

      The lack of H3K27me3 and EZH2 binding at the INK4b-ARF-INK4a region in AML-193 cells does not result from global changes in H3K27me3 or EZH2 protein levels compared with other AML cell lines (supplemental Figure 4). Although there was a previous report demonstrating an association of H3K9me3 with p15INK4b DNA methylation in AML cells,27 we found low levels of H3K9me3 at p15INK4b irrespective of the p15INK4b DNA methylation status (Figure 2D). In addition, our data indicated that low levels of H3K9me3 also exist at p14ARF-p16INK4a in AML cell lines, with the exception of KG-1 and KG-1a cells.

    View all citing articles on Scopus
    View full text