Oncotarget

Research Papers:

Gene-body hypermethylation controlled cryptic promoter and miR26A1-dependent EZH2 regulation of TET1 gene activity in chronic lymphocytic leukemia

Pradeep Kumar Kopparapu, Mohammad Hamdy Abdelrazak Morsy, Chandrasekhar Kanduri and Meena Kanduri _

PDF  |  HTML  |  Supplementary Files  |  How to cite

Oncotarget. 2017; 8:77595-77608. https://doi.org/10.18632/oncotarget.20668

Metrics: PDF 1854 views  |   HTML 2981 views  |   ?  


Abstract

Pradeep Kumar Kopparapu1,*, Mohammad Hamdy Abdelrazak Morsy1,*, Chandrasekhar Kanduri2 and Meena Kanduri1

1Department of Clinical Chemistry and Transfusion Medicine, Institute of Biomedicine, Sahlgrenska Academy, Gothenburg University, Gothenburg, Sweden

2Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, Sahlgrenska Academy, Gothenburg University, Gothenburg, Sweden

*These authors contributed equally to this work

Correspondence to:

Meena Kanduri, email: [email protected]

Keywords: gene-body hypermethylation, chronic lymphocytic leukemia, cryptic promoters, EZH2 and TET1 gene

Received: April 28, 2017     Accepted: June 29, 2017     Published: September 06, 2017

ABSTRACT

The Ten-eleven-translocation 1 (TET1) protein is a member of dioxygenase protein family that catalyzes the oxidation of 5-methylcytosine to 5-hydroxymethylcytosine. TET1 is differentially expressed in many cancers, including leukemia. However, very little is known about mechanism behind TET1 deregulation. Previously, by characterizing global methylation patterns in CLL patients using MBD-seq, we found TET1 as one of the differentially methylated regions with gene-body hypermethylation. Herein, we characterize mechanisms that control TET1 gene activity at the transcriptional level. We show that treatment of CLL cell lines with 5-aza 2´-deoxycytidine (DAC) results in the activation of miR26A1, which causes decrease in both mRNA and protein levels of EZH2, which in turn results in the decreased occupancy of EZH2 over the TET1 promoter and consequently the loss of TET1 expression. In addition, DAC treatment also leads to the activation of antisense transcription overlapping the TET1 gene from a cryptic promoter, located in the hypermethylated intronic region. Increased expression of intronic transcripts correlates with decreased TET1 promoter activity through the loss of RNA Pol II occupancy. Thus, our data demonstrate that TET1 gene activation in CLL depends on miR26A1 regulated EZH2 binding at the TET1 promoter and silencing of novel cryptic promoter by gene-body hypermethylation.


Creative Commons License All site content, except where otherwise noted, is licensed under a Creative Commons Attribution 4.0 License.
PII: 20668