1. Academic Validation
  2. Histone lysine demethylase KDM5B facilitates proliferation and suppresses apoptosis in human acute myeloid leukemia cells through the miR-140-3p/BCL2 axis

Histone lysine demethylase KDM5B facilitates proliferation and suppresses apoptosis in human acute myeloid leukemia cells through the miR-140-3p/BCL2 axis

  • RNA. 2024 Jan 31:rna.079865.123. doi: 10.1261/rna.079865.123.
Jiaojuan Huang 1 Shuiling Jin 2 Rongqun Guo 3 Wei Wu 4 Chengxuan Yang 5 Yali Qin 1 Qingchuan Chen 1 Ximiao He 1 Jing Qu 1 Zhenhua Yang 6
Affiliations

Affiliations

  • 1 School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology.
  • 2 Department of Oncology, the First Affiliated Hospital of Zhengzhou University.
  • 3 Department of Hematology, the First Affiliated Hospital of Zhengzhou University.
  • 4 Department of Clinical Laboratory, institute of translational medicine, Renmin Hospital of Wuhan University.
  • 5 Department of Galactophore, Xinxiang First People's Hospital, Xinxiang.
  • 6 School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology; [email protected].
Abstract

The histone lysine demethylase KDM5B is frequently upregulated in various human Cancer cells. However, its expression and functional role in human acute myeloid leukemia (AML) cells remain unclear. Here, we found that the expression level of KDM5B is high in primary human AML cells. We have demonstrated that knocking down KDM5B leads to Apoptosis and impairs proliferation in primary human AML and some human AML cell lines. We further identified miR-140-3p as a downstream target gene of KDM5B. KDM5B expression was inversely correlated with miR-140-3p level in primary human AML cells. Molecular studies showed that silencing KDM5B enhanced H3K4 tri-methylation (H3K4me3) at the promoter of miR-140-3p, leading to high expression of miR-140-3p, which in turn inhibited B-cell CLL/lymphoma 2 (BCL2) expression. Finally, we demonstrated that the defective proliferation induced by KDM5B knockdown (KD) can be rescued with the miR-140-3p inhibitor or enhanced by combining KDM5B KD with a BCL2 inhibitor. Altogether, our data support the conclusion that KDM5B promotes tumorigenesis in human AML cells through the miR-140-3p/BCL2 axis. Targeting the KDM5B/miR-140-3p/BCL2 pathway may hold therapeutic promise for treating human AML.

Keywords

Acute myeloid leukemia; BCL2; Histone H3K4 methylation; KDM5B; miR-140-3p.

Figures
Products