KDM3B is the H3K9 demethylase involved in transcriptional activation of lmo2 in leukemia

Riferimento: 
Mol Cell Biol. 2012 Jul;32(14):2917-33.
Autori: 
Kim JY, Kim KB, Eom GH, Choe N, Kee HJ, Son HJ, Oh ST, Kim DW, Pak JH, Baek HJ, Kook H, Hahn Y, Kook H, Chakravarti D, Seo SB.
Fonte: 
Mol Cell Biol. 2012 Jul;32(14):2917-33.
Anno: 
2012
Azione: 
Abbiamo scoperto che il genoma KDM3B reprime la differenziazione indotta dall'acido all-trans retinoico (ATRA) nelle cellule di leucemia promielocitica.
Target: 
ATRA- KDM3B/leucemia promielocitica.

ABSTRACT
Histone lysine methylation and demethylation are considered critical steps in transcriptional regulation. In this report, we performed chromatin immunoprecipitation with microarray technology (ChIP-chip) analysis to examine the genome-wide occupancy of H3K9-me2 during all-trans-retinoic acid (ATRA)-induced differentiation of HL-60 promyelocytic leukemia cells. Using this approach, we found that KDM3B, which contains a JmjC domain, was downregulated during differentiation through the recruitment of a corepressor complex. Furthermore, KDM3B displayed histone H3K9-me1/2 demethylase activity and induced leukemogenic oncogene lmo2 expression via a synergistic interaction with CBP. Here, we found that KDM3B repressed leukemia cell differentiation and was upregulated in blood cells from acute lymphoblastic leukemia (ALL)-type leukemia patients. The combined results of this study provide evidence that the H3K9-me1/2 demethylase KDM3B might play a role in leukemogenesis via activation of lmo2 through interdependent actions with the histone acetyltransferase (HAT) complex containing CBP.
Free PMC Article

Sostanze: