Corepressor-dependent silencing of fetal hemoglobin expression by BCL11A.

Document Type

Article

Publication Date

4-16-2013

Keywords

Animals, Carrier Proteins, Cell Line, Tumor, Chromatin, Chromatin Immunoprecipitation, Chromatography, Liquid, Co-Repressor Proteins, Erythroid Precursor Cells, Fetal Hemoglobin, Gene Expression Regulation, Developmental, Humans, Mice, Multiprotein Complexes, Nuclear Proteins, Proteomics, RNA Interference, Real-Time Polymerase Chain Reaction, Tandem Mass Spectrometry, beta-Globins

JAX Source

Proc Natl Acad Sci U S A 2013 Apr 16; 110(16):6518-23.

Volume

110

Issue

16

First Page

6518

Last Page

6523

ISSN

1091-6490

PMID

23576758

Abstract

Reactivation of fetal hemoglobin (HbF) in adults ameliorates the severity of the common β-globin disorders. The transcription factor BCL11A is a critical modulator of hemoglobin switching and HbF silencing, yet the molecular mechanism through which BCL11A coordinates the developmental switch is incompletely understood. Particularly, the identities of BCL11A cooperating protein complexes and their roles in HbF expression and erythroid development remain largely unknown. Here we determine the interacting partner proteins of BCL11A in erythroid cells by a proteomic screen. BCL11A is found within multiprotein complexes consisting of erythroid transcription factors, transcriptional corepressors, and chromatin-modifying enzymes. We show that the lysine-specific demethylase 1 and repressor element-1 silencing transcription factor corepressor 1 (LSD1/CoREST) histone demethylase complex interacts with BCL11A and is required for full developmental silencing of mouse embryonic β-like globin genes and human γ-globin genes in adult erythroid cells in vivo. In addition, LSD1 is essential for normal erythroid development. Furthermore, the DNA methyltransferase 1 (DNMT1) is identified as a BCL11A-associated protein in the proteomic screen. DNMT1 is required to maintain HbF silencing in primary human adult erythroid cells. DNMT1 haploinsufficiency combined with BCL11A deficiency further enhances γ-globin expression in adult animals. Our findings provide important insights into the mechanistic roles of BCL11A in HbF silencing and clues for therapeutic targeting of BCL11A in β-hemoglobinopathies. Proc Natl Acad Sci U S A 2013 Apr 16; 110(16):6518-23.

Share

COinS