4.7 Article

Impairment of human terminal erythroid differentiation by histone deacetylase 5 deficiency

Journal

BLOOD
Volume 138, Issue 17, Pages 1615-1627

Publisher

AMER SOC HEMATOLOGY
DOI: 10.1182/blood.2020007401

Keywords

-

Categories

Funding

  1. National Institutes of Health (NIH), National Heart, Lung, and Blood Institute [HL140625, HL149626]
  2. NIH, National Institute of Diabetes and Digestive and Kidney Diseases [DK032094]
  3. Hugoton Foundation
  4. Natural Science Foundation of China [U1804282, 81530005, 81570099, 81770112, 81700102, 81900108]

Ask authors/readers for more resources

The study highlights the significant role of HDAC5 in human erythropoiesis, as knockdown of HDAC5 leads to increased apoptosis, decreased chromatin condensation, and impaired enucleation of erythroblasts.
Histone deacetylases (HDACs) are a group of enzymes that catalyze the removal of acetyl groups from histone and nonhistone proteins. HDACs have been shown to have diverse func-tions in a wide range of biological processes. However, their roles in mammalian erythropoi-esis remain to be fully defined. This study showed that, of the 11 classic HDAC family members, 6 (HDAC1, -2, -3, and HDAC5, -6, -7) are expressed in human erythroid cells, with HDAC5 most significantly upregulated during terminal erythroid differentiation. Knock-down of HDAC5 by either short hairpin RNA or small interfering RNA in human CD34(+) cells followed by erythroid cell culture led to increased apoptosis, decreased chromatin conden-sation, and impaired enucleation of erythroblasts. Biochemical analyses revealed that HDAC5 deficiency resulted in activation of p53 in association with increased acetylation of p53. Furthermore, although acetylation of histone 4 (H4) is decreased during normal terminal erythroid differentiation, HDAC5 deficiency led to increased acetylation of H4 (K12) in late-stage erythroblasts. This increased acetylation was accompanied by decreased chromatin condensation, implying a role for H4 (K12) deacetylation in chromatin condensation. ATAC-seq and RNA sequencing analyses revealed that HDAC5 knockdown leads to increased chromatin accessibility genome-wide and global changes in gene expression. Moreover, pharmacological inhibition of HDAC5 by the inhibitor LMK235 also led to increased H4 acetylation, impaired chromatin condensation, and enucleation. Taken together, our findings have uncovered previously unrecognized roles and molecular mechanisms of action for HDAC5 in human erythropoiesis. These results may provide insights into understanding the anemia associated with HDAC inhibitor treatment.

Authors

I am an author on this paper
Click your name to claim this paper and add it to your profile.

Reviews

Primary Rating

4.7
Not enough ratings

Secondary Ratings

Novelty
-
Significance
-
Scientific rigor
-
Rate this paper

Recommended

No Data Available
No Data Available