1. Academic Validation
  2. A novel histone deacetylase pathway regulates mitosis by modulating Aurora B kinase activity

A novel histone deacetylase pathway regulates mitosis by modulating Aurora B kinase activity

  • Genes Dev. 2006 Sep 15;20(18):2566-79. doi: 10.1101/gad.1455006.
Yun Li 1 Gary D Kao Benjamin A Garcia Jeffrey Shabanowitz Donald F Hunt Jun Qin Caroline Phelan Mitchell A Lazar
Affiliations

Affiliation

  • 1 Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania School of Medicine, Philadelphia, 19104, USA.
Abstract

Histone deacetylase (HDAC) inhibitors perturb the cell cycle and have great potential as anti-cancer agents, but their mechanism of action is not well established. HDACs classically function as repressors of gene expression, tethered to sequence-specific transcription factors. Here we report that HDAC3 is a critical, transcription-independent regulator of mitosis. HDAC3 forms a complex with A-Kinase-Anchoring Proteins AKAP95 and HA95, which are targeted to mitotic chromosomes. Deacetylation of H3 in mitosis requires AKAP95/HA95 and HDAC3 and provides a hypoacetylated H3 tail that is the preferred substrate for Aurora B kinase. Phosphorylation of H3S10 by Aurora B leads to dissociation of HP1 proteins from methylated H3K9 residues on mitotic heterochromatin. This transcription-independent pathway, involving interdependent changes in histone modification and protein association, is required for normal progression through mitosis and is an unexpected target of HDAC inhibitors, a class of drugs currently in clinical trials for treating Cancer.

Figures