HDAC inhibitors (HDACI) are promising anti-tumor agents that have just entered the clinics. Despite their potent activity, the fundamental question of how HDAC inhibitors achieve their anti-tumor effects remains poorly understood. It is not known which of the HDAC family members or which cellular pathway is the most critical target for the antitumor activity of HDACI. This knowledge would facilitate the development of more effective inhibitors that target specific HDAC member(s) and identify the pathway(s) critical for HDACI to kill tumors, thereby aiding in the rational design of future cancer therapy. We have discovered that inactivation of a single HDAC member, HDAC10, recapitulates all major effects of HDACI, including growth arrest, cell death, induction of a cell cycle inhibitor, and reactive oxygen production (ROS). We further showed that HDAC10 inactivation and HDACI treatment both dramatically activate autophagy, a cellular response intimately linked to metabolic stress and cell death. These findings strongly suggest that HDAC10 is a key target mediating the anti-tumor activity of HDACI. Remarkably, we found that HDAC10 is localized to mitochondria. We hypothesize that HDACI elicits anti-tumor effects by targeting the mitochondrial deacetylase HDAC10, which controls mitochondrial function and autophagy important for tumor cell growth and proliferation. Specifically, we propose:

Public Health Relevance

HDAC inhibitors (HDACI) are promising anti-tumor agents. The fundamental question of how HDAC inhibitors achieve their anti-tumor effects remains poorly understood. We have identified a mitochondria-localized HDAC. Inactivation of this HDAC alone recapitulates all major phenotypes induced by HDACI. This finding indicates mitochondria function as a critical target for anti-tumor activity of HDACI. Elucidating the role of HDAC in mitochondria would provide new mechanistic understanding of how HDACI promotes cell death and uncover novel regulation of mitochondria function by HDAC and reversible acetylation.

Agency
National Institute of Health (NIH)
Institute
National Cancer Institute (NCI)
Type
Research Project (R01)
Project #
5R01CA133220-02
Application #
7895482
Study Section
Basic Mechanisms of Cancer Therapeutics Study Section (BMCT)
Program Officer
Spalholz, Barbara A
Project Start
2009-08-01
Project End
2011-07-31
Budget Start
2010-08-01
Budget End
2011-07-31
Support Year
2
Fiscal Year
2010
Total Cost
$323,700
Indirect Cost
Name
Duke University
Department
Pharmacology
Type
Schools of Medicine
DUNS #
044387793
City
Durham
State
NC
Country
United States
Zip Code
27705