Heart failure is a major cause of disease and death in the US and worldwide. Features of this disease are heart enlargement (hypertrophy) and heart stiffness (fibrosis), which result in impaired heart contraction and relaxation. The five-year deat rate following first admission of heart failure is >40%, stressing the need for new therapeutic strategies. Histone deacetylase (HDAC) inhibitors have emerged as a new class of drugs that have been shown as effective in reducing cardiac hypertrophy and ultimately improving heart failure in pre- clinical models. We previously published that HDAC inhibition blocks nuclear ERK1/2 signaling, and thus hypertrophy, by upregulating the nuclear ERK1/2 phosphatase, dual-specificity phosphatase 5 (DUSP5) in cardiac myocytes. However, nothing is known about the in vivo function of DUSP5 in the heart. Recent studies demonstrate that nuclear ERK1/2 signaling is a central regulator of pathological cardiac hypertrophy in animal models of HF. In preliminary studies, we postulated that DUSP5 null mice would develop exaggerated cardiac hypertrophy in response to stress due to enhanced nuclear ERK phosphorylation. Indeed, DUSP5-deficient mice had significantly larger left ventricles (LVs), as well as enhanced ERK1/2 activation, compared to wild- type littermates in response to treatment with the -adrenergic agonist, isoproterenol (ISO). Curiously, ISO did not induce RV hypertrophy in wild-type mice, but triggered a ~40% increase in RV mass in DUSP5 null mice. These data suggest a prominent role for DUSP5 in the control of RV hypertrophy. Significantly, relative to LV hypertrophy, little is known about the molecular mechanisms controlling pathological RV growth. This application will fill a critical void by testing the hypothesis that DUSP5 functions as a signl-dependent repressor of cardiac hypertrophy by dephosphorylating nuclear ERK1/2, and this mechanism is especially critical for suppression of RV hypertrophy. To test our hypothesis, we have developed two specific aims.
Aim 1 will elucidate the mechanism(s) by which HDACs control DUSP5 expression in cardiac myocytes using chromatin immunoprecipitation (ChIP) and DNA methylation techniques to determine epigenetic regulatory events within the dusp5 promoter.
In Aim 2, we will address the in vivo role ofDUSP5 in the control of nuclear ERK1/2 phosphorylation and pathological RV hypertrophy in animal models of heart failure. The proposed study will significantly add to our limited knowledge regarding RV growth, potentially facilitating development of better therapies for patients with right-sided heart failure.

Public Health Relevance

Heart failure (HF) is a major cause of disease and death worldwide. Prolonged heart enlargement (hypertrophy) plays a critical role in the development of heart failure, yet little is known about heart hypertrophy and in particular right ventricular (RV) hypertrophy. This project will investigate the role of dual-specificity phosphatase 5 (DUSP5) as a repressor of heart hypertrophy, with particular emphasis on the function of this phosphatase in the control of RV enlargement. Knowledge gained from this work has the potential to facilitate development of better therapies for patients with right-sided heart failure.

Agency
National Institute of Health (NIH)
Institute
National Heart, Lung, and Blood Institute (NHLBI)
Type
Postdoctoral Individual National Research Service Award (F32)
Project #
5F32HL124893-02
Application #
8979448
Study Section
Special Emphasis Panel (ZRG1)
Program Officer
Wang, Wayne C
Project Start
2014-09-01
Project End
2015-12-31
Budget Start
2015-09-01
Budget End
2015-12-31
Support Year
2
Fiscal Year
2015
Total Cost
Indirect Cost
Name
University of Colorado Denver
Department
Internal Medicine/Medicine
Type
Schools of Medicine
DUNS #
041096314
City
Aurora
State
CO
Country
United States
Zip Code
80045
Angiolilli, Chiara; Kabala, Pawel A; Grabiec, Aleksander M et al. (2017) Histone deacetylase 3 regulates the inflammatory gene expression programme of rheumatoid arthritis fibroblast-like synoviocytes. Ann Rheum Dis 76:277-285
Nozik-Grayck, Eva; Woods, Crystal; Stearman, Robert S et al. (2016) Histone deacetylation contributes to low extracellular superoxide dismutase expression in human idiopathic pulmonary arterial hypertension. Am J Physiol Lung Cell Mol Physiol 311:L124-34
Angiolilli, Chiara; Grabiec, Aleksander M; Ferguson, Bradley S et al. (2016) Inflammatory cytokines epigenetically regulate rheumatoid arthritis fibroblast-like synoviocyte activation by suppressing HDAC5 expression. Ann Rheum Dis 75:430-8
Lemon, Douglas D; Harrison, Brooke C; Horn, Todd R et al. (2015) Promiscuous actions of small molecule inhibitors of the protein kinase D-class IIa HDAC axis in striated muscle. FEBS Lett 589:1080-8
Ferguson, Bradley S; McKinsey, Timothy A (2015) Non-sirtuin histone deacetylases in the control of cardiac aging. J Mol Cell Cardiol 83:14-20
McLendon, Patrick M; Ferguson, Bradley S; Osinska, Hanna et al. (2014) Tubulin hyperacetylation is adaptive in cardiac proteotoxicity by promoting autophagy. Proc Natl Acad Sci U S A 111:E5178-86