Heart disease and resultant heart failure remain a leading cause of death and disability in humans, with heart failure being the most expensive single diagnosis in US healthcare. Protein degradation by the ubiquitin-proteasome system (UPS) is pivotal to protein quantity and quality control in the cell. UPS dysfunction especially proteasome functional insufficiency and increased proteotoxic stress (IPTS) play a major pathogenic role in the progression from a large subset of heart disease to heart failure and, accordingly, proteasome enhancement promises to become a new strategy to battle heart disease with IPTS. Developing more effective therapeutics to enhance the proteasome, however, requires better understanding how proteasome function is regulated. Recent advance in cell biology excitingly unravels the proteasome as a nodal point for controlling UPS proteolytic function. For example, it is recently reported that the 19S proteasome subunit RPN6/PSMD11 can be specifically phosphorylated at Ser14 by cAMP-dependent kinase (PKA), resulting in marked increases in the proteolytic function of the proteasome in cultured cells. We have reported that cGMP-dependent kinase (PKG) activates the proteasome, facilitates proteasomal degradation of misfolded proteins and thereby protects against cardiac IPTS but the proteasome phosphosite(s) of PKG remains undefined. Phosphodiesterase (PDE) 1, an enzyme known to degrade both cGMP and cAMP and thereby suppress PKG and PKA, is upregulated in mouse and human failing hearts. We have discovered that PDE1 is increased in mouse hearts with IPTS and PDE1 inhibition (PDE1i) improves UPS performance in a PKA- and PKG-dependent manner and confers striking therapeutic benefits to a bona fide mouse model of heart disease with IPTS. We newly created the Rpn6S14A and Rpn6S14D gene knock-in mice and confirmed that proteasome activation by PKA is specifically blocked and mimicked in them, respectively. Our pilot studies reveal that elevating cGMP but not cAMP further increases myocardial 26S proteasome activities in the Rpn6S14D mice. Hence, we propose and this project will test the novel hypothesis that distinctive but complementary molecular mechanisms are taken by PKG and PKA to activate the proteasome such that dual activation of PKG and PKA can complementarily enhance proteasome proteolytic function and thereby protect against cardiac IPTS. We will identify proteasome phosphosite(s) responsible for the proteasome activation by PKG using tandem mass spectrometry based proteomics combined with biochemical and genetic interrogations, determine the role of proteasome phosphoregulation by PKA in PDE1i-induced protection against cardiac IPTS, and test the effects of PKG and PKA duo-activation on UPS performance and protection against cardiac IPTS. Completion of this project will not only delineate the molecular mechanism by which PKG regulates the proteasome but also provide comprehensive experimental basis for developing PKG and PKA duo-activation as a novel strategy to treat heart disease with IPTS, a large subset of heart disease in humans.

Public Health Relevance

Despite recent advances in both basic research and clinical management, heart disease and resultant heart failure remain the leading cause of death and disability in the US and even for the entire mankind. This research project will test a potential new therapeutic strategy by exploitation of how heart muscle cells control their protein breakdown pathways to increase their ability to remove unwanted and unneeded proteins in the cell during diseased states. This will help deepen our understanding of the molecular mechanisms underlying the progression from a large subset of heart diseases to heart failure and contribute to the search for new strategies to prevent or more effectively treat this common and yet life-threatening disorder.

Agency
National Institute of Health (NIH)
Institute
National Heart, Lung, and Blood Institute (NHLBI)
Type
Research Project (R01)
Project #
2R01HL072166-15A1
Application #
10046317
Study Section
Myocardial Ischemia and Metabolism Study Section (MIM)
Program Officer
Adhikari, Bishow B
Project Start
2003-07-01
Project End
2024-06-30
Budget Start
2020-09-05
Budget End
2021-06-30
Support Year
15
Fiscal Year
2020
Total Cost
Indirect Cost
Name
University of South Dakota
Department
Other Basic Sciences
Type
Schools of Medicine
DUNS #
929930808
City
Vermillion
State
SD
Country
United States
Zip Code
57069
Sane, Sanam; Hafner, Andre; Srinivasan, Rekha et al. (2018) UBXN2A enhances CHIP-mediated proteasomal degradation of oncoprotein mortalin-2 in cancer cells. Mol Oncol 12:1753-1777
Hu, Chengjun; Tian, Yihao; Xu, Hongxin et al. (2018) Inadequate ubiquitination-proteasome coupling contributes to myocardial ischemia-reperfusion injury. J Clin Invest 128:5294-5306
Wu, Penglong; Yuan, Xun; Li, Faqian et al. (2017) Myocardial Upregulation of Cathepsin D by Ischemic Heart Disease Promotes Autophagic Flux and Protects Against Cardiac Remodeling and Heart Failure. Circ Heart Fail 10:
Abdullah, Ammara; Eyster, Kathleen M; Bjordahl, Travis et al. (2017) Murine Myocardial Transcriptome Analysis Reveals a Critical Role of COPS8 in the Gene Expression of Cullin-RING Ligase Substrate Receptors and Redox and Vesicle Trafficking Pathways. Front Physiol 8:594
Reihe, Casey A; Pekas, Nickolas; Wu, Penglong et al. (2017) Systemic inhibition of neddylation by 3-day MLN4924 treatment regime does not impair autophagic flux in mouse hearts and brains. Am J Cardiovasc Dis 7:134-150
Pan, Bo; Zhang, Hanming; Cui, Taixing et al. (2017) TFEB activation protects against cardiac proteotoxicity via increasing autophagic flux. J Mol Cell Cardiol 113:51-62
Frick, Andreas (2017) Common and Distinct Gray Matter Alterations in Social Anxiety Disorder and Major Depressive Disorder. EBioMedicine 21:53-54
Liao, Yuning; Liu, Ningning; Hua, Xianliang et al. (2017) Proteasome-associated deubiquitinase ubiquitin-specific protease 14 regulates prostate cancer proliferation by deubiquitinating and stabilizing androgen receptor. Cell Death Dis 8:e2585
Li, Jie; Ma, Wenxia; Yue, Guihua et al. (2017) Cardiac proteasome functional insufficiency plays a pathogenic role in diabetic cardiomyopathy. J Mol Cell Cardiol 102:53-60
Hou, Ning; Wen, Ying; Yuan, Xun et al. (2017) Activation of Yap1/Taz signaling in ischemic heart disease and dilated cardiomyopathy. Exp Mol Pathol 103:267-275

Showing the most recent 10 out of 105 publications