Heart failure (HF) is the leading cause of adult hospitalization in the industrialized world and imposes a substantial burden on the public health. Over the past two decades, a large body of research using transgenic models has implicated a growing number of transcription factors and microRNAs as mediators of myocardial hypertrophy and dysfunction. Because few of these mediators have been confirmed in human hearts, there has been minimal progress in applying these insights to human HF therapeutics. We propose to overcome these barriers by leveraging unique bioresources at three U.S. transplant centers that have amassed repositories of high-quality myocardium from more than 1,900 human subjects over the past 15 years. The overall goals of this proposal are to use integrative genomics to test whether transcriptional regulatory programs identified in animal models are relevant in human HF and to perform unbiased screens for regulators of myocardial gene expression in human subjects. Integrative genomics elucidates disease mechanism by combining phenotype data with whole-genome genotypes and gene expression in a tissue of interest. To apply these approaches to the human heart, we have assembled a multidisciplinary team of experts in heart failure, clinical investigation, cardiac biology, and the genetics of complex disorders.
In Aim 1, we will perform a case-control study (n=1000) using integrated SNP and expression data to test which of 40 pre-specified transcriptional regulators contribute to advanced HF in humans.
In Aim 2, we will perform a cross-sectional study (n=1000) using integrated SNP and expression data across a broad range of myocardial phenotypes to test whether the same candidate regulators contribute to pathological remodeling. In both aims, secondary analyses and network modeling will enable genome-wide screens for unanticipiated mechanisms of transcriptional regulation in the human heart.
In Aim 3, we will test whether our most promising results identify genetic risk factors for cardiac remodeling in the general population through collaboration with EchoGen, a genome-wide association meta-analysis of echocardiographic traits in seven community-based cohorts (N=18,000). This research will test the relevance of knowledge derived from years of animal research while employing an unbiased discovery approach to reveal unanticipated mechanisms of human myocardial disease. Doing so will accelerate the translation of scientific knowledge to HF therapeutics. Moreover, all data and biosamples will be made available to the scientific community to promote a broad and durable impact on HF research.

Public Health Relevance

In the midst of an ongoing heart failure epidemic, this research will determine the clinical relevance of heart failure mechanisms identified in animal models, help identify new therapeutic targets, and define mechanisms through which genetic variation influences the development of heart failure. By advancing clinical research in myocardial mechanisms of disease progression, this project will accelerate the development of therapeutic applications to improve the care of patients with heart failure.

Agency
National Institute of Health (NIH)
Institute
National Heart, Lung, and Blood Institute (NHLBI)
Type
Research Project (R01)
Project #
1R01HL105993-01A1
Application #
8187371
Study Section
Clinical and Integrative Cardiovascular Sciences Study Section (CICS)
Program Officer
Paltoo, Dina
Project Start
2011-08-01
Project End
2015-04-30
Budget Start
2011-08-01
Budget End
2012-04-30
Support Year
1
Fiscal Year
2011
Total Cost
$2,325,744
Indirect Cost
Name
University of Pennsylvania
Department
Internal Medicine/Medicine
Type
Schools of Medicine
DUNS #
042250712
City
Philadelphia
State
PA
Country
United States
Zip Code
19104
Hu, Ray; Morley, Michael P; Brandimarto, Jeffrey et al. (2018) Genetic Reduction in Left Ventricular Protein Kinase C-? and Adverse Ventricular Remodeling in Human Subjects. Circ Genom Precis Med 11:e001901
Truitt, Rachel; Mu, Anbin; Corbin, Elise A et al. (2018) Increased Afterload Augments Sunitinib-Induced Cardiotoxicity in an Engineered Cardiac Microtissue Model. JACC Basic Transl Sci 3:265-276
Chen, Christina Yingxian; Caporizzo, Matthew A; Bedi, Kenneth et al. (2018) Suppression of detyrosinated microtubules improves cardiomyocyte function in human heart failure. Nat Med 24:1225-1233
Peyster, Eliot G; Madabhushi, Anant; Margulies, Kenneth B (2018) Advanced Morphologic Analysis for Diagnosing Allograft Rejection: The Case of Cardiac Transplant Rejection. Transplantation 102:1230-1239
Zhang, Xiaoming; Yoon, Jin-Young; Morley, Michael et al. (2018) A common variant alters SCN5A-miR-24 interaction and associates with heart failure mortality. J Clin Invest 128:1154-1163
Bec, Julien; Phipps, Jennifer E; Gorpas, Dimitris et al. (2017) In vivo label-free structural and biochemical imaging of coronary arteries using an integrated ultrasound and multispectral fluorescence lifetime catheter system. Sci Rep 7:8960
Wild, Philipp S; Felix, Janine F; Schillert, Arne et al. (2017) Large-scale genome-wide analysis identifies genetic variants associated with cardiac structure and function. J Clin Invest 127:1798-1812
Vikram, Ajit; Lewarchik, Christopher M; Yoon, Jin-Young et al. (2017) Sirtuin 1 regulates cardiac electrical activity by deacetylating the cardiac sodium channel. Nat Med 23:361-367
Parikh, Victoria N; Ashley, Euan A (2017) Next-Generation Sequencing in Cardiovascular Disease: Present Clinical Applications and the Horizon of Precision Medicine. Circulation 135:406-409
Narayan, Hari K; Finkelman, Brian; French, Benjamin et al. (2017) Detailed Echocardiographic Phenotyping in Breast Cancer Patients: Associations With Ejection Fraction Decline, Recovery, and Heart Failure Symptoms Over 3 Years of Follow-Up. Circulation 135:1397-1412

Showing the most recent 10 out of 41 publications