of Parent R01: LMNA mutations accounts for approximately 6% of cases in familial dilated cardiomyopathies (DCM). Compared to other DCM patients, LMNA cardiolaminopathy patients exhibit a severe clinical course, conduction abnormalities, and a high rate of heart failure. LMNA DCM has a complex pathophysiology and it has been hypothesized that this form of cardiac disease is due to defects in function of cardiomyocytes (CMs) as well as defects in non-CM populations such as endothelial cells (ECs). However, the detailed molecular mechanisms of LMNA cardiolaminopathy remain elusive. Accordingly, the parent R01 proposes to generate human induced pluripotent stem cell- derived cardiomyocytes (iPSC-CMs) from LMNA DCM patients as well as matched family controls and healthy unrelated controls. To illuminate molecular mechanisms, we are performing structural, electrophysiological, transcriptomic, and mechanistic analyses using patient-specific as well as genome-edited isogenic iPSC-CMs and iPSC-ECs. We will also perform drug screening studies on these cell types to target dysregulated signaling pathways and identify targeting compounds. Collectively, these studies are anticipated to uncover mechanistic insights of LMNA cardiolaminopathy, a major cause of DCM, and may help identify novel therapeutic candidates that can target CMs and ECs, two key cell populations. Proposed Supplement: In the proposed Research Supplement, we will extend the parent R01 to include two critically related sets of experiments. First, we will expand the cohort of DCM patients and healthy control to include 20 additional individuals of African American ethnicity, which will lay the groundwork for exploring population-specific effects of DCM and allow systematic comparison of population differences in the proposed drug screening effort. Secondly, we will take advantages of latest advances in single-cell RNA sequencing (scRNA-seq) and characterize DCM iPSC-CMs and iPSC-ECs at a single cell level and identify disease mechanisms free from confounding effects of cellular heterogeneity. The proposed experiments will be performed by, and will enable the training of, Mr. Damon Williams, a recent baccalaureate graduate who is a member of a group underrepresented in biomedical research (African Americans).

Public Health Relevance

LMNA cardiolaminopathy accounts for 6% of all familial cardiomyopathies, and more than 160 mutations have been identified as pathogenic. In the proposed studies, we will use induced pluripotent stem cells (iPSCs) from LMNA cardiolaminopathy patients and genome-edited isogenic iPSC lines to elucidate the pathogenic mechanisms cardiolaminopathy. Results obtained here may provide clues for the development of novel therapeutic drugs for this common inherited heart disease.

Agency
National Institute of Health (NIH)
Institute
National Heart, Lung, and Blood Institute (NHLBI)
Type
Research Project (R01)
Project #
3R01HL113006-06S1
Application #
9674139
Study Section
Program Officer
Wong, Renee P
Project Start
2012-05-01
Project End
2021-07-31
Budget Start
2018-07-15
Budget End
2018-07-31
Support Year
6
Fiscal Year
2018
Total Cost
Indirect Cost
Name
Stanford University
Department
Internal Medicine/Medicine
Type
Schools of Medicine
DUNS #
009214214
City
Stanford
State
CA
Country
United States
Zip Code
94304
Garg, Priyanka; Garg, Vivek; Shrestha, Rajani et al. (2018) Human Induced Pluripotent Stem Cell-Derived Cardiomyocytes as Models for Cardiac Channelopathies: A Primer for Non-Electrophysiologists. Circ Res 123:224-243
Lee, Andrew S; Inayathullah, Mohammed; Lijkwan, Maarten A et al. (2018) Prolonged survival of transplanted stem cells after ischaemic injury via the slow release of pro-survival peptides from a collagen matrix. Nat Biomed Eng 2:104-113
Zhao, Xin; Chen, Haodong; Xiao, Dan et al. (2018) Comparison of Non-human Primate versus Human Induced Pluripotent Stem Cell-Derived Cardiomyocytes for Treatment of Myocardial Infarction. Stem Cell Reports 10:422-435
Oikonomopoulos, Angelos; Kitani, Tomoya; Wu, Joseph C (2018) Pluripotent Stem Cell-Derived Cardiomyocytes as a Platform for Cell Therapy Applications: Progress and Hurdles for Clinical Translation. Mol Ther 26:1624-1634
Lee, Jaecheol; Shao, Ning-Yi; Paik, David T et al. (2018) SETD7 Drives Cardiac Lineage Commitment through Stage-Specific Transcriptional Activation. Cell Stem Cell 22:428-444.e5
Knowles, Joshua W; Ashley, Euan A (2018) Cardiovascular disease: The rise of the genetic risk score. PLoS Med 15:e1002546
Wnorowski, Alexa; Yang, Huaxiao; Wu, Joseph C (2018) Progress, obstacles, and limitations in the use of stem cells in organ-on-a-chip models. Adv Drug Deliv Rev :
Liu, Qing; Van Bortle, Kevin; Zhang, Yue et al. (2018) Disruption of mesoderm formation during cardiac differentiation due to developmental exposure to 13-cis-retinoic acid. Sci Rep 8:12960
Paik, David T; Tian, Lei; Lee, Jaecheol et al. (2018) Large-Scale Single-Cell RNA-Seq Reveals Molecular Signatures of Heterogeneous Populations of Human Induced Pluripotent Stem Cell-Derived Endothelial Cells. Circ Res 123:443-450
Sharma, Arun; Zhang, Yuan; Buikema, Jan W et al. (2018) Stage-specific Effects of Bioactive Lipids on Human iPSC Cardiac Differentiation and Cardiomyocyte Proliferation. Sci Rep 8:6618

Showing the most recent 10 out of 73 publications