Among the familial cardiomyopathies, cardiac laminopathy caused by LMNA mutations accounts for approximately 6% of all cases. Compared to other DCM patients with left ventricular dilation, 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 due to limited patient-derived bio-specimens and lack of appropriate disease models. To overcome the problem, in this R01 grant renewal application, we will 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 clarify the detailed molecular mechanisms, we will conduct structural, electrophysiological, developmental, transcriptome, and mechanistic analyses using patient-specific as well as genome-edited isogenic iPSC-CMs and iPSC-ECs. Importantly, we will perform drug screening studies targeting dysregulated signaling pathways. To confirm the beneficial action of identified targeting compounds, we will then conduct transcriptomic and functional analysis of isogenic LMNA iPSC-CM and iPSC-ECs using high throughput platforms. Collectively, these studies will uncover mechanistic insights of LMNA cardiolaminopathy, a major cause of DCM, which may help identify novel therapeutic candidates that can target CMs and ECs, two key cell populations.

Public Health Relevance

This study will provide much needed information to understand the complex cardiomyopathy induced by LMNA mutations. LMNA cardiolaminopathy is a common cause of familial cardiac disease, accounting for 6% of all cases, and more than 160 mutations have been identified as pathogenic. However, the detailed molecular mechanisms LMNA cardiolaminopathy remain elusive due to lack of appropriate disease models. 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 molecular pathogenesis. Our preliminary data have revealed specific deregulated signaling pathways in LMNA iPSC-CMs and iPSC-ECs that might represent potential targets for focused drug screening assays. Results obtained here may provide further insights into the molecular pathogenesis of LMNA cardiolaminopathy and promote the development of novel therapeutic drugs that can rescue the disease phenotype.

Agency
National Institute of Health (NIH)
Institute
National Heart, Lung, and Blood Institute (NHLBI)
Type
Research Project (R01)
Project #
2R01HL113006-06
Application #
9383096
Study Section
Special Emphasis Panel (ZRG1)
Program Officer
Buxton, Denis B
Project Start
2012-05-01
Project End
2021-07-31
Budget Start
2017-08-01
Budget End
2018-07-31
Support Year
6
Fiscal Year
2017
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; Oikonomopoulos, Angelos; Chen, Haodong et al. (2018) Genome Editing of Induced Pluripotent Stem Cells to Decipher Cardiac Channelopathy Variant. J Am Coll Cardiol 72:62-75
Rhee, June-Wha; Wu, Joseph C (2018) Cardiac Cell Cycle Activation as a Strategy to Improve iPSC-Derived Cardiomyocyte Therapy. Circ Res 122:14-16
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

Showing the most recent 10 out of 73 publications