Dilated cardiomyopathy (DCM) is a leading cause of morbidity and mortality worldwide, with an estimated prevalence of 1:250 in the general population. Recent human genetic studies suggest that the complex heritability of DCM may be due to a combination of polygenic causes. However, the complex interactions among various genetic variants are less understood, hampering the development of effective therapeutics for DCM. To achieve the latter, we will use state-of-the-art approaches by employing the induced pluripotent stem cell-derived cardiomyocyte (iPSC-CM) platform coupled with tissue engineering, single cell RNA sequencing (scRNA-seq), and CRISPR/dCas9 genome screening technologies to interrogate the interplay between genetic variants in the etiology of DCM and identify novel drug target for polygenic DCM treatment.
In Aim 1, we will generate iPSC lines from 20 polygenic DCM patients and 10 controls from the same family cohort, and create 120 genome- edited isogenic iPSC lines using CRISPR/Cas9. We will then differentiate the polygenic DCM iPSCs and corresponding isogenic iPSC lines to iPSC-CMs. Using bioinformatic analysis, we will generate a gene expression ?score index? to assess the pathogenicity of each genetic variant and interactions between different genetic variants.
In Aim 2, we will generate 3D engineered heart tissues (EHTs) to elucidate functional consequences of polygenic DCM variants at the 3D level. We will make 3D EHTs consisting of iPSC-CMs, iPSC- derived endothelial cells (iPSC-ECs), and iPSC-derived fibroblasts (iPSC-FBs) from these 20 polygenic DCM patients and their genome-edited isogenic iPSC lines, which allow us to simulate complex cell-cell interactions and understand the crosstalk among different cell types.
In Aim 3, we will use CRISPR/dCas9 genome screening approach in combination with scRNA-seq technology to identify genes suitable as drug targets in polygenic DCM iPSC-CMs. This novel technique provides a unique and cost-efficient way to systemically screen for druggable genes associated with polygenic DCM. Collectively, the proposed aims will help us comprehensively elucidate the genetic and molecular basis of polygenic DCM and discover novel drug targets for patient-specific therapeutics.

Public Health Relevance

Recent human genetic studies suggest that the complex heritability of dilated cardiomyopathy (DCM) may be due to a combination of polygenic causes; however, the functional evidence for these possible etiologies remains unclear. We previously demonstrated that patient-specific induced pluripotent stem cell-derived cardiomyocytes (iPSC-CMs) can serve as an effective platform for modeling DCM by recapitulating the clinical phenotype of DCM in a dish. In this proposal, we will interrogate the interplay between genetic variants in the etiology of polygenic DCM and screen drug targets for polygenic DCM treatment by employing the iPSC-CM platform coupled with engineered heart tissue (EHT), single cell RNA sequencing (scRNA-seq), and CRISPR/dCas9 genome screening technologies. The knowledge we collect here should be instrumental in developing novel patient-specific therapeutics to treat polygenic DCM.

Agency
National Institute of Health (NIH)
Institute
National Heart, Lung, and Blood Institute (NHLBI)
Type
Research Project (R01)
Project #
2R01HL130020-05
Application #
9884902
Study Section
Cardiac Contractility, Hypertrophy, and Failure Study Section (CCHF)
Program Officer
Burns, Kristin
Project Start
2015-12-16
Project End
2023-11-30
Budget Start
2019-12-15
Budget End
2020-11-30
Support Year
5
Fiscal Year
2020
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
94305
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
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
Churko, Jared M; Garg, Priyanka; Treutlein, Barbara et al. (2018) Defining human cardiac transcription factor hierarchies using integrated single-cell heterogeneity analysis. Nat Commun 9:4906
Chang, Alex C Y; Chang, Andrew C H; Kirillova, Anna et al. (2018) Telomere shortening is a hallmark of genetic cardiomyopathies. Proc Natl Acad Sci U S A 115:9276-9281
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 :
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
Ma, Ning; Zhang, Joe Z; Itzhaki, Ilanit et al. (2018) Determining the Pathogenicity of a Genomic Variant of Uncertain Significance Using CRISPR/Cas9 and Human-Induced Pluripotent Stem Cells. Circulation 138:2666-2681
Liu, Chun; Oikonomopoulos, Angelos; Sayed, Nazish et al. (2018) Modeling human diseases with induced pluripotent stem cells: from 2D to 3D and beyond. Development 145:

Showing the most recent 10 out of 31 publications