Combinatorial interactions of genetic variants in human cardiomyopathy

Dilated cardiomyopathy (DCM) is a leading cause of morbidity and mortality worldwide; yet how genetic variation and environmental factors impact DCM heritability remains unclear. Here, we report that compound genetic interactions between DNA sequence variants contribute to the complex heritability of DCM. By using genetic data from a large family with a history of DCM, we discovered that heterozygous sequence variants in the TROPOMYOSIN 1 (TPM1) and VINCULIN (VCL) genes cosegregate in individuals affected by DCM. In vitro studies of patient-derived and isogenic human-pluripotent-stem-cell-derived cardiomyocytes that were genome-edited via CRISPR to create an allelic series of TPM1 and VCL variants revealed that cardiomyocytes with both TPM1 and VCL variants display reduced contractility and sarcomeres that are less organized. Analyses of mice genetically engineered to harbour these human TPM1 and VCL variants show that stress on the heart may also influence the variable penetrance and expressivity of DCM-associated genetic variants in vivo. We conclude that compound genetic variants can interact combinatorially to induce DCM, particularly when influenced by other disease-provoking stressors.Genome-edited human pluripotent stem cells and genome-edited mouse models reveal that combinatorial genetic interactions contribute to the complex genetic heritability of human cardiomyopathy.

[1]  Juan C. del Álamo,et al.  Three-Dimensional Quantification of Cellular Traction Forces and Mechanosensing of Thin Substrata by Fourier Traction Force Microscopy , 2013, PloS one.

[2]  Donald M Bers,et al.  Epigenetic Regulation of Phosphodiesterases 2A and 3A Underlies Compromised β-Adrenergic Signaling in an iPSC Model of Dilated Cardiomyopathy. , 2015, Cell stem cell.

[3]  D. Hedges,et al.  Dilated cardiomyopathy: the complexity of a diverse genetic architecture , 2013, Nature Reviews Cardiology.

[4]  B. Maron,et al.  Double or compound sarcomere mutations in hypertrophic cardiomyopathy: a potential link to sudden death in the absence of conventional risk factors. , 2012, Heart rhythm.

[5]  P. Robinson,et al.  Doubly heterozygous LMNA and TTN mutations revealed by exome sequencing in a severe form of dilated cardiomyopathy , 2013, European Journal of Human Genetics.

[6]  G. Phillips,et al.  Crystal structure of tropomyosin at 7 Ångstroms resolution , 2000, Proteins.

[7]  A. Kimura Molecular genetics and pathogenesis of cardiomyopathy , 2015, Journal of Human Genetics.

[8]  Karen S. Frese,et al.  Atlas of the clinical genetics of human dilated cardiomyopathy. , 2014, European heart journal.

[9]  M. Robinson,et al.  Differential analyses for RNA-seq: transcript-level estimates improve gene-level inferences , 2015, F1000Research.

[10]  Thomas R. Gingeras,et al.  STAR: ultrafast universal RNA-seq aligner , 2013, Bioinform..

[11]  M. Robinson,et al.  Differential analyses for RNA-seq: transcript-level estimates improve gene-level inferences. , 2015, F1000Research.

[12]  M. Suematsu,et al.  Distinct metabolic flow enables large-scale purification of mouse and human pluripotent stem cell-derived cardiomyocytes. , 2013, Cell stem cell.

[13]  Christopher M. DeBoever,et al.  iPSCORE: A Resource of 222 iPSC Lines Enabling Functional Characterization of Genetic Variation across a Variety of Cell Types , 2017, Stem cell reports.

[14]  P. Singal,et al.  Role of oxidative stress in transition of hypertrophy to heart failure. , 1996, Journal of the American College of Cardiology.

[15]  Holger Klein,et al.  dupRadar: a Bioconductor package for the assessment of PCR artifacts in RNA-Seq data , 2016, BMC Bioinformatics.

[16]  F. Whitby,et al.  Mutations that alter the surface charge of alpha-tropomyosin are associated with dilated cardiomyopathy. , 2001, Journal of molecular and cellular cardiology.

[17]  G. Abecasis,et al.  Merlin—rapid analysis of dense genetic maps using sparse gene flow trees , 2002, Nature Genetics.

[18]  George Church,et al.  Titin mutations in iPS cells define sarcomere insufficiency as a cause of dilated cardiomyopathy , 2015, Science.

[19]  E. Boerwinkle,et al.  Genetic Risk, Adherence to a Healthy Lifestyle, and Coronary Disease. , 2016, The New England journal of medicine.

[20]  Benjamin Meder,et al.  A genome-wide association study identifies 6p21 as novel risk locus for dilated cardiomyopathy. , 2014, European heart journal.

[21]  R. Jaenisch,et al.  Generating genetically modified mice using CRISPR/Cas-mediated genome engineering , 2014, Nature Protocols.

[22]  Davide Heller,et al.  STRING v10: protein–protein interaction networks, integrated over the tree of life , 2014, Nucleic Acids Res..

[23]  Yusu Gu,et al.  (cid:1) 1 Integrin Gene Excision in the Adult Murine Cardiac Myocyte Causes Defective Mechanical and Signaling Responses , 2012 .

[24]  K. Frazer,et al.  Brief Report: Oxidative Stress Mediates Cardiomyocyte Apoptosis in a Human Model of Danon Disease and Heart Failure , 2015, Stem cells.

[25]  W. Huber,et al.  Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2 , 2014, Genome Biology.

[26]  O. Frazier,et al.  Metabolic Gene Expression in Fetal and Failing Human Heart , 2001, Circulation.

[27]  Sean P. Palecek,et al.  Directed cardiomyocyte differentiation from human pluripotent stem cells by modulating Wnt/β-catenin signaling under fully defined conditions , 2012, Nature Protocols.

[28]  S. Rosenkranz,et al.  Alterations of β-adrenergic signaling and cardiac hypertrophy in transgenic mice overexpressing TGF-β1 , 2002 .

[29]  Rudolf Jaenisch,et al.  One-Step Generation of Mice Carrying Mutations in Multiple Genes by CRISPR/Cas-Mediated Genome Engineering , 2013, Cell.

[30]  M. Bainbridge,et al.  A Potential Oligogenic Etiology of Hypertrophic Cardiomyopathy: A Classic Single-Gene Disorder , 2017, Circulation research.

[31]  G. Church,et al.  CRISPR-Cas-mediated targeted genome editing in human cells. , 2014, Methods in molecular biology.

[32]  M. Carter,et al.  A Regulatory Mechanism That Detects Premature Nonsense Codons in T-cell Receptor Transcripts in Vivo Is Reversed by Protein Synthesis Inhibitors in Vitro* , 1995, The Journal of Biological Chemistry.

[33]  Pak Chung Sham,et al.  Genetic Power Calculator: design of linkage and association genetic mapping studies of complex traits , 2003, Bioinform..

[34]  Prashant Mali,et al.  Genome editing in human stem cells. , 2014, Methods in enzymology.

[35]  M. Fornage,et al.  Heart Disease and Stroke Statistics—2017 Update: A Report From the American Heart Association , 2017, Circulation.

[36]  Adam J Engler,et al.  Preparation of Hydrogel Substrates with Tunable Mechanical Properties , 2010, Current protocols in cell biology.

[37]  James Y. Zou Analysis of protein-coding genetic variation in 60,706 humans , 2015, Nature.

[38]  S. Jamieson,et al.  Beta 1- and beta 2-adrenergic-receptor subpopulations in nonfailing and failing human ventricular myocardium: coupling of both receptor subtypes to muscle contraction and selective beta 1-receptor down-regulation in heart failure. , 1986, Circulation research.

[39]  Lior Pachter,et al.  Near-optimal probabilistic RNA-seq quantification , 2016, Nature Biotechnology.

[40]  Christopher M. DeBoever,et al.  iPSCORE: A Resource of 222 iPSC Lines Enabling Functional Characterization of Genetic Variation across a Variety of Cell Types. , 2017, Stem cell reports.

[41]  R. Maroofian,et al.  Digenic inheritance of mutations in the cardiac troponin (TNNT2) and cardiac beta myosin heavy chain (MYH7) as the cause of severe dilated cardiomyopathy. , 2017, European journal of medical genetics.

[42]  Donald Wolfgeher,et al.  Population-Based Variation in Cardiomyopathy Genes , 2012, Circulation. Cardiovascular genetics.

[43]  L. Fauchier,et al.  A genome-wide association study identifies two loci associated with heart failure due to dilated cardiomyopathy. , 2011, European heart journal.

[44]  Christine L. Mummery,et al.  Three-dimensional cardiac microtissues composed of cardiomyocytes and endothelial cells co-differentiated from human pluripotent stem cells , 2017, Development.

[45]  P. Stenson,et al.  The Human Gene Mutation Database: towards a comprehensive repository of inherited mutation data for medical research, genetic diagnosis and next-generation sequencing studies , 2017, Human Genetics.

[46]  Adam J Engler,et al.  Mechanical Forces Reshape Differentiation Cues That Guide Cardiomyogenesis. , 2016, Circulation research.

[47]  E. Adamson,et al.  Vinculin knockout results in heart and brain defects during embryonic development. , 1998, Development.

[48]  Eli J. Fine,et al.  DNA targeting specificity of RNA-guided Cas9 nucleases , 2013, Nature Biotechnology.

[49]  E. McNally,et al.  Genetic Variation in Cardiomyopathy and Cardiovascular Disorders. , 2015, Circulation journal : official journal of the Japanese Circulation Society.

[50]  Gwendolyn M. Jang,et al.  Meta- and Orthogonal Integration of Influenza "OMICs" Data Defines a Role for UBR4 in Virus Budding. , 2015, Cell host & microbe.

[51]  Bronwen L. Aken,et al.  GENCODE: The reference human genome annotation for The ENCODE Project , 2012, Genome research.

[52]  J. Ross,et al.  Segregation of atrial-specific and inducible expression of an atrial natriuretic factor transgene in an in vivo murine model of cardiac hypertrophy , 1991, Proceedings of the National Academy of Sciences of the United States of America.

[53]  Richard D. Friedman,et al.  Consanguinity Mapping of Congenital Heart Disease in a South Indian Population , 2010, PloS one.

[54]  Lan Lin,et al.  rMATS: Robust and flexible detection of differential alternative splicing from replicate RNA-Seq data , 2014, Proceedings of the National Academy of Sciences.