Rewiring of 3D Chromatin Topology Orchestrates Transcriptional Reprogramming and the Development of Human Dilated Cardiomyopathy

Background: Transcriptional reconfiguration is central to heart failure, the most common cause of which is dilated cardiomyopathy (DCM). The effect of 3-dimensional chromatin topology on transcriptional dysregulation and pathogenesis in human DCM remains elusive. Methods: We generated a compendium of 3-dimensional epigenome and transcriptome maps from 101 biobanked human DCM and nonfailing heart tissues through highly integrative chromatin immunoprecipitation (H3K27ac [acetylation of lysine 27 on histone H3]), in situ high–throughput chromosome conformation capture, chromatin immunoprecipitation sequencing, assay for transposase-accessible chromatin using sequencing, and RNA sequencing. We used human induced pluripotent stem cell–derived cardiomyocytes and mouse models to interrogate the key transcription factor implicated in 3-dimensional chromatin organization and transcriptional regulation in DCM pathogenesis. Results: We discovered that the active regulatory elements (H3K27ac peaks) and their connectome (H3K27ac loops) were extensively reprogrammed in DCM hearts and contributed to transcriptional dysregulation implicated in DCM development. For example, we identified that nontranscribing NPPA-AS1 (natriuretic peptide A antisense RNA 1) promoter functions as an enhancer and physically interacts with the NPPA (natriuretic peptide A) and NPPB (natriuretic peptide B) promoters, leading to the cotranscription of NPPA and NPPB in DCM hearts. We revealed that DCM-enriched H3K27ac loops largely resided in conserved high-order chromatin architectures (compartments, topologically associating domains) and their anchors unexpectedly had equivalent chromatin accessibility. We discovered that the DCM-enriched H3K27ac loop anchors exhibited a strong enrichment for HAND1 (heart and neural crest derivatives expressed 1), a key transcription factor involved in early cardiogenesis. In line with this, its protein expression was upregulated in human DCM and mouse failing hearts. To further validate whether HAND1 is a causal driver for the reprogramming of enhancer–promoter connectome in DCM hearts, we performed comprehensive 3-dimensional epigenome mappings in human induced pluripotent stem cell–derived cardiomyocytes. We found that forced overexpression of HAND1 in human induced pluripotent stem cell–derived cardiomyocytes induced a distinct gain of enhancer–promoter connectivity and correspondingly increased the expression of their connected genes implicated in DCM pathogenesis, thus recapitulating the transcriptional signature in human DCM hearts. Electrophysiology analysis demonstrated that forced overexpression of HAND1 in human induced pluripotent stem cell–derived cardiomyocytes induced abnormal calcium handling. Furthermore, cardiomyocyte-specific overexpression of Hand1 in the mouse hearts resulted in dilated cardiac remodeling with impaired contractility/Ca2+ handling in cardiomyocytes, increased ratio of heart weight/body weight, and compromised cardiac function, which were ascribed to recapitulation of transcriptional reprogramming in DCM. Conclusions: This study provided novel chromatin topology insights into DCM pathogenesis and illustrated a model whereby a single transcription factor (HAND1) reprograms the genome-wide enhancer–promoter connectome to drive DCM pathogenesis.

[1]  M. Nóbrega,et al.  Genetic Variation in Enhancers Modifies Cardiomyopathy Gene Expression and Progression. , 2021, Circulation.

[2]  P. Pinton,et al.  Mitochondrial Function and Dysfunction in Dilated Cardiomyopathy , 2021, Frontiers in Cell and Developmental Biology.

[3]  Bjoern Peters,et al.  Promoter-interacting expression quantitative trait loci are enriched for functional genetic variants , 2020, Nature Genetics.

[4]  M. Nóbrega,et al.  Altered Enhancer and Promoter Usage Leads to Differential Gene Expression in the Normal and Failed Human Heart , 2020, Circulation. Heart failure.

[5]  T. Misteli The Self-Organizing Genome: Principles of Genome Architecture and Function , 2020, Cell.

[6]  J. Wong,et al.  The Gridlock transcriptional repressor impedes vertebrate heart regeneration by restricting expression of lysine methyltransferase , 2020, Development.

[7]  R. Qu,et al.  An Overview of the Cytoskeleton-Associated Role of PDLIM5 , 2020, Frontiers in Physiology.

[8]  E. Kranias,et al.  Phosphorylation of cardiac myosin–binding protein-C contributes to calcium homeostasis , 2020, The Journal of Biological Chemistry.

[9]  May Yin Lee,et al.  Epigenomes of Human Hearts Reveal New Genetic Variants Relevant for Cardiac Disease and Phenotype , 2020, Circulation research.

[10]  Rolf Backofen,et al.  Galaxy HiCExplorer 3: a web server for reproducible Hi-C, capture Hi-C and single-cell Hi-C data analysis, quality control and visualization , 2020, Nucleic Acids Res..

[11]  Howard Y. Chang,et al.  Circular ecDNA promotes accessible chromatin and high oncogene expression , 2019, Nature.

[12]  William A. Flavahan,et al.  Altered chromosomal topology drives oncogenic programs in SDH-deficient GISTs , 2019, Nature.

[13]  N. Pereira,et al.  Genetics of dilated cardiomyopathy: practical implications for heart failure management , 2019, Nature Reviews Cardiology.

[14]  P. Ellinor,et al.  Antisense regulation of atrial natriuretic peptide expression. , 2019, JCI insight.

[15]  Wei Xie,et al.  The role of 3D genome organization in development and cell differentiation , 2019, Nature Reviews Molecular Cell Biology.

[16]  Xiao-gang Guo,et al.  Novel polymorphisms in PDLIM3 and PDLIM5 gene encoding Z‐line proteins increase risk of idiopathic dilated cardiomyopathy , 2019, Journal of cellular and molecular medicine.

[17]  Ze-Min Fang,et al.  Histone methyltransferase SMYD2: ubiquitous regulator of disease , 2019, Clinical Epigenetics.

[18]  Stefan Schoenfelder,et al.  Long-range enhancer–promoter contacts in gene expression control , 2019, Nature Reviews Genetics.

[19]  Guoliang Li,et al.  Chromatin interaction analysis with updated ChIA-PET Tool (V3) , 2019 .

[20]  Yibin Wang,et al.  Epigenetics in dilated cardiomyopathy , 2019, Current opinion in cardiology.

[21]  P. Bovolenta,et al.  Mapping the Global Chromatin Connectivity Network for Sox2 Function in Neural Stem Cell Maintenance , 2019, Cell stem cell.

[22]  Nezar Abdennur,et al.  Cooler: scalable storage for Hi-C data and other genomically-labeled arrays , 2019, bioRxiv.

[23]  Sandy L. Klemm,et al.  Chromatin accessibility and the regulatory epigenome , 2019, Nature Reviews Genetics.

[24]  Jasmin I. Maier,et al.  AIF1L regulates actomyosin contractility and filopodial extensions in human podocytes , 2018, PloS one.

[25]  Howard Y. Chang,et al.  Promoter of lncRNA Gene PVT1 Is a Tumor-Suppressor DNA Boundary Element , 2018, Cell.

[26]  Min Wang,et al.  Loss of microRNA-128 promotes cardiomyocyte proliferation and heart regeneration , 2018, Nature Communications.

[27]  Aristotelis Tsirigos,et al.  Stratification of TAD boundaries reveals preferential insulation of super-enhancers by strong boundaries , 2018, Nature Communications.

[28]  P. Elliott,et al.  Evolving concepts in dilated cardiomyopathy , 2018, European journal of heart failure.

[29]  J. Epstein,et al.  Genome-Nuclear Lamina Interactions Regulate Cardiac Stem Cell Lineage Restriction , 2017, Cell.

[30]  Howard Y. Chang,et al.  Enhancer connectome in primary human cells identifies target genes of disease-associated DNA elements , 2017, Nature Genetics.

[31]  E. Petretto,et al.  Natural genetic variation of the cardiac transcriptome in non-diseased donors and patients with dilated cardiomyopathy , 2017, Genome Biology.

[32]  Nadav Ahituv,et al.  Gene Regulatory Elements, Major Drivers of Human Disease. , 2017, Annual review of genomics and human genetics.

[33]  Michael Q. Zhang,et al.  In Situ Capture of Chromatin Interactions by Biotinylated dCas9 , 2017, Cell.

[34]  William Stafford Noble,et al.  HiCRep: assessing the reproducibility of Hi-C data using a stratum-adjusted correlation coefficient , 2017, bioRxiv.

[35]  Lan T M Dao,et al.  Genome-wide characterization of mammalian promoters with distal enhancer functions , 2017, Nature Genetics.

[36]  B. Li,et al.  A tiling1deletion based genetic screen for cis-regulatory element identification in mammalian cells , 2017, Nature Methods.

[37]  K. Hong,et al.  Progesterone Protects Against Bisphenol A–Induced Arrhythmias in Female Rat Cardiac Myocytes via Rapid Signaling , 2017, Endocrinology.

[38]  I. Moskowitz,et al.  TBX5: A Key Regulator of Heart Development. , 2017, Current topics in developmental biology.

[39]  Miao Yu,et al.  Mapping of long-range chromatin interactions by proximity ligation-assisted ChIP-seq , 2016, Cell Research.

[40]  Clint L. Miller,et al.  PDE1C deficiency antagonizes pathological cardiac remodeling and dysfunction , 2016, Proceedings of the National Academy of Sciences.

[41]  Howard Y. Chang,et al.  HiChIP: efficient and sensitive analysis of protein-directed genome architecture , 2016, Nature Methods.

[42]  Ri-tai Huang,et al.  HAND1 loss-of-function mutation associated with familial dilated cardiomyopathy , 2016, Clinical chemistry and laboratory medicine.

[43]  Neva C. Durand,et al.  Juicer Provides a One-Click System for Analyzing Loop-Resolution Hi-C Experiments. , 2016, Cell systems.

[44]  Måns Magnusson,et al.  MultiQC: summarize analysis results for multiple tools and samples in a single report , 2016, Bioinform..

[45]  Jesse R. Dixon,et al.  Chromatin Domains: The Unit of Chromosome Organization. , 2016, Molecular cell.

[46]  G. Fonarow,et al.  Epidemiology and aetiology of heart failure , 2016, Nature Reviews Cardiology.

[47]  L. Zentilin,et al.  A mouse model for adult cardiac-specific gene deletion with CRISPR/Cas9 , 2015, Proceedings of the National Academy of Sciences.

[48]  Jean-Philippe Vert,et al.  HiC-Pro: an optimized and flexible pipeline for Hi-C data processing , 2015, Genome Biology.

[49]  Michael P. Snyder,et al.  Mango: a bias-correcting ChIA-PET analysis pipeline , 2015, Bioinform..

[50]  A. Visel,et al.  Disruptions of Topological Chromatin Domains Cause Pathogenic Rewiring of Gene-Enhancer Interactions , 2015, Cell.

[51]  J. Dekker,et al.  Condensin-Driven Remodeling of X-Chromosome Topology during Dosage Compensation , 2015, Nature.

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

[53]  Fidel Ramírez,et al.  deepTools: a flexible platform for exploring deep-sequencing data , 2014, Nucleic Acids Res..

[54]  Yong-liang Wang,et al.  Progressive decay of Ca2+ homeostasis in the development of diabetic cardiomyopathy , 2014, Cardiovascular Diabetology.

[55]  Wei Shi,et al.  featureCounts: an efficient general purpose program for assigning sequence reads to genomic features , 2013, Bioinform..

[56]  L. D. de Windt,et al.  Regulation of fetal gene expression in heart failure. , 2013, Biochimica et biophysica acta.

[57]  Howard Y. Chang,et al.  Transposition of native chromatin for fast and sensitive epigenomic profiling of open chromatin, DNA-binding proteins and nucleosome position , 2013, Nature Methods.

[58]  Helga Thorvaldsdóttir,et al.  Integrative Genomics Viewer (IGV): high-performance genomics data visualization and exploration , 2012, Briefings Bioinform..

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

[60]  R. Moss,et al.  Ablation of the Cardiac-Specific Gene Leucine-Rich Repeat Containing 10 (Lrrc10) Results in Dilated Cardiomyopathy , 2012, PloS one.

[61]  Job Dekker,et al.  Analysis of long-range chromatin interactions using Chromosome Conformation Capture. , 2012, Methods.

[62]  E. Liu,et al.  Large-Scale Functional Organization of Long-Range Chromatin Interaction Networks , 2012, Cell reports.

[63]  Yong Zhang,et al.  Identifying ChIP-seq enrichment using MACS , 2012, Nature Protocols.

[64]  J. Sedat,et al.  Spatial partitioning of the regulatory landscape of the X-inactivation centre , 2012, Nature.

[65]  Euan A. Ashley,et al.  Patient-Specific Induced Pluripotent Stem Cells as a Model for Familial Dilated Cardiomyopathy , 2012, Science Translational Medicine.

[66]  Raymond K. Auerbach,et al.  Extensive Promoter-Centered Chromatin Interactions Provide a Topological Basis for Transcription Regulation , 2012, Cell.

[67]  M. Ashraf,et al.  Reduced collagen deposition in infarcted myocardium facilitates induced pluripotent stem cell engraftment and angiomyogenesis for improvement of left ventricular function. , 2011, Journal of the American College of Cardiology.

[68]  S. Moon,et al.  A novel Fbxo25 acts as an E3 ligase for destructing cardiac specific transcription factors. , 2011, Biochemical and biophysical research communications.

[69]  Timothy J. Durham,et al.  "Systematic" , 1966, Comput. J..

[70]  R. Young,et al.  Histone H3K27ac separates active from poised enhancers and predicts developmental state , 2010, Proceedings of the National Academy of Sciences.

[71]  Aaron R. Quinlan,et al.  BIOINFORMATICS APPLICATIONS NOTE , 2022 .

[72]  Mark D. Robinson,et al.  edgeR: a Bioconductor package for differential expression analysis of digital gene expression data , 2009, Bioinform..

[73]  Gonçalo R. Abecasis,et al.  The Sequence Alignment/Map format and SAMtools , 2009, Bioinform..

[74]  Jing Chen,et al.  ToppGene Suite for gene list enrichment analysis and candidate gene prioritization , 2009, Nucleic Acids Res..

[75]  Richard Durbin,et al.  Sequence analysis Fast and accurate short read alignment with Burrows – Wheeler transform , 2009 .

[76]  I. Sjaastad,et al.  Slowing of cardiomyocyte Ca2+ release and contraction during heart failure progression in postinfarction mice. , 2009, American journal of physiology. Heart and circulatory physiology.

[77]  E. Liu,et al.  Regulation of Estrogen Receptor-mediated Long Range Transcription via Evolutionarily Conserved Distal Response Elements* , 2008, Journal of Biological Chemistry.

[78]  Da-Zhi Wang,et al.  The MEF2D transcription factor mediates stress-dependent cardiac remodeling in mice. , 2008, The Journal of clinical investigation.

[79]  I. Sjaastad,et al.  Increased cardiomyocyte function and Ca2+ transients in mice during early congestive heart failure. , 2007, Journal of molecular and cellular cardiology.

[80]  Wouter de Laat,et al.  Quantitative analysis of chromosome conformation capture assays (3C-qPCR) , 2007, Nature Protocols.

[81]  B. Aronow,et al.  Myocyte Enhancer Factors 2A and 2C Induce Dilated Cardiomyopathy in Transgenic Mice* , 2006, Journal of Biological Chemistry.

[82]  P. Shannon,et al.  Cytoscape: a software environment for integrated models of biomolecular interaction networks. , 2003, Genome research.

[83]  M. Bang,et al.  Myopalladin, a Novel 145-Kilodalton Sarcomeric Protein with Multiple Roles in Z-Disc and I-Band Protein Assemblies , 2001, The Journal of cell biology.

[84]  E. Olson,et al.  Regulation of skeletal myogenesis by association of the MEF2 transcription factor with class II histone deacetylases. , 2000, Molecular cell.

[85]  J. Cross,et al.  The Hand1 bHLH transcription factor is essential for placentation and cardiac morphogenesis , 1998, Nature Genetics.

[86]  S. Chien,et al.  Regulation of cardiac gene expression during myocardial growth and hypertrophy: molecular studies of an adaptive physiologic response , 1991, FASEB journal : official publication of the Federation of American Societies for Experimental Biology.