Sequential Defects in Cardiac Lineage Commitment and Maturation Cause Hypoplastic Left Heart Syndrome

Background Complex molecular programs in specific cell lineages govern human heart development. Hypoplastic left heart syndrome (HLHS) is the most common and severe manifestation within the spectrum of left ventricular outflow tract obstruction defects occurring in association with ventricular hypoplasia. The pathogenesis of HLHS is unknown, but hemodynamic disturbances are assumed to play a prominent role. Methods To identify perturbations in gene programs controlling ventricular muscle lineage development in HLHS, we performed: i) whole-exome sequencing of 87 HLHS parent-offspring trios, ii) nuclear transcriptomics of cardiomyocytes from ventricles of 4 patients with HLHS and 15 controls at different stages of heart development, iii) single cell RNA sequencing and iv) 3D modeling in iPSCs from 3 patients with HLHS and 3 controls. Results Gene set enrichment and protein network analyses of damaging de-novo mutations and dysregulated genes from ventricles of patients with HLHS suggested alterations in specific gene programs and cellular processes critical during fetal ventricular cardiogenesis, including cell-cycle and cardiomyocyte maturation. Single-cell and 3D modeling with iPSCs demonstrated intrinsic defects in the cell-cycle/UPR/autophagy hub resulting in disrupted differentiation of early cardiac progenitor lineages leading to defective cardiomyocyte-subtype differentiation/maturation in HLHS. Additionally, premature cell-cycle exit of ventricular cardiomyocytes from HLHS patients prevented normal tissue responses to developmental signals for growth leading to multinucleation/polyploidy, accumulation of DNA damage, and exacerbated apoptosis, all potential drivers of left ventricular hypoplasia in absence of hemodynamic cues. Conclusions Our results highlight that despite genetic heterogeneity in HLHS, many mutations converge on sequential cellular processes primarily driving cardiac myogenesis, suggesting novel therapeutic approaches.

[1]  B. Aronow,et al.  iPSC modeling shows uncompensated mitochondrial mediated oxidative stress underlies early heart failure in hypoplastic left heart syndrome , 2021, bioRxiv.

[2]  Robert H. Anderson,et al.  Clarification of the definition of hypoplastic left heart syndrome , 2021, Nature Reviews Cardiology.

[3]  Timothy J. Nelson,et al.  Intrinsic Endocardial Defects Contribute to Hypoplastic Left Heart Syndrome. , 2020, Cell stem cell.

[4]  E. Wolf,et al.  Somatic gene editing ameliorates skeletal and cardiac muscle failure in pig and human models of Duchenne muscular dystrophy , 2020, Nature Medicine.

[5]  Joakim Lundeberg,et al.  A Spatiotemporal Organ-Wide Gene Expression and Cell Atlas of the Developing Human Heart , 2019, Cell.

[6]  Joe Z. Zhang,et al.  Single-Cell RNA Sequencing of Human Embryonic Stem Cell Differentiation Delineates Adverse Effects of Nicotine on Embryonic Development , 2019, Stem cell reports.

[7]  K. Chien,et al.  Population and Single-Cell Analysis of Human Cardiogenesis Reveals Unique LGR5 Ventricular Progenitors in Embryonic Outflow Tract. , 2019, Developmental cell.

[8]  Robert H. Anderson,et al.  Hypoplastic Left Heart Syndrome: A New Paradigm for an Old Disease? , 2019, Journal of cardiovascular development and disease.

[9]  Lu Wen,et al.  Single-Cell Transcriptome Analysis Maps the Developmental Track of the Human Heart. , 2019, Cell reports.

[10]  T. Schwarzmayr,et al.  Long-term functional and structural preservation of precision-cut human myocardium under continuous electromechanical stimulation in vitro , 2019, Nature Communications.

[11]  Guang Li,et al.  Single cell expression analysis reveals anatomical and cell cycle-dependent transcriptional shifts during heart development , 2019, Development.

[12]  W. Chung,et al.  De novo variants in congenital diaphragmatic hernia identify MYRF as a new syndrome and reveal genetic overlaps with other developmental disorders , 2018, PLoS genetics.

[13]  Kashish Chetal,et al.  Defining human cardiac transcription factor hierarchies using integrated single-cell heterogeneity analysis , 2018, Nature Communications.

[14]  S. Teichmann,et al.  Single cell RNA-seq and ATAC-seq analysis of cardiac progenitor cell transition states and lineage settlement , 2018, Nature Communications.

[15]  L. Iop,et al.  Interplay of cell–cell contacts and RhoA/MRTF‐A signaling regulates cardiomyocyte identity , 2018, The EMBO journal.

[16]  E. Zackai,et al.  De novo variants in Myelin regulatory factor (MYRF) as candidates of a new syndrome of cardiac and urogenital anomalies , 2018, American journal of medical genetics. Part A.

[17]  R. Backofen,et al.  Distinct epigenetic programs regulate cardiac myocyte development and disease in the human heart in vivo , 2017, Nature Communications.

[18]  Yufeng Shen,et al.  Contribution of rare inherited and de novo variants in 2,871 congenital heart disease probands , 2017, Nature Genetics.

[19]  D. Barron,et al.  Re-evaluation of hypoplastic left heart syndrome from a developmental and morphological perspective , 2017, Orphanet Journal of Rare Diseases.

[20]  Simon Watkins,et al.  The complex genetics of hypoplastic left heart syndrome , 2017, Nature Genetics.

[21]  Min Yu,et al.  Induced pluripotent stem cell modelling of HLHS underlines the contribution of dysfunctional NOTCH signalling to impaired cardiogenesis , 2017, Human molecular genetics.

[22]  Timothy J. Nelson,et al.  NOTCH1‐Dependent Nitric Oxide Signaling Deficiency in Hypoplastic Left Heart Syndrome Revealed Through Patient‐Specific Phenotypes Detected in Bioengineered Cardiogenesis , 2017, Stem cells.

[23]  F. Cecconi,et al.  Autophagy and the Cell Cycle: A Complex Landscape , 2017, Front. Oncol..

[24]  Beth L. Pruitt,et al.  Disease Model of GATA4 Mutation Reveals Transcription Factor Cooperativity in Human Cardiogenesis , 2016, Cell.

[25]  J. Seidman,et al.  Single-Cell Resolution of Temporal Gene Expression during Heart Development. , 2016, Developmental cell.

[26]  Mariella G. Filbin,et al.  Single-cell RNA-seq supports a developmental hierarchy in human oligodendroglioma , 2016, Nature.

[27]  Michael P Snyder,et al.  iPSC-derived cardiomyocytes reveal abnormal TGFβ signaling in left ventricular non-compaction cardiomyopathy , 2016, Nature Cell Biology.

[28]  Iuliana Ionita-Laza,et al.  De Novo Synonymous Mutations in Regulatory Elements Contribute to the Genetic Etiology of Autism and Schizophrenia , 2016, Neuron.

[29]  K. Poss,et al.  Building and re-building the heart by cardiomyocyte proliferation , 2016, Development.

[30]  Sebastian A. Leidel,et al.  A Dynamic Unfolded Protein Response Contributes to the Control of Cortical Neurogenesis. , 2015, Developmental cell.

[31]  P. Burridge,et al.  Chemically Defined Culture and Cardiomyocyte Differentiation of Human Pluripotent Stem Cells , 2015, Current protocols in human genetics.

[32]  Timothy J. Nelson,et al.  Compound heterozygous NOTCH1 mutations underlie impaired cardiogenesis in a patient with hypoplastic left heart syndrome , 2015, Human Genetics.

[33]  Jens R. Nyengaard,et al.  Dynamics of Cell Generation and Turnover in the Human Heart , 2015, Cell.

[34]  Kali T. Witherspoon,et al.  Excess of rare, inherited truncating mutations in autism , 2015, Nature Genetics.

[35]  M. Mann,et al.  Antisense-mediated exon skipping: a therapeutic strategy for titin-based dilated cardiomyopathy , 2015, EMBO molecular medicine.

[36]  A. Regev,et al.  Spatial reconstruction of single-cell gene expression , 2015, Nature Biotechnology.

[37]  Jesper V Olsen,et al.  Rapid and deep proteomes by faster sequencing on a benchtop quadrupole ultra-high-field Orbitrap mass spectrometer. , 2014, Journal of proteome research.

[38]  Matthias Mann,et al.  The Q Exactive HF, a Benchtop Mass Spectrometer with a Pre-filter, High-performance Quadrupole and an Ultra-high-field Orbitrap Analyzer* , 2014, Molecular & Cellular Proteomics.

[39]  F. Lescroart,et al.  Cardiac cell lineages that form the heart. , 2014, Cold Spring Harbor perspectives in medicine.

[40]  S. Emani,et al.  Fetal Aortic Valvuloplasty for Evolving Hypoplastic Left Heart Syndrome: Postnatal Outcomes of the First 100 Patients , 2014, Circulation.

[41]  J. Chan,et al.  Unfolded Protein Response Is Required for the Definitive Endodermal Specification of Mouse Embryonic Stem Cells via Smad2 and β-Catenin Signaling* , 2014, The Journal of Biological Chemistry.

[42]  K. Naruse,et al.  Directed Differentiation of Patient-Specific Induced Pluripotent Stem Cells Identifies the Transcriptional Repression and Epigenetic Modification of NKX2-5, HAND1, and NOTCH1 in Hypoplastic Left Heart Syndrome , 2014, PloS one.

[43]  Praveen Shukla,et al.  Chemically defined generation of human cardiomyocytes , 2014, Nature Methods.

[44]  Cole Trapnell,et al.  The dynamics and regulators of cell fate decisions are revealed by pseudotemporal ordering of single cells , 2014, Nature Biotechnology.

[45]  Cole Trapnell,et al.  Pseudo-temporal ordering of individual cells reveals dynamics and regulators of cell fate decisions , 2014, Nature Biotechnology.

[46]  E. Sernagor,et al.  An Induced Pluripotent Stem Cell Model of Hypoplastic Left Heart Syndrome (HLHS) Reveals Multiple Expression and Functional Differences in HLHS‐Derived Cardiac Myocytes , 2014, Stem cells translational medicine.

[47]  M. Mann,et al.  Minimal, encapsulated proteomic-sample processing applied to copy-number estimation in eukaryotic cells , 2014, Nature Methods.

[48]  P. Shannon,et al.  Fetal reprogramming and senescence in hypoplastic left heart syndrome and in human pluripotent stem cells during cardiac differentiation. , 2013, The American journal of pathology.

[49]  Jia Wang,et al.  Highly efficient induction and long-term maintenance of multipotent cardiovascular progenitors from human pluripotent stem cells under defined conditions , 2013, Cell Research.

[50]  Murim Choi,et al.  De novo mutations in histone modifying genes in congenital heart disease , 2013, Nature.

[51]  Jean Qiu,et al.  A novel image-based cytometry method for autophagy detection in living cells , 2012, Autophagy.

[52]  C. Hetz The unfolded protein response: controlling cell fate decisions under ER stress and beyond , 2012, Nature Reviews Molecular Cell Biology.

[53]  B. McCrindle,et al.  Left ventricular hypoplasia: a spectrum of disease involving the left ventricular outflow tract, aortic valve, and aorta. , 2012, Journal of the American College of Cardiology.

[54]  Dong Ryul Lee,et al.  Screening ethnically diverse human embryonic stem cells identifies a chromosome 20 minimal amplicon conferring growth advantage , 2011, Nature Biotechnology.

[55]  M. Newton,et al.  Karyotypic abnormalities in human induced pluripotent stem cells and embryonic stem cells , 2011, Nature Biotechnology.

[56]  Elaine Fuchs,et al.  Skin Stem Cells Orchestrate Directional Migration by Regulating Microtubule-ACF7 Connections through GSK3β , 2011, Cell.

[57]  M. Seyfarth,et al.  Patient-specific induced pluripotent stem-cell models for long-QT syndrome. , 2010 .

[58]  C. Sander,et al.  Automated Network Analysis Identifies Core Pathways in Glioblastoma , 2010, PloS one.

[59]  Chengji J. Zhou,et al.  Cardiac neural crest and outflow tract defects in Lrp6 mutant mice , 2009, Developmental dynamics : an official publication of the American Association of Anatomists.

[60]  H. Boyd,et al.  Recurrence of Congenital Heart Defects in Families , 2009, Circulation.

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

[62]  M. Mann,et al.  MaxQuant enables high peptide identification rates, individualized p.p.b.-range mass accuracies and proteome-wide protein quantification , 2008, Nature Biotechnology.

[63]  C. Chu,et al.  Mitochondrially localized ERK2 regulates mitophagy and autophagic cell stress , 2008, Autophagy.

[64]  K. Chien,et al.  Islet1 cardiovascular progenitors: a single source for heart lineages? , 2007, Development.

[65]  Lisa J. Martin,et al.  Hypoplastic left heart syndrome is heritable. , 2007, Journal of the American College of Cardiology.

[66]  Yunfu Sun,et al.  Multipotent Embryonic Isl1 + Progenitor Cells Lead to Cardiac, Smooth Muscle, and Endothelial Cell Diversification , 2006, Cell.

[67]  S. Kattman,et al.  Multipotent flk-1+ cardiovascular progenitor cells give rise to the cardiomyocyte, endothelial, and vascular smooth muscle lineages. , 2006, Developmental cell.

[68]  Amy S. Lee,et al.  In Vivo Regulation of Grp78/BiP Transcription in the Embryonic Heart , 2006, Journal of Biological Chemistry.

[69]  Matthias E. Futschik,et al.  Noise-robust Soft Clustering of Gene Expression Time-course Data , 2005, J. Bioinform. Comput. Biol..

[70]  F. Giostra,et al.  Methods and Results , 2014 .

[71]  T. Mikawa,et al.  TBX5 transcription factor regulates cell proliferation during cardiogenesis. , 2001, Developmental biology.

[72]  S. Jackson,et al.  Regulation of p53 in response to DNA damage , 1999, Oncogene.

[73]  J. Lippincott-Schwartz,et al.  Brefeldin A: insights into the control of membrane traffic and organelle structure , 1992, The Journal of cell biology.

[74]  A. Nadas,et al.  The hypoplastic left heart syndrome; an analysis of 101 cases. , 1958, Pediatric clinics of North America.

[75]  R. Clark,et al.  Autophagy in neurite injury and neurodegeneration: in vitro and in vivo models. , 2009, Methods in enzymology.

[76]  M H Paul,et al.  Experimental production of hypoplastic left heart syndrome in the chick embryo. , 1973, The American journal of cardiology.

[77]  M. Lev Pathologic anatomy and interrelationship of hypoplasia of the aortic tract complexes. , 1952, Laboratory investigation; a journal of technical methods and pathology.