PRDM16 Is a Compact Myocardium-Enriched Transcription Factor Required to Maintain Compact Myocardial Cardiomyocyte Identity in Left Ventricle

Supplemental Digital Content is available in the text. Background: Left ventricular noncompaction cardiomyopathy (LVNC) was discovered half a century ago as a cardiomyopathy with excessive trabeculation and a thin ventricular wall. In the decades since, numerous studies have demonstrated that LVNC primarily has an effect on left ventricles (LVs) and is often associated with LV dilation and dysfunction. However, in part because of the lack of suitable mouse models that faithfully mirror the selective LV vulnerability in patients, mechanisms underlying the susceptibility of LVs to dilation and dysfunction in LVNC remain unknown. Genetic studies have revealed that deletions and mutations in PRDM16 (PR domain-containing 16) cause LVNC, but previous conditional Prdm16 knockout mouse models do not mirror the LVNC phenotype in patients, and the underlying molecular mechanisms by which PRDM16 deficiency causes LVNC are still unclear. Methods: Prdm16 cardiomyocyte–specific knockout (Prdm16cKO) mice were generated and analyzed for cardiac phenotypes. RNA sequencing and chromatin immunoprecipitation deep sequencing were performed to identify direct transcriptional targets of PRDM16 in cardiomyocytes. Single-cell RNA sequencing in combination with spatial transcriptomics was used to determine cardiomyocyte identity at the single-cell level. Results: Cardiomyocyte-specific ablation of Prdm16 in mice caused LV-specific dilation and dysfunction, as well as biventricular noncompaction, which fully recapitulated LVNC in patients. PRDM16 functioned mechanistically as a compact myocardium-enriched transcription factor that activated compact myocardial genes while repressing trabecular myocardial genes in LV compact myocardium. Consequently, Prdm16cKO LV compact myocardial cardiomyocytes shifted from their normal transcriptomic identity to a transcriptional signature resembling trabecular myocardial cardiomyocytes or neurons. Chamber-specific transcriptional regulation by PRDM16 was attributable in part to its cooperation with LV-enriched transcription factors Tbx5 and Hand1. Conclusions: These results demonstrate that disruption of proper specification of compact cardiomyocytes may play a key role in the pathogenesis of LVNC. They also shed light on underlying mechanisms of the LV-restricted transcriptional program governing LV chamber growth and maturation, providing a tangible explanation for the susceptibility of LV in a subset of LVNC cardiomyopathies.

[1]  P. Seale,et al.  Prdm16 Deficiency Leads to Age-Dependent Cardiac Hypertrophy, Adverse Remodeling, Mitochondrial Dysfunction, and Heart Failure. , 2020, Cell reports.

[2]  Howard Y. Chang,et al.  Single-cell RNA sequencing in cardiovascular development, disease and medicine , 2020, Nature Reviews Cardiology.

[3]  B. Oh,et al.  Cardiac-specific inactivation of Prdm16 effects cardiac conduction abnormalities and cardiomyopathy-associated phenotypes. , 2020, American journal of physiology. Heart and circulatory physiology.

[4]  M. Novák Position , 2020, Introduction to Sensors for Electrical and Mechanical Engineers.

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

[6]  Guocheng Yuan,et al.  A reference map of murine cardiac transcription factor chromatin occupancy identifies dynamic and conserved enhancers , 2019, Nature Communications.

[7]  G. Perkins,et al.  Nexilin is a New Component of Junctional Membrane Complexes Required for Cardiac T-Tubule Formation. , 2019, Circulation.

[8]  P. Khaitovich,et al.  Gene expression across mammalian organ development , 2019, Nature.

[9]  R. Hofstra,et al.  Cardiac Phenotypes, Genetics, and Risks in Familial Noncompaction Cardiomyopathy. , 2019, Journal of the American College of Cardiology.

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

[11]  Mauro W. Costa,et al.  Deletion of Nkx2-5 in trabecular myocardium reveals the developmental origins of pathological heterogeneity associated with ventricular non-compaction cardiomyopathy , 2018, PLoS genetics.

[12]  Matthew B. Johnson,et al.  The Epigenetic State of PRDM16-Regulated Enhancers in Radial Glia Controls Cortical Neuron Position , 2018, Neuron.

[13]  W. Shou,et al.  Potential Common Pathogenic Pathways for the Left Ventricular Noncompaction Cardiomyopathy (LVNC) , 2018, Pediatric Cardiology.

[14]  J. Li,et al.  Neddylation mediates ventricular chamber maturation through repression of Hippo signaling , 2018, Proceedings of the National Academy of Sciences.

[15]  J. Michael Cherry,et al.  The Encyclopedia of DNA elements (ENCODE): data portal update , 2017, Nucleic Acids Res..

[16]  Roberta B. Nowak,et al.  HSPB7 is indispensable for heart development by modulating actin filament assembly , 2017, Proceedings of the National Academy of Sciences.

[17]  Jared Evans,et al.  Diagnostic Yield of Whole Exome Sequencing in Pediatric Dilated Cardiomyopathy , 2017, Journal of cardiovascular development and disease.

[18]  Robert H. Anderson,et al.  Identification of a hybrid myocardial zone in the mammalian heart after birth , 2017, Nature Communications.

[19]  S. Morrison,et al.  Prdm16 is required for the maintenance of neural stem cells in the postnatal forebrain and their differentiation into ependymal cells , 2017, Genes & development.

[20]  Robert H. Anderson,et al.  Key Questions Relating to Left Ventricular Noncompaction Cardiomyopathy: Is the Emperor Still Wearing Any Clothes? , 2017, The Canadian journal of cardiology.

[21]  J. Aerts,et al.  SCENIC: Single-cell regulatory network inference and clustering , 2017, Nature Methods.

[22]  Claudia Stöllberger,et al.  Left ventricular noncompaction cardiomyopathy: cardiac, neuromuscular, and genetic factors , 2017, Nature Reviews Cardiology.

[23]  Ju Chen,et al.  Adaptor proteins NUMB and NUMBL promote cell cycle withdrawal by targeting ERBB2 for degradation , 2017, The Journal of clinical investigation.

[24]  Bin Zhou,et al.  Transcriptomic Profiling Maps Anatomically Patterned Subpopulations among Single Embryonic Cardiac Cells. , 2016, Developmental cell.

[25]  D. Andreini,et al.  Long-Term Prognostic Value of Cardiac Magnetic Resonance in Left Ventricle Noncompaction: A Prospective Multicenter Study. , 2016, Journal of the American College of Cardiology.

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

[27]  Md. Abul Hassan Samee,et al.  Complex Interdependence Regulates Heterotypic Transcription Factor Distribution and Coordinates Cardiogenesis , 2016, Cell.

[28]  P. Cohen,et al.  The Multifaceted Roles of PRDM16: Adipose Biology and Beyond , 2016, Trends in Endocrinology & Metabolism.

[29]  Rick B. Vega,et al.  Mitochondrial biogenesis and dynamics in the developing and diseased heart , 2015, Genes & development.

[30]  B. Gerber,et al.  Prognostic Impact of Hypertrabeculation and Noncompaction Phenotype in Dilated Cardiomyopathy: A CMR Study. , 2015, JACC. Cardiovascular imaging.

[31]  A. McCulloch,et al.  HIF1α Represses Cell Stress Pathways to Allow Proliferation of Hypoxic Fetal Cardiomyocytes. , 2015, Developmental cell.

[32]  Kyoung-Jae Won,et al.  PRDM16 binds MED1 and controls chromatin architecture to determine a brown fat transcriptional program , 2015, Genes & development.

[33]  Axel Visel,et al.  Dynamic GATA4 enhancers shape the chromatin landscape central to heart development and disease , 2014, Nature Communications.

[34]  Y. Hsueh,et al.  Cortactin-binding protein 2 increases microtubule stability and regulates dendritic arborization , 2014, Journal of Cell Science.

[35]  W. de Laat,et al.  A Large Permissive Regulatory Domain Exclusively Controls Tbx3 Expression in the Cardiac Conduction System , 2014, Circulation research.

[36]  Alexander S. Banks,et al.  Ablation of PRDM16 and Beige Adipose Causes Metabolic Dysfunction and a Subcutaneous to Visceral Fat Switch , 2014, Cell.

[37]  G. Musso,et al.  Fine mapping of the 1p36 deletion syndrome identifies mutation of PRDM16 as a cause of cardiomyopathy. , 2013, American journal of human genetics.

[38]  F. Conlon,et al.  Tcf21 regulates the specification and maturation of proepicardial cells , 2013, Development.

[39]  B. Bruneau Signaling and transcriptional networks in heart development and regeneration. , 2013, Cold Spring Harbor perspectives in biology.

[40]  Carlos Torroja,et al.  Mutations in the NOTCH pathway regulator MIB1 cause left ventricular noncompaction cardiomyopathy , 2013, Nature Medicine.

[41]  Tobias Hohenauer,et al.  The Prdm family: expanding roles in stem cells and development , 2012, Development.

[42]  M. Bulyk,et al.  Dual transcriptional activator and repressor roles of TBX20 regulate adult cardiac structure and function. , 2012, Human molecular genetics.

[43]  R. Sandberg,et al.  Sequentially acting Sox transcription factors in neural lineage development. , 2011, Genes & development.

[44]  I. Lemischka,et al.  Prdm16 is a physiologic regulator of hematopoietic stem cells. , 2011, Blood.

[45]  Joshua W. Vincentz,et al.  Analysis of the Hand1 cell lineage reveals novel contributions to cardiovascular, neural crest, extra‐embryonic, and lateral mesoderm derivatives , 2010, Developmental dynamics : an official publication of the American Association of Anatomists.

[46]  C. Glass,et al.  Simple combinations of lineage-determining transcription factors prime cis-regulatory elements required for macrophage and B cell identities. , 2010, Molecular cell.

[47]  B. Bjork,et al.  Prdm16 is required for normal palatogenesis in mice. , 2010, Human molecular genetics.

[48]  Allan R. Jones,et al.  A robust and high-throughput Cre reporting and characterization system for the whole mouse brain , 2009, Nature Neuroscience.

[49]  C. Çevik,et al.  Biventricular Noncompaction: A Case Report , 2008, Echocardiography.

[50]  B. Spiegelman,et al.  PRDM16 controls a brown fat/skeletal muscle switch , 2008, Nature.

[51]  Bastiaan J. Boukens,et al.  Transcription Factor Tbx3 Is Required for the Specification of the Atrioventricular Conduction System , 2008, Circulation research.

[52]  W. McKenna,et al.  Left ventricular noncompaction and cardiomyopathy: cause, contributor, or epiphenomenon? , 2008, Current opinion in cardiology.

[53]  R. Markwald,et al.  Periostin Is Required for Maturation and Extracellular Matrix Stabilization of Noncardiomyocyte Lineages of the Heart , 2008, Circulation research.

[54]  B. Spiegelman,et al.  Transcriptional control of brown fat determination by PRDM16. , 2007, Cell metabolism.

[55]  D. Srivastava,et al.  Essential roles of the bHLH transcription factor Hrt2 in repression of atrial gene expression and maintenance of postnatal cardiac function , 2007, Proceedings of the National Academy of Sciences.

[56]  M. Bennett,et al.  Pan‐myocardial expression of Cre recombinase throughout mouse development , 2007, Genesis.

[57]  M. Chin,et al.  Abstract 1238: CHF1/Hey2 Plays a Pivotal Role in Left Ventricular Maturation through Suppression of Ectopic Atrial Gene Expression , 2006 .

[58]  Michael D. Schneider,et al.  Cardiomyocyte-Specific Deletion of the Coxsackievirus and Adenovirus Receptor Results in Hyperplasia of the Embryonic Left Ventricle and Abnormalities of Sinuatrial Valves , 2006, Circulation research.

[59]  Barry J Maron,et al.  Contemporary definitions and classification of the cardiomyopathies: an American Heart Association Scientific Statement from the Council on Clinical Cardiology, Heart Failure and Transplantation Committee; Quality of Care and Outcomes Research and Functional Genomics and Translational Biology Interd , 2006, Circulation.

[60]  J. Belmont,et al.  Clinical Characterization of Left Ventricular Noncompaction in Children: A Relatively Common Form of Cardiomyopathy , 2003, Circulation.

[61]  Takuro Nakamura,et al.  A novel EVI1 gene family, MEL1, lacking a PR domain (MEL1S) is expressed mainly in t(1;3)(p36;q21)-positive AML and blocks G-CSF-induced myeloid differentiation. , 2003, Blood.

[62]  B. Hogan,et al.  An essential role of Bmp4 in the atrioventricular septation of the mouse heart. , 2003, Genes & development.

[63]  A. Moorman,et al.  Cardiac chamber formation: development, genes, and evolution. , 2003, Physiological reviews.

[64]  John B Carlin,et al.  The epidemiology of childhood cardiomyopathy in Australia. , 2003, The New England journal of medicine.

[65]  M. Crackower,et al.  Temporally Regulated and Tissue-Specific Gene Manipulations in the Adult and Embryonic Heart Using a Tamoxifen-Inducible Cre Protein , 2001, Circulation research.

[66]  Robert H. Anderson,et al.  Developmental patterning of the myocardium , 2000, The Anatomical record.

[67]  J. Seidman,et al.  Chamber-specific cardiac expression of Tbx5 and heart defects in Holt-Oram syndrome. , 1999, Developmental biology.

[68]  J. Rossant,et al.  Defects in heart and lung development in compound heterozygotes for two different targeted mutations at the N-myc locus. , 1993, Development.

[69]  R. Arcilla,et al.  Ventricular trabeculations in the chick embryo heart and their contribution to ventricular and muscular septal development. , 1985, Circulation research.

[70]  G. Aquaro,et al.  Magnetic resonance assessment of prevalence and correlates of right ventricular abnormalities in isolated left ventricular noncompaction. , 2014, The American journal of cardiology.

[71]  W. Shou,et al.  Myocardial Mycn is essential for mouse ventricular wall morphogenesis. , 2013, Developmental biology.

[72]  H. Kok,et al.  Left ventricular non-compaction cardiomyopathy. , 2012, JBR-BTR : organe de la Societe royale belge de radiologie (SRBR) = orgaan van de Koninklijke Belgische Vereniging voor Radiologie.

[73]  David J. Miller,et al.  Gene Regulatory Networks in the Evolution and Development of the Heart , 2006 .