A Spatiotemporal Organ-Wide Gene Expression and Cell Atlas of the Developing Human Heart

The process of cardiac morphogenesis in humans is incompletely understood. Its full characterization requires a deep exploration of the organ-wide orchestration of gene expression with a single-cell spatial resolution. Here, we present a molecular approach that reveals the comprehensive transcriptional landscape of cell types populating the embryonic heart at three developmental stages and that maps cell-type-specific gene expression to specific anatomical domains. Spatial transcriptomics identified unique gene profiles that correspond to distinct anatomical regions in each developmental stage. Human embryonic cardiac cell types identified by single-cell RNA sequencing confirmed and enriched the spatial annotation of embryonic cardiac gene expression. In situ sequencing was then used to refine these results and create a spatial subcellular map for the three developmental phases. Finally, we generated a publicly available web resource of the human developing heart to facilitate future studies on human cardiogenesis.

[1]  John A Wolf,et al.  Transcriptome In Vivo Analysis (TIVA) of spatially defined single cells in intact live mouse and human brain tissue , 2014, Nature Methods.

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

[3]  A. Moorman,et al.  Development of the human heart , 2014, American journal of medical genetics. Part A.

[4]  U. Dräger,et al.  Dynamic patterns of retinoic acid synthesis and response in the developing mammalian heart. , 1998, Developmental biology.

[5]  Guo-Cheng Yuan,et al.  Transcriptome-scale super-resolved imaging in tissues by RNA seqFISH+ , 2019, Nature.

[6]  A. Oudenaarden,et al.  Genome-wide RNA Tomography in the Zebrafish Embryo , 2014, Cell.

[7]  Erik L. G. Wernersson,et al.  An Application-Directed, Versatile DNA FISH Platform for Research and Diagnostics. , 2018, Methods in molecular biology.

[8]  I. Shepherd,et al.  Characterization of spatial and temporal expression pattern of SCG10 during zebrafish development. , 2009, Gene expression patterns : GEP.

[9]  Patrik L. Ståhl,et al.  Visualization and analysis of gene expression in tissue sections by spatial transcriptomics , 2016, Science.

[10]  E. Arenas,et al.  Wnt/β-Catenin Stimulation and Laminins Support Cardiovascular Cell Progenitor Expansion from Human Fetal Cardiac Mesenchymal Stromal Cells , 2016, Stem cell reports.

[11]  Christopher S. McGinnis,et al.  DoubletFinder: Doublet detection in single-cell RNA sequencing data using artificial nearest neighbors , 2018, bioRxiv.

[12]  M. Buckingham,et al.  The deployment of cell lineages that form the mammalian heart , 2018, Nature Reviews Cardiology.

[13]  X. Zhuang,et al.  RNA Imaging with Multiplexed Error-Robust Fluorescence In Situ Hybridization (MERFISH). , 2016, Methods in enzymology.

[14]  E Jansson,et al.  Myoglobin content and citrate synthase activity in different parts of the normal human heart. , 1990, Journal of applied physiology.

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

[16]  Nikolaus Rajewsky,et al.  The Drosophila embryo at single-cell transcriptome resolution , 2017, Science.

[17]  David J. Anderson,et al.  Molecular probes for the development and plasticity of neural crest derivatives , 1985, Cell.

[18]  Patrik L. Ståhl,et al.  Spatial detection of fetal marker genes expressed at low level in adult human heart tissue , 2017, Scientific Reports.

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

[20]  Erik L. G. Wernersson,et al.  iFISH is a publically available resource enabling versatile DNA FISH to study genome architecture , 2019, Nature Communications.

[21]  A. van Oudenaarden,et al.  Single-Cell Sequencing of the Healthy and Diseased Heart Reveals Cytoskeleton-Associated Protein 4 as a New Modulator of Fibroblasts Activation , 2018, Circulation.

[22]  B. Göttgens,et al.  Defining the earliest step of cardiovascular lineage segregation by single-cell RNA-seq , 2018, Science.

[23]  Catherine E. Braine,et al.  Spatiotemporal dynamics of molecular pathology in amyotrophic lateral sclerosis , 2018, Science.

[24]  Fabian J Theis,et al.  The Human Cell Atlas , 2017, bioRxiv.

[25]  Monika S. Kowalczyk,et al.  Single-cell RNA-seq reveals changes in cell cycle and differentiation programs upon aging of hematopoietic stem cells , 2015, Genome research.

[26]  Guangchuang Yu,et al.  clusterProfiler: an R package for comparing biological themes among gene clusters. , 2012, Omics : a journal of integrative biology.

[27]  S. Shete,et al.  Myozenin 2 Is a Novel Gene for Human Hypertrophic Cardiomyopathy , 2007, Circulation research.

[28]  X. Zhuang,et al.  Spatially resolved, highly multiplexed RNA profiling in single cells , 2015, Science.

[29]  J. Epstein,et al.  Brief Ultrarapid Communication Islet1 Derivatives in the Heart Are of Both Neural Crest and Second Heart Field Origin Methods and Results: We Used an Intersectional Fate-mapping System Using the Rc::frepe Allele, Which Reports Dual Flpe and Cre Recombination. Combining Isl1 , 2022 .

[30]  Paul Theodor Pyl,et al.  HTSeq—a Python framework to work with high-throughput sequencing data , 2014, bioRxiv.

[31]  Kenneth D. Harris,et al.  Probabilistic cell typing enables fine mapping of closely related cell types in situ , 2019, Nature Methods.

[32]  Evan Z. Macosko,et al.  Slide-seq: A scalable technology for measuring genome-wide expression at high spatial resolution , 2019, Science.

[33]  George M. Church,et al.  Highly Multiplexed Subcellular RNA Sequencing in Situ , 2014, Science.

[34]  J. D. Engel,et al.  Differential regulation of transcription factor gene expression and phenotypic markers in developing sympathetic neurons. , 1995, Development.

[35]  Karl Deisseroth,et al.  An interactive framework for whole-brain maps at cellular resolution , 2017, Nature Neuroscience.

[36]  Joakim Lundeberg,et al.  TagGD: Fast and Accurate Software for DNA Tag Generation and Demultiplexing , 2013, PloS one.

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

[38]  M. Majesky,et al.  Tbx18 regulates development of the epicardium and coronary vessels. , 2013, Developmental biology.

[39]  Carolina Wählby,et al.  In situ sequencing for RNA analysis in preserved tissue and cells , 2013, Nature Methods.

[40]  Patrik L. Ståhl,et al.  Barcoded solid-phase RNA capture for Spatial Transcriptomics profiling in mammalian tissue sections , 2018, Nature Protocols.

[41]  M. Jahangiri,et al.  Extracellular Matrix Proteomics Reveals Interplay of Aggrecan and Aggrecanases in Vascular Remodeling of Stented Coronary Arteries , 2018, Circulation.

[42]  P. Ray,et al.  XIST expression from the maternal X chromosome in human male preimplantation embryos at the blastocyst stage. , 1997, Human molecular genetics.

[43]  Grace X. Y. Zheng,et al.  Massively parallel digital transcriptional profiling of single cells , 2016, Nature Communications.

[44]  I. Yanai,et al.  Building a tumor atlas: integrating single-cell RNA-Seq data with spatial transcriptomics in pancreatic ductal adenocarcinoma , 2018, bioRxiv.

[45]  J. Marioni,et al.  High-throughput spatial mapping of single-cell RNA-seq data to tissue of origin , 2015, Nature Biotechnology.

[46]  J. Lundeberg,et al.  Preparation of plant tissue to enable Spatial Transcriptomics profiling using barcoded microarrays , 2018, Nature Protocols.

[47]  A. Keyte,et al.  Evolutionary and developmental origins of the cardiac neural crest: building a divided outflow tract. , 2014, Birth defects research. Part C, Embryo today : reviews.

[48]  Richard Bonneau,et al.  High-definition spatial transcriptomics for in situ tissue profiling , 2019, Nature Methods.

[49]  Joel Sjöstrand,et al.  ST Pipeline: an automated pipeline for spatial mapping of unique transcripts , 2017, Bioinform..

[50]  Ju Chen,et al.  A myocardial lineage derives from Tbx18 epicardial cells , 2008, Nature.

[51]  J. Lundeberg,et al.  Gene expression profiling of periodontitis-affected gingival tissue by spatial transcriptomics , 2018, Scientific Reports.

[52]  Antoon F. M. Moorman,et al.  An interactive three-dimensional digital atlas and quantitative database of human development , 2016, Science.

[53]  R. Mirsky,et al.  Schwann Cell Precursors; Multipotent Glial Cells in Embryonic Nerves , 2019, Front. Mol. Neurosci..

[54]  K. Yutzey,et al.  Transcriptional Control of Cell Lineage Development in Epicardium-Derived Cells , 2013, Journal of developmental biology.

[55]  I. Yanai,et al.  Integrating single-cell RNA-Seq with spatial transcriptomics in pancreatic ductal adenocarcinoma using multimodal intersection analysis , 2019 .

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

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

[58]  Andrey Alexeyenko,et al.  Spatially resolved transcriptome profiling in model plant species , 2017, Nature Plants.

[59]  P. Riley,et al.  The epicardium signals the way towards heart regeneration , 2014, Stem cell research.

[60]  I. Adameyko,et al.  Nerve-associated neural crest: peripheral glial cells generate multiple fates in the body. , 2017, Current opinion in genetics & development.

[61]  Fred L. Bookstein,et al.  Principal Warps: Thin-Plate Splines and the Decomposition of Deformations , 1989, IEEE Trans. Pattern Anal. Mach. Intell..

[62]  William E. Allen,et al.  Three-dimensional intact-tissue sequencing of single-cell transcriptional states , 2018, Science.