Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Article
  • Published:

Integrated multi-omic characterization of congenital heart disease

Abstract

The heart, the first organ to develop in the embryo, undergoes complex morphogenesis that when defective results in congenital heart disease (CHD). With current therapies, more than 90% of patients with CHD survive into adulthood, but many suffer premature death from heart failure and non-cardiac causes1. Here, to gain insight into this disease progression, we performed single-nucleus RNA sequencing on 157,273 nuclei from control hearts and hearts from patients with CHD, including those with hypoplastic left heart syndrome (HLHS) and tetralogy of Fallot, two common forms of cyanotic CHD lesions, as well as dilated and hypertrophic cardiomyopathies. We observed CHD-specific cell states in cardiomyocytes, which showed evidence of insulin resistance and increased expression of genes associated with FOXO signalling and CRIM1. Cardiac fibroblasts in HLHS were enriched in a low-Hippo and high-YAP cell state characteristic of activated cardiac fibroblasts. Imaging mass cytometry uncovered a spatially resolved perivascular microenvironment consistent with an immunodeficient state in CHD. Peripheral immune cell profiling suggested deficient monocytic immunity in CHD, in agreement with the predilection in CHD to infection and cancer2. Our comprehensive phenotyping of CHD provides a roadmap towards future personalized treatments for CHD.

This is a preview of subscription content, access via your institution

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: Profiling of tissues from paediatric controls and patients with CHD or heart failure.
Fig. 2: snRNA-seq showing the unique transcriptional signature of cardiomyocytes in paediatric patients with CHD.
Fig. 3: Profiling of cardiac fibroblasts in paediatric patients.
Fig. 4: Tissue histology and validation of tissue snRNA-seq results across paediatric cardiac diseases.
Fig. 5: High-dimensional histopathology of paediatric cardiac diseases.
Fig. 6: Intercellular communication in paediatric cardiovascular disease.

Similar content being viewed by others

Data availability

Raw and processed next-generation sequencing data have been deposited at the NCBI Gene Expression Omnibus under accession number GSE203275. The snRNA-seq data are available online at the Broad Single Cell Portal under study number SCP1852.

Code availability

The pseduobulk RNA-seq analysis script we used is available at https://hbctraining.github.io/scRNA-seq/lessons/pseudobulk_DESeq2_scrnaseq.html.

References

  1. Raissadati, A., Nieminen, H., Jokinen, E. & Sairanen, H. Progress in late results among pediatric cardiac surgery patients: a population-based 6-decade study with 98% follow-up. Circulation 131, 347–353 (2015).

    Article  PubMed  Google Scholar 

  2. Diller, G. P. et al. Survival prospects and circumstances of death in contemporary adult congenital heart disease patients under follow-up at a large tertiary centre. Circulation 132, 2118–2125 (2015).

    Article  PubMed  Google Scholar 

  3. Hsu, D. T. & Pearson, G. D. Heart failure in children: part II: diagnosis, treatment, and future directions. Circ. Heart. Fail. 2, 490–498 (2009).

    Article  PubMed  Google Scholar 

  4. Friedberg, M. K. & Reddy, S. Right ventricular failure in congenital heart disease. Curr. Opin. Pediatr. 31, 604–610 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  5. Gurvitz, M. et al. Emerging research directions in adult congenital heart disease: A report from an NHLBI/ACHA working group. J. Am. Coll. Cardiol. 67, 1956–1964 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  6. Ntiloudi, D. et al. Adult congenital heart disease: a paradigm of epidemiological change. Int. J. Cardiol. 218, 269–274 (2016).

    Article  PubMed  Google Scholar 

  7. Fontan, F. et al. Outcome after a “perfect” Fontan operation. Circulation 81, 1520–1536 (1990).

    Article  CAS  PubMed  Google Scholar 

  8. Liu, X. et al. The complex genetics of hypoplastic left heart syndrome. Nat. Genet. 49, 1152–1159 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Brisson, A. R., Matsui, D., Rieder, M. J. & Fraser, D. D. Translational research in pediatrics: tissue sampling and biobanking. Pediatrics 129, 153–162 (2012).

    Article  PubMed  Google Scholar 

  10. Sim, C. B. et al. Sex-specific control of human heart maturation by the progesterone receptor. Circulation 143, 1614–1628 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Litviňuková, M. et al. Cells of the adult human heart. Nature 588, 466–472 (2020).

    Article  ADS  PubMed  PubMed Central  CAS  Google Scholar 

  12. Tucker, N. R. et al. Transcriptional and cellular diversity of the human heart. Circulation 142, 466–482 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Dann, E., Henderson, N. C., Teichmamnn, S. A., Morgan, M. D. & Marioni, J. C. Differential abundance testing on single-cell data using k-nearest neighbor graphs. Nat. Biotechnol. 40, 245–253 (2022).

    Article  CAS  PubMed  Google Scholar 

  14. Liu, H. et al. Control of cytokinesis by β-adrenergic receptors indicates an approach for regulating cardiomyocyte endowment. Sci. Transl. Med. 11, eaaw6419 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. van Duijvenboden, K. et al. Conserved NPPB+ border zone switches from MEF2- to AP-1-driven gene program. Circulation 140, 864–879 (2019).

    Article  PubMed  CAS  Google Scholar 

  16. Lee, R. et al. Regulated inositol-requiring protein 1-dependent decay as a mechanism of corin RNA and protein deficiency in advanced human systolic heart failure. J. Am. Heart Assoc. 3, e001104 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  17. Goetze, J. P. et al. Cardiac natriuretic peptides. Nat. Rev. Cardiol. 17, 698–717 (2020).

    Article  CAS  PubMed  Google Scholar 

  18. Yang, S. F., Chou, R. H., Li, S. Y., Huang, S. S. & Huang, P. H. Serum corin level is associated with subsequent decline in renal function in patients with suspected coronary artery disease. J. Am. Heart Assoc. 7, e008157 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  19. Eleuteri, E. et al. Fibrosis markers and CRIM1 increase in chronic heart failure of increasing severity. Biomarkers 19, 214–221 (2014).

    Article  CAS  PubMed  Google Scholar 

  20. Beckman, E. J. Management of the pediatric organ donor. J. Pediatr. Pharmacol. Ther. 24, 276–289 (2019).

    MathSciNet  PubMed  PubMed Central  Google Scholar 

  21. Tallquist, M. D. & Molkentin, J. D. Redefining the identity of cardiac fibroblasts. Nat. Rev. Cardiol. 14, 484–491 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  22. Corbel, M. et al. Inhibition of bleomycin-induced pulmonary fibrosis in mice by the matrix metalloproteinase inhibitor batimastat. J. Pathol. 193, 538–545 (2001).

    Article  CAS  PubMed  Google Scholar 

  23. Nguyen, X. X., Muhammad, L., Nietert, P. J. & Feghali-Bostwick, C. IGFBP-5 promotes fibrosis via increasing its own expression and that of other pro-fibrotic mediators. Front. Endocrinol. 9, 601 (2018).

    Article  Google Scholar 

  24. Numaga-Tomita, T. et al. TRPC3–GEF–H1 axis mediates pressure overload-induced cardiac fibrosis. Sci. Rep. 6, 39383 (2016).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  25. Xiao, Y. et al. Hippo pathway deletion in adult resting cardiac fibroblasts initiates a cell state transition with spontaneous and self-sustaining fibrosis. Genes Dev. 33, 1491–1505 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Long, F. et al. Targeting JMJD3 histone demethylase mediates cardiac fibrosis and cardiac function following myocardial infarction. Biochem. Biophys. Res. Commun. 528, 671–677 (2020).

    Article  CAS  PubMed  Google Scholar 

  27. Kalucka, J. et al. Single-cell transcriptome atlas of murine endothelial cells. Cell 180, 764–779.e720 (2020).

    Article  CAS  PubMed  Google Scholar 

  28. Su, T. et al. Single-cell analysis of early progenitor cells that build coronary arteries. Nature 559, 356–362 (2018).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  29. Thiriot, A. et al. Differential DARC/ACKR1 expression distinguishes venular from non-venular endothelial cells in murine tissues. BMC Biol. 15, 45 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  30. Vanlandewijck, M. et al. A molecular atlas of cell types and zonation in the brain vasculature. Nature 554, 475–480 (2018).

    Article  ADS  CAS  PubMed  Google Scholar 

  31. Jiang, X. et al. Elevated lymphatic vessel density measured by Lyve-1 expression in areas of replacement fibrosis in the ventricular septum of patients with hypertrophic obstructive cardiomyopathy (HOCM). Heart Vessels 35, 78–85 (2020).

    Article  PubMed  Google Scholar 

  32. Bertero, E., Kutschka, I., Maack, C. & Dudek, J. Cardiolipin remodeling in Barth syndrome and other hereditary cardiomyopathies. Biochim. Biophys. Acta 1866, 165803 (2020).

    Article  CAS  Google Scholar 

  33. Salehi, A. H., Xanthoudakis, S. & Barker, P. A. NRAGE, a p75 neurotrophin receptor-interacting protein, induces caspase activation and cell death through a JNK-dependent mitochondrial pathway. J. Biol. Chem. 277, 48043–48050 (2002).

    Article  CAS  PubMed  Google Scholar 

  34. Nishimura, H. et al. Autoimmune dilated cardiomyopathy in PD-1 receptor-deficient mice. Science 291, 319–322 (2001).

    Article  ADS  CAS  PubMed  Google Scholar 

  35. Zhang, B. CD73: a novel target for cancer immunotherapy. Cancer Res. 70, 6407–6411 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Hyun, S. Y. et al. Immunosuppressive role of CD11b+ CD33+ HLA-DR myeloid-derived suppressor cells-like blast subpopulation in acute myeloid leukemia. Cancer Med. 9, 7007–7017 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Afanasyeva, M. et al. Impaired up-regulation of CD25 on CD4+ T cells in IFN-γ knockout mice is associated with progression of myocarditis to heart failure. Proc. Natl Acad. Sci. USA 102, 180–185 (2005).

    Article  ADS  CAS  PubMed  Google Scholar 

  38. Grabie, N., Lichtman, A. H. & Padera, R. T cell checkpoint regulators in the heart. Cardiovasc. Res. 115, 869–877 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Koenig, A. L. et al. Single-cell transcriptomics reveals cell-type-specific diversification in human heart failure. Nat. Cardiovasc. Res. 1, 263–280 (2022).

    Article  Google Scholar 

  40. Caron, M. et al. Single-cell analysis of childhood leukemia reveals a link between developmental states and ribosomal protein expression as a source of intra-individual heterogeneity. Sci Rep. 10, 8079 (2020).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  41. Zheng, G. X. et al. Massively parallel digital transcriptional profiling of single cells. Nat. Commun. 8, 14049 (2017).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  42. Crozat, K. et al. The XC chemokine receptor 1 is a conserved selective marker of mammalian cells homologous to mouse CD8α+ dendritic cells. J. Exp. Med. 207, 1283–1292 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Vanhoutte, D. et al. Thbs1 induces lethal cardiac atrophy through PERK-ATF4 regulated autophagy. Nat. Commun. 12, 3928 (2021).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  44. Jin, S. et al. Inference and analysis of cell-cell communication using CellChat. Nat. Commun. 12, 1088 (2021).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  45. Blair, C. A. et al. A protocol for collecting human cardiac tissue for research. VAD J. 2, 12 (2016).

    Google Scholar 

  46. Corces, M. R. et al. An improved ATAC-seq protocol reduces background and enables interrogation of frozen tissues. Nat. Methods 14, 959–962 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Hill, M. C. & Martin, J. F. Epigenetic assays in purified cardiomyocyte nuclei. Methods Mol. Biol. 2158, 307–321 (2021).

    Article  CAS  PubMed  Google Scholar 

  48. Morikawa, Y. et al. Actin cytoskeletal remodeling with protrusion formation is essential for heart regeneration in Hippo-deficient mice. Sci. Signal. 8, ra41 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  49. Chevrier, S. et al. Compensation of signal spillover in suspension and imaging mass cytometry. Cell Syst. 6, 612–620.e615 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Fleming, S. J., Marioni, J. C. & Babadi, M. CellBender remove-background: a deep generative model for unsupervised removal of background noise from scRNA-seq datasets. Preprint at bioRxiv https://doi.org/10.1101/791699 (2019).

  51. Korsunsky, I. et al. Fast, sensitive and accurate integration of single-cell data with Harmony. Nat. Methods 16, 1289–1296 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Li, L. et al. Pitx2 maintains mitochondrial function during regeneration to prevent myocardial fat deposition. Development 145, dev168609 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  53. Yu, G., Wang, L. G., Han, Y. & He, Q. Y. clusterProfiler: an R package for comparing biological themes among gene clusters. OMICS 16, 284–287 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Love, M. I., Huber, W. & Anders, S. Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol 15, 550 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  55. Marini, F. & Binder, H. pcaExplorer: an R/Bioconductor package for interacting with RNA-seq principal components. BMC Bioinf. 20, 331 (2019).

    Article  Google Scholar 

  56. Ramilowski, J. A. et al. A draft network of ligand–receptor-mediated multicellular signalling in human. Nat. Commun. 6, 7866 (2015).

    Article  ADS  CAS  PubMed  Google Scholar 

  57. Tirosh, I. et al. Dissecting the multicellular ecosystem of metastatic melanoma by single-cell RNA-seq. Science 352, 189–196 (2016).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  58. Monroe, T. O. et al. YAP partially reprograms chromatin accessibility to directly induce adult cardiogenesis in vivo. Dev. Cell 48, 765–779.e767 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Langmead, B. & Salzberg, S. L. Fast gapped-read alignment with Bowtie 2. Nat. Methods 9, 357–359 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Zhang, Y. et al. Model-based analysis of ChIP-seq (MACS). Genome Biol. 9, R137 (2008).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  61. Quinlan, A. R. & Hall, I. M. BEDTools: a flexible suite of utilities for comparing genomic features. Bioinformatics 26, 841–842 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Hansen, K. D., Irizarry, R. A. & Wu, Z. Removing technical variability in RNA-seq data using conditional quantile normalization. Biostatistics 13, 204–216 (2012).

    Article  PubMed  PubMed Central  MATH  Google Scholar 

  63. Heinz, S. et al. Simple combinations of lineage-determining transcription factors prime cis-regulatory elements required for macrophage and B cell identities. Mol. Cell 38, 576–589 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

This work was supported by the Department of Defense (CDMRP) W81XWH-17-1-0418 (J.F.M.); National Institutes of Health (1F31HL156681-01 (H.L.), F30HL145908 (Z.A.K.), 5T32HL007208-42 (M.C.H.), R56 HL142704 and R01HL142704 (J.W.), and R01HL 127717, R01HL 130804 and R01HL 118761 (J.F.M.); the Vivian L. Smith Foundation (J.F.M.); Baylor Research Advocates for Student Scientists and Baylor College of Medicine Medical Scientist Training Program (Z.A.K.); the LeDucq Foundation’s Transatlantic Networks of Excellence in Cardiovascular Research (14CVD01: Defining the Genomic Topology of Atrial Fibrillation), the MacDonald Research Fund Award (16RDM001), and a grant from the Saving Tiny Hearts Society (J.F.M.); and NIH HL149164, HL148785 and University of Kentucky Myocardial Recovery Alliance (E.J.B. and K.S.C.). The TCBR is supported by: Children’s Discovery Institute of Washington University and St Louis Children’s Hospital (PM-LI-2019-829) (J.N. and K.L.), the Baylor College of Medicine Pathology and Histology Core and the BCM Breast Cancer Core. This project was supported by the Optical Imaging and Vital Microscopy (OiVM) core at BCM. This research was performed in the Flow Cytometry and Cellular Imaging Core Facility, which is supported in part by the National Institutes of Health through M. D. Anderson's Cancer Center Support Grant CA016672, the NCI’s Research Specialist 1 R50 CA243707-01A1, and a Shared Instrumentation Award from the Cancer Prevention Research Institution of Texas (CPRIT), RP121010. This project was supported by the Cytometry and Cell Sorting Core at Baylor College of Medicine with funding from the CPRIT Core Facility Support Award (CPRIT-RP180672), the NIH (CA125123 and RR024574) and the assistance of J. M. Sederstrom. This project was supported in part by the Genomic and RNA Profiling Core at Baylor College of Medicine with funding from the NIH S10 grant (1S10OD023469). We acknowledge the Gill Cardiovascular Biorepository at the University of Kentucky for providing paediatric control myocardium samples. N. Stancel provided editorial support. Artwork for some figures was generated with BioRender.com.

Author information

Authors and Affiliations

Authors

Contributions

Conceptualization: I.A., M.C.H. and J.F.M. Methodology: M.C.H., Z.A.K., H.L. and Y.M. Writing, original draft: M.C.H. and J.F.M. Writing, review and editing: J.F.M., M.C.H., I.A., D.T., J.W., Z.A.K., H.L. and Y.M. Funding acquisition: I.A., J.F.M. and J.W. Resources: J.F.M., J.W., I.A., E.J.B., K.S.C., J.N., K.L. and L.W. Supervision: J.F.M. and I.A. Data curation: M.C.H. and T.J.M.

Corresponding author

Correspondence to James F. Martin.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature thanks Eva van Rooij and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

Additional information

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Extended data figures and tables

Extended Data Fig. 1 Cardiac tissue snRNA-seq profiling.

a, The number of nuclei detected per sample. Calculations for cell (nuclei) per library performed be dividing total number of nuclei by the number of technical replicates (libraries). b, UMAP showing sample identity. Colored according to Extended Data Fig. 1a. c, Cluster composition across cell types. (Top) Number of samples detected per cluster. (Bottom) Stacked bar graph indicating the percentage of each samples contribution to the indicated cluster. Colored according to Extended Data Fig. 1a. d, Pseduobulk RNA-seq from all cell types collapsed. All technical and biological and technical replicates (libraries) are shown. e, Feature plot showing expression of marker genes across global UMAP from Fig. 1b.

Extended Data Fig. 2 Characterization of pediatric cardiomyocytes.

a, Bar plot indicating the number of cardiomyocyte nuclei detected from each pediatric human sample. b, Stacked bar plot showing the composition of each patient sample across the indicated CM clusters. c, Beeswarm plot showing the log-fold distribution of changes across disease and donors in neighborhoods from different cardiomyocyte clusters. Differentially abundant neighborhoods are shown in color. d, A scree plot displaying the proportion of explained variance for all principal components derived from the pseudo-bulk RNA-seq analysis of CMs. e, Bar plot showing the significance of relation with PC1 (red) and PC2 (blue). f, Venn diagram for the intersection of all individual Diagnoses from pseudobulk RNA-seq analysis. g, Venn diagram for the intersection of all age-related and diagnosis-related genes from pseudo-bulk analysis of CM nuclei. h, Gene signature scores projected across UMAP embedding of CM nuclei. i, Violin plot of CM maturation gene module scores for all CMs separated by patient. The patients are ordered by age, from youngest (left) to oldest (right). j, Violin plot of CM maturation gene module scores for all CMs derived from Sim et. al.

Extended Data Fig. 3 Cardiomyocyte transcriptomic Maturation.

a, Heatmap of log2 foldchange values from pseudo-bulk RNA-seq analysis of CMs. Adjusted p-value < 0.01. b, Gene ontology (GO) analysis of mature and young gene signatures identified in Extended Data Fig. 2j.

Extended Data Fig. 4 Epigenomic characterization of cardiomyocytes in CHD.

a, Feature plots showing gene expression in cardiomyocytes. b–d, RNAscope for CRIM1 and CORIN. e, Genome browser tracks displaying cardiomyocyte-specific ATAC-seq data. f, Venn diagram showing the overlap of genes annotated from ATAC-seq peaks and snRNA-seq clusters.

Extended Data Fig. 5 Transcriptional profiling of pediatric cardiac fibroblasts and endothelial cells.

a, Bar plot indicating the number of fibroblast nuclei detected from each sample. b, Stacked bar plot showing the composition of each sample across the indicated CF clusters. c, Stacked bar graph displaying the total number of detected CF nuclei from each patient group. The composition of each CF cluster is highlighted. d, Enrichment map for gene pathway over-representation analysis colored by CF cluster. e, UMAP manifold of cardiac ECs colored by cluster. (f) Dot plot of snRNA-seq expression for EC marker genes. g, Embedding of the Milo K-NN differential abundance testing results for ECs. All nodes represent neighborhoods, colored by their log fold changes for disease versus donor. Neighborhoods with insignificant log fold changes (FDR 10%) are white. Layout of nodes determined by UMAP embedding, shown in Extended Data Fig. 5e. h, Beeswarm plot showing the log-fold distribution of changes across disease and donors in neighborhoods from different EC clusters. Differentially abundant neighborhoods are shown in color. i, Top, cluster composition bar plot colored by patient diagnosis. Bottom, heatmap displaying average expression for all differentially expressed genes for EC clusters. j, Enrichment map for gene pathway over-representation analysis colored by EC cluster.

Extended Data Fig. 6 Additional tissue histology.

a–e, H&E and trichrome staining of additional myocardial samples from donor (a), TOF (b), Neo-HLHS (c), HF-HLHS (d), and DCM (e) patients. Left image is a 2-mm core, and the dashed box outlines the highlighted perivascular region at high magnification in the right image.

Extended Data Fig. 7 Transcriptional profiling of pediatric cardiac immune cell populations.

a, Representative images of cell-segmentation and phenotyping analysis performed on imaging mass cytometry (IMC) images. b, Representative images of IMC (top) and immunofluorescence (bottom)of LYZ marker expression. Solid boxes indicate same regions with LYZ expression. c, Embedding of the Milo K-NN differential abundance testing results for cardiac immune cell populations. All nodes represent neighborhoods, colored by their log fold changes for disease versus Donor. Neighborhoods with insignificant log fold changes (FDR 10%) are white. Layout of nodes determined by UMAP embedding, shown in Fig. 6f. d, Beeswarm plot showing the log-fold distribution of changes across disease and donors in neighborhoods from different immune cell clusters. Differentially abundant neighborhoods are shown in color. Compiled from Extended Data Fig. 7a. (e) Top, cluster composition bar plot colored by patient diagnosis. Bottom, heatmap displaying average expression for all differentially expressed genes for myeloid and lymphoid cell clusters. (f) Enrichment map for gene pathway over-representation analysis colored by cardiac immune cell cluster. (g) Left, protein expression from IMC data across UMAP embedding. Right, feature plot displaying gene expression from snRNA-seq. (h) Feature plot displaying gene expression from snRNA-seq. Related to Fig. 1b.

Extended Data Fig. 8 Single-cell transcriptomic analysis of PBMCs from pediatric patients with CHD.

a, Diagram depicting the overall study design for hematological profiling in CHD patients. b, UMAP embedding of PBMCs colored by cell type. c, Feature plots showing the expression of marker genes. d, UMAP embedding of PBMC scRNA-seq data colored by patient sample Identification number. e, Cluster composition analysis of PBMC scRNA-seq data. f, Cluster composition stacked-bar plot highlighting the percent of cells from each diagnosis group within each single cell cluster. g, Density plot for each indicated diagnosis over the UMAP embedding from Extended Data Fig. 8b. h, UMAP embedding of peripheral monocyte cells. i, Dot plot displaying marker gene expression from monocyte cell clusters. j, Cluster composition analysis of monocyte clusters colored by patient diagnosis. k, UMAP embedding of peripheral NK cells. l, Dot plot displaying marker gene expression from NK cell clusters. m, Cluster composition analysis of NK cell clusters colored by patient diagnosis.

Extended Data Fig. 9 Transcriptomic and epigenomic characterization of peripheral immune cell populations in CHD.

a, Top, PCA plot of NK and CD14+ cell ATAC-seq data. Bottom, Genome Browser tracks for NK cell and CD14+ cell ATAC-seq data. Each track is patient matched by diagnosis. b, Heatmap displaying differential chromatin accessibility analysis of NK cells versus CD14+ monocytes. c, Genome browser tracks displaying genes identified from differential expression analysis in scRNA-seq. d, Left, heatmap displaying the average mRNA expression per cluster of each cytokine (rows) detected in the plasma of CHD patients. Right, heatmap showing the protein expression of each cytokine (row) detected in CHD patient-derived plasma (columns). e, Heatmap depicting the expression of each cognate receptor from the putative ligands identified in Extended Data Fig. 9d from the cardiac snRNA-seq data. f, receptor-ligand map showing the highly expressed receptors identified in Extended Data Fig. 9e with their respective ligands identified in patient plasma. Connections colored by snRNA-seq cluster.

Supplementary information

Supplementary Information

This file contains a guide to Supplementary Tables 1–18 (tables supplied separately).

Reporting Summary

Supplementary Tables

This zipped file contains Supplementary Tables 1–18.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Hill, M.C., Kadow, Z.A., Long, H. et al. Integrated multi-omic characterization of congenital heart disease. Nature 608, 181–191 (2022). https://doi.org/10.1038/s41586-022-04989-3

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41586-022-04989-3

This article is cited by

Comments

By submitting a comment you agree to abide by our Terms and Community Guidelines. If you find something abusive or that does not comply with our terms or guidelines please flag it as inappropriate.

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing