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:

Construction of a human cell landscape at single-cell level

Abstract

Single-cell analysis is a valuable tool for dissecting cellular heterogeneity in complex systems1. However, a comprehensive single-cell atlas has not been achieved for humans. Here we use single-cell mRNA sequencing to determine the cell-type composition of all major human organs and construct a scheme for the human cell landscape (HCL). We have uncovered a single-cell hierarchy for many tissues that have not been well characterized. We established a ‘single-cell HCL analysis’ pipeline that helps to define human cell identity. Finally, we performed a single-cell comparative analysis of landscapes from human and mouse to identify conserved genetic networks. We found that stem and progenitor cells exhibit strong transcriptomic stochasticity, whereas differentiated cells are more distinct. Our results provide a useful resource for the study of human biology.

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: Constructing an HCL using microwell-seq.
Fig. 2: Immune activation of non-immune cells in the HCL.
Fig. 3: Application of scHCL analysis for stem cell biology.
Fig. 4: Cross-species comparison of cell landscapes.

Similar content being viewed by others

Data availability

The CNGB Nucleotide Sequence Archive accession number is CNP0000325 (https://db.cngb.org/search/?q=CNP0000325). The GEO accession number is GSE134355. The human DGE data are available at https://figshare.com/articles/HCL_DGE_Data/7235471. The mouse DGE data are available at https://figshare.com/articles/MCA_DGE_Data/5435866. Source Data for Figs. 2, 4 and Extended Data Figs. 2, 5, 7, 8, 9, 10, 11 are provided with the paper. HCL data can also be accessed at http://bis.zju.edu.cn/HCL/ or https://db.cngb.org/HCL/.

Code availability

Detailed codes for figures are provided at https://github.com/ggjlab/HCL/. An online R package is available for scHCL (https://github.com/ggjlab/scHCL/).

References

  1. Tanay, A. & Regev, A. Scaling single-cell genomics from phenomenology to mechanism. Nature 541, 331–338 (2017).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  2. Tang, F. et al. mRNA-seq whole-transcriptome analysis of a single cell. Nat. Methods 6, 377–382 (2009).

    Article  CAS  PubMed  Google Scholar 

  3. Ramsköld, D. et al. Full-length mRNA-seq from single-cell levels of RNA and individual circulating tumor cells. Nat. Biotechnol. 30, 777–782 (2012).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  4. Treutlein, B. et al. Reconstructing lineage hierarchies of the distal lung epithelium using single-cell RNA-seq. Nature 509, 371–375 (2014).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  5. Shalek, A. K. et al. Single-cell transcriptomics reveals bimodality in expression and splicing in immune cells. Nature 498, 236–240 (2013).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  6. Macosko, E. Z. et al. Highly parallel genome-wide expression profiling of individual cells using nanoliter droplets. Cell 161, 1202–1214 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Klein, A. M. et al. Droplet barcoding for single-cell transcriptomics applied to embryonic stem cells. Cell 161, 1187–1201 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Plass, M. et al. Cell type atlas and lineage tree of a whole complex animal by single-cell transcriptomics. Science 360, eaaq1723 (2018).

    Article  PubMed  CAS  Google Scholar 

  9. Wagner, D. E. et al. Single-cell mapping of gene expression landscapes and lineage in the zebrafish embryo. Science 360, 981–987 (2018).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  10. Sebe-Pedros, A. et al. Cnidarian cell type diversity and regulation revealed by whole-organism single-cell RNA-seq. Cell 173, 1520–1534.e1520 (2018).

    Article  CAS  PubMed  Google Scholar 

  11. Plasschaert, L. W. et al. A single-cell atlas of the airway epithelium reveals the CFTR-rich pulmonary ionocyte. Nature 560, 377–381 (2018).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  12. Fincher, C. T., Wurtzel, O., de Hoog, T., Kravarik, K. M. & Reddien, P. W. Cell type transcriptome atlas for the planarian Schmidtea mediterranea. Science 360, eaaq1736 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  13. Farrell, J. A. et al. Single-cell reconstruction of developmental trajectories during zebrafish embryogenesis. Science 360, eaar3131 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  14. Cao, J. et al. Comprehensive single-cell transcriptional profiling of a multicellular organism. Science 357, 661–667 (2017).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  15. Spanjaard, B. et al. Simultaneous lineage tracing and cell-type identification using CRISPR-Cas9-induced genetic scars. Nat. Biotechnol. 36, 469–473 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Han, X. et al. Mapping the mouse cell atlas by microwell-seq. Cell 172, 1091–1107.e1017 (2018).

    Article  CAS  PubMed  Google Scholar 

  17. Tabula Muris Consortium. Single-cell transcriptomics of 20 mouse organs creates a Tabula Muris. Nature 562, 367–372 (2018).

    Article  ADS  CAS  Google Scholar 

  18. Cao, J. et al. The single-cell transcriptional landscape of mammalian organogenesis. Nature 566, 496–502 (2019).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  19. Pijuan-Sala, B. et al. A single-cell molecular map of mouse gastrulation and early organogenesis. Nature 566, 490–495 (2019).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  20. Zhong, S. et al. A single-cell RNA-seq survey of the developmental landscape of the human prefrontal cortex. Nature 555, 524–528 (2018).

    Article  ADS  CAS  PubMed  Google Scholar 

  21. Young, M. D. et al. Single-cell transcriptomes from human kidneys reveal the cellular identity of renal tumors. Science 361, 594–599 (2018).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  22. Lake, B. B. et al. Integrative single-cell analysis of transcriptional and epigenetic states in the human adult brain. Nat. Biotechnol. 36, 70–80 (2018).

    Article  CAS  PubMed  Google Scholar 

  23. Gao, S. et al. Tracing the temporal-spatial transcriptome landscapes of the human fetal digestive tract using single-cell RNA-sequencing. Nat. Cell Biol. 20, 721–734 (2018).

    Article  CAS  PubMed  Google Scholar 

  24. Velten, L. et al. Human haematopoietic stem cell lineage commitment is a continuous process. Nat. Cell Biol. 19, 271–281 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Li, L. et al. Single-cell RNA-seq analysis maps development of human germline cells and gonadal niche interactions. Cell Stem Cell 20, 858–873.e854 (2017).

    Article  CAS  PubMed  Google Scholar 

  26. Guo, J. et al. Chromatin and single-cell rna-seq profiling reveal dynamic signaling and metabolic transitions during human spermatogonial stem cell development. Cell Stem Cell 21, 533–546.e536 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Segerstolpe, Å. et al. Single-cell transcriptome profiling of human pancreatic islets in health and type 2 diabetes. Cell Metab. 24, 593–607 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Muraro, M. J. et al. A single-cell transcriptome atlas of the human pancreas. Cell Syst. 3, 385–394.e383 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Baron, M. et al. A single-cell transcriptomic map of the human and mouse pancreas reveals inter- and intra-cell population structure. Cell Syst. 3, 346–360.e344 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Blakeley, P. et al. Defining the three cell lineages of the human blastocyst by single-cell RNA-seq. Development 142, 3151–3165 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Yan, L. et al. Single-cell RNA-Seq profiling of human preimplantation embryos and embryonic stem cells. Nat. Struct. Mol. Biol. 20, 1131–1139 (2013).

    Article  CAS  PubMed  Google Scholar 

  32. Aizarani, N. et al. A human liver cell atlas reveals heterogeneity and epithelial progenitors. Nature 572, 199–204 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Vodyanik, M. A., Bork, J. A., Thomson, J. A. & Slukvin, I. I. Human embryonic stem cell-derived CD34+ cells: efficient production in the coculture with OP9 stromal cells and analysis of lymphohematopoietic potential. Blood 105, 617–626 (2005).

    Article  CAS  PubMed  Google Scholar 

  34. Rezania, A. et al. Reversal of diabetes with insulin-producing cells derived in vitro from human pluripotent stem cells. Nat. Biotechnol. 32, 1121–1133 (2014).

    Article  CAS  PubMed  Google Scholar 

  35. Satija, R., Farrell, J. A., Gennert, D., Schier, A. F. & Regev, A. Spatial reconstruction of single-cell gene expression data. Nat. Biotechnol. 33, 495–502 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Tosches, M. A. et al. Evolution of pallium, hippocampus, and cortical cell types revealed by single-cell transcriptomics in reptiles. Science 360, 881–888 (2018).

    Article  ADS  CAS  PubMed  Google Scholar 

  37. Vento-Tormo, R. et al. Single-cell reconstruction of the early maternal-fetal interface in humans. Nature 563, 347–353 (2018).

    Article  ADS  CAS  PubMed  Google Scholar 

  38. Wolf, F. A. et al. PAGA: graph abstraction reconciles clustering with trajectory inference through a topology preserving map of single cells. Genome Biol. 20, 59 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  39. Chu, L. F. et al. Single-cell RNA-seq reveals novel regulators of human embryonic stem cell differentiation to definitive endoderm. Genome Biol. 17, 173 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  40. Roerink, S. F. et al. Intra-tumour diversification in colorectal cancer at the single-cell level. Nature 556, 457–462 (2018).

    Article  ADS  CAS  PubMed  Google Scholar 

  41. Camp, J. G. et al. Multilineage communication regulates human liver bud development from pluripotency. Nature 546, 533–538 (2017).

    Article  ADS  CAS  PubMed  Google Scholar 

  42. Camp, J. G. et al. Human cerebral organoids recapitulate gene expression programs of fetal neocortex development. Proc. Natl Acad. Sci. USA 112, 15672–15677 (2015).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  43. La Manno, G. et al. Molecular diversity of midbrain development in mouse, human, and stem cells. Cell 167, 566–580.e519 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  44. Hodge, R. D. et al. Conserved cell types with divergent features in human versus mouse cortex. Nature 573, 61–68 (2019).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  45. Aibar, S. et al. SCENIC: single-cell regulatory network inference and clustering. Nat. Methods 14, 1083–1086 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Dobin, A. et al. STAR: ultrafast universal RNA-seq aligner. Bioinformatics 29, 15–21 (2013).

    Article  CAS  PubMed  Google Scholar 

  47. Buettner, F. et al. Computational analysis of cell-to-cell heterogeneity in single-cell RNA-sequencing data reveals hidden subpopulations of cells. Nat. Biotechnol. 33, 155–160 (2015).

    Article  CAS  PubMed  Google Scholar 

  48. Wolf, F. A., Angerer, P. & Theis, F. J. SCANPY: large-scale single-cell gene expression data analysis. Genome Biol. 19, 15 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  49. Galili, T. dendextend: an R package for visualizing, adjusting and comparing trees of hierarchical clustering. Bioinformatics 31, 3718–3720 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Suo, S. et al. Revealing the critical regulators of cell identity in the mouse cell atlas. Cell Rep. 25, 1436–1445.e3 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Shannon, P. et al. Cytoscape: a software environment for integrated models of biomolecular interaction networks. Genome Res. 13, 2498–2504 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. La Manno, G. et al. RNA velocity of single cells. Nature 560, 494–498 (2018).

    Article  ADS  PubMed  PubMed Central  CAS  Google Scholar 

  53. Crow, M., Paul, A., Ballouz, S., Huang, Z. J. & Gillis, J. Characterizing the replicability of cell types defined by single cell RNA-sequencing data using MetaNeighbor. Nat. Commun. 9, 884 (2018).

    Article  ADS  PubMed  PubMed Central  CAS  Google Scholar 

  54. Guo, M., Bao, E. L., Wagner, M., Whitsett, J. A. & Xu, Y. SLICE: determining cell differentiation and lineage based on single cell entropy. Nucleic Acids Res. 45, e54 (2017).

    Article  PubMed  CAS  Google Scholar 

  55. Grün, D. et al. De novo prediction of stem cell identity using single-cell transcriptome data. Cell Stem Cell 19, 266–277 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  56. Guo, J. et al. The adult human testis transcriptional cell atlas. Cell Res. 28, 1141–1157 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Han, X. et al. Mapping human pluripotent stem cell differentiation pathways using high throughput single-cell RNA-sequencing. Genome Biol. 19, 47 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Karamitros, D. et al. Single-cell analysis reveals the continuum of human lympho-myeloid progenitor cells. Nat. Immunol. 19, 85–97 (2018).

    Article  CAS  PubMed  Google Scholar 

  59. Nguyen, Q. H. et al. Profiling human breast epithelial cells using single cell RNA sequencing identifies cell diversity. Nat. Commun. 9, 2028 (2018).

    Article  ADS  PubMed  PubMed Central  CAS  Google Scholar 

  60. Tsang, J. C. H. et al. Integrative single-cell and cell-free plasma RNA transcriptomics elucidates placental cellular dynamics. Proc. Natl Acad. Sci. USA 114, E7786–E7795 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Villani, A. C. et al. Single-cell RNA-seq reveals new types of human blood dendritic cells, monocytes, and progenitors. Science 356, eaah4573 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  62. Wang, M. et al. Single-cell RNA sequencing analysis reveals sequential cell fate transition during human spermatogenesis. Cell Stem Cell 23, 599–614.e4 (2018).

    Article  CAS  PubMed  Google Scholar 

  63. Celniker, S. E. et al. Unlocking the secrets of the genome. Nature 459, 927–930 (2009).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  64. Gu, Z., Gu, L., Eils, R., Schlesner, M. & Brors, B. circlize implements and enhances circular visualization in R. Bioinformatics 30, 2811–2812 (2014).

    Article  CAS  PubMed  Google Scholar 

  65. Fuxman Bass, J. I. et al. Using networks to measure similarity between genes: association index selection. Nat. Methods 10, 1169–1176 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

Download references

Acknowledgements

We thank G-BIO (Hangzhou), Sidansai Biotechnology (Shanghai), BGI (Shenzhen) and CNGB (Shenzhen) for supporting this project; Vazyme for supplying the customized enzymes in the study; the Core Facilities of Zhejiang University School of Medicine for technical support; the Center of Cryo-Electron Microscopy at Zhejiang University for computational support; and Y. Zhu, J. Zhu, L. Huang, L. Shao, Z. Wang, H. Huang, X. Wu, W. Lin, M. Bai, Q. Sun, X. Wu, M. Yao, F. Zhu, Z. Li, L. Huang, L. Shao, Z. Wang and X. Chen for help with sample collection. This publication is part of the Human Cell Atlas: www.humancellatlas.org/publications/. G.G. is a participant of the Human Cell Atlas Project (International), the Alliance for Atlas of Blood Cells (China), and the Cell Atlas Project (Zhejiang University Stem Cell Institute). This work was supported by the National Natural Science Foundation of China (grants 91842301, 81770188, 31722027, 31922049, 31701290, and 31871473), the National Key Research and Development Program (grants 2018YFA0107804, 2018YFA0107801, 2018YFA0800503, and 2018YFC1005003), the Zhejiang Provincial Natural Science Foundation of China (grant R17H080001), and the Fundamental Research Funds for the Central Universities (G.G.).

Author information

Authors and Affiliations

Authors

Contributions

The project was conceived by G.G. Tissue digestion experiments were performed by X.H., Z.Z., R.W. and Y.C. Microwell-seq experiments were performed by X.H., Z.Z., R.W., H.C., F.Y., M.J., J. Wu and S.L. Single-cell data processing, clustering and trajectory analyses were performed by L.F., H.S., J. Wang, Y.X. and C.Y. scHCL analyses and website construction were performed by H.S., Y.Z. and M.C. Cross-species and gene regulation analyses were performed by J. Wang and H.S. Immunostaining experiments were performed by H.C. Stem cell differentiation experiments were performed by H.C., X.M. and S.Z. Sequencing experiments were performed by R.L., Y.G. and M.W. Fetal tissue collections were conducted by Y.C., Y.W. and D.Z. Adult brain tissue collections were conducted by X.J., J.Z., R.Z. and H. Hu. Adult hematopoietic cell collections were conducted by H. Huang. Other adult tissue collections were conducted by H.T., W.G., T.Z., Q.Z., X.B., L.Z., C.W., T.L., J.C. and W.W.. The paper was written by G.G., X.H., Z.Z., L.F., H.S., R.W., Y.C., H.C. and J. Wang. Funding was acquired by G.G. and X.H.

Corresponding authors

Correspondence to Xiaoping Han or Guoji Guo.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Peer review information Nature thanks Berthold Gottgens, Rosario Isasi and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

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 Construction of the HCL.

a, Comparison of t-SNE maps for lung data sets from Tabula Muris (10X Genomics), down-sampled Tabula Muris data (10X Genomics), and MCA data (microwell-seq). Note that downsampling in sequencing depth does not affect cell-type clusters in the Tabula Muris data. Notably, lung data from MCA (sequenced at lower depth) detects more cell-type clusters, including important lung epithelial cells such as AT1 cells, club cells and biopotent progenitors. b, Numbers of cells processed by 31 December 2019 at the HCL for each tissue type. c, Venn diagrams of gene numbers detected in bulk RNA sequencing and microwell-seq (genes with fewer than three counts are excluded). Scatter plots on the right show high correlations (more than 0.8) of average gene expression between bulk RNA sequencing and microwell-seq. We analysed 17,058 genes for kidney and 16,910 genes for lung. d, The percentage of cell types recovered in sub-samples of adult lung, kidney and adrenal gland single-cell data. The major cell-type numbers in representative tissues are near plateau at around 8,000 cells; we collected more than 10,000 cells per tissue on average. e, A Seurat analysis of donor batch effect from four fetal kidney samples (n = 22,439 cells) and three adult kidney samples (n = 22,692 cells). The mixing of different donor single cells in each cell-type cluster suggests a relatively low batch effect in the data. The cluster contribution bar charts on the right suggest that one of the fetal kidney donors lacks C19 and one of the adult kidney donors lacks C22.

Extended Data Fig. 2 Genetic network analysis of the HCL.

a, Verification of pseudo-cell analysis for network interpretation from HCL data. Left, numbers of genes in single cell, pseudo-cell 5, pseudo-cell 10, pseudo-cell 20, pseudo-cell 50 and pseudo-cell 100 from mouse lung single-cell data14,15 generated by 10X Genomics, Smart-seq2 and microwell-seq. Genes were calculated in each cell or pseudo-cell. Sample sizes for each box from left to right were: 10X Genomics: 5,449, 1,089, 540, 272, 112 and 62; Smart-seq2: 1,620, 324, 158, 83, 33 and 20; microwell-seq: 6,940, 1,390, 686, 349, 142 and 81. Right, silhouette value in single cell, pseudo-cell 5, pseudo-cell 10, pseudo-cell 20, pseudo-cell 50 and pseudo-cell 100 from mouse lung single-cell data. A high silhouette value represents good separation. Sample sizes for each box from left to right were: 6,940, 1,390, 686, 349, 142 and 81. Box plots: centre line, median; boxes, first and third quartiles of the distribution; whiskers, highest and lowest data points within 1.5 × IQR. b, t-SNE map of HCL pseudo-cell data showing improved cell-type clustering (n = 30,053 pseudo-cells). c, TF–TF correlation heat map covering 1,521 human TFs generated using HCL pseudo-cell data. The correlation data are listed in Supplementary Table 2. d, Representative TF network in the HCL (PCC > 0.5). Note that the HCL TF network is highly related within small modules but discrete among different modules.

Source Data

Extended Data Fig. 3 t-SNE maps for examples of analysed fetal tissues in the HCL.

t-SNE maps for single-cell data from fetal skin 2 (a; n = 5,294 cells), fetal brain 5 (b, n = 5,096), fetal pancreas 2 (c, n = 6,939), fetal female gonad 1 (d, n = 2,710), fetal rib 3 (e, n = 4,560), fetal male gonad 1 (f, n = 3,358), chorionic villus 1 (g, n = 9,898), and fetal calvaria 1 (h, n = 5,129). Cells are coloured by cell-type cluster.

Extended Data Fig. 4 t-SNE maps for examples of analysed adult tissues in the HCL.

t-SNE maps for single-cell data from adult gallbladder 1 (a, n = 9,769 cells), adult liver 4 (b, n = 4,384), adult transverse colon 1 (c, n = 5,765), adult duodenum 1 (d, n = 4,681), adult ileum 2 (e, n = 3,367), adult trachea 2 (f, n = 9,949), adult thyroid gland 1 (g, n = 6,319), and adult peripheral blood 1 (h, n = 2,719). Cells are coloured according to cell-type cluster.

Extended Data Fig. 5 Analysis of human lung and kidney.

a, t-SNE map of fetal kidney 4 single-cell data (n = 4,511 cells). The experiment was replicated four times with similar results. b, t-SNE map of adult kidney 2 single-cell data (n = 8,877 cells). The experiment was replicated three times with similar results. c, t-SNE map of fetal lung 1 single-cell data (n = 4,526 cells). The experiment was replicated twice with similar results. d, t-SNE map of adult lung 1 single-cell data (n = 8,426 cells). The experiment was replicated three times with similar results. All cells in ad are coloured according to cell-type cluster. eh, Ligand and receptor analysis of fetal kidney 4 (e), adult kidney 2 (f), fetal lung 1 (g) and adult lung 1 (h) using the method CellPhoneDB. The colours represent cell types; line thickness indicates the degree of association between cell types.

Source Data

Extended Data Fig. 6 Examples of novel populations.

a, t-SNE map of adult pleura 1 single-cell data (n = 19,695 cells). Cells are coloured according to cell-type cluster in a, c and d. b, Gene expression heat map showing the top 20 differentially expressed genes for each cell cluster in adult pleura 1. Dark blue, high expression; light blue, low expression. Representative genes are labelled in the corresponding area on the right. c, t-SNE map of adult omentum 3 single-cell data (n = 1,354 cells). d, t-SNE map of fetal muscle single-cell data (n = 18,345 cells). e, Feature plot in the t-SNE map of adult lung 1 single-cell data (n = 8,426 cells). Cells are coloured according to the expression of the indicated marker genes. f, Immunofluorescence assay for the epithelial cell marker KRT17 and the CXC chemokine CXCL2 in human adult lung tissue. Scale bar, 50 μm. The experiment was replicated three times with similar results.

Extended Data Fig. 7 Cross-tissue cellular network.

a, b, t-SNE maps of single-cell data for human tissue-specific stromal cells (n = 9,452 cells). Cells are coloured according to stromal cell subtype (a) or tissue type (b). c, Bar plots showing the contributions of donors to each of the stromal and endothelial cell clusters.

Source Data

Extended Data Fig. 8 Analysis of fetal-to-adult transition.

a, Heat map showing the similarity (PCC) between cell types of adult kidney and fetal kidney. Blue, low similarity; red, high similarity. b, Heat map showing the similarity (PCC) between cell types of adult lung and fetal lung. Blue, low similarity; red, high similarity. c, d, Branching gene expression trajectory analysis of non-immune cells in fetal and adult human tissues using PAGA. c, Coloured by developmental stages; d, coloured by cell lineages. Differential gene expression analysis was performed for representative lineage progression; top markers for fetal (light blue) and adult (dark blue) cells are shown. Marker lists are provided in the source data. e, Single-cell entropy of non-immune cells in each fetal and adult tissue pair (right) and for combined adult and fetal data (left) measured by SLICE54. Box plots: centre line, median; boxes, first and third quartiles of the distribution; whiskers, highest and lowest data points within 1.5 × IQR. Sample from left to right: 2,770, 3,557, 522, 279, 340, 193, 270, 38, 885, 631, 208, 353, 187, 111, 268, 112, 294, 314, 583, 739.

Source Data

Extended Data Fig. 9 scHCL analysis.

a, scHCL results for bulk human ES cell data (n = 17 samples). Each row represents one cell type in our reference. Each column represents a bulk sample. PCC was used to evaluate cell-type gene expression similarity. Red, high correlation; grey, low correlation. Some cell-type data come from published works, as denoted by first author name: Zhong20, Li25, Segerstolpe27, Baron29, VentoTormo37, Camp42, LaManno43, Han57. b, scHCL results for colorectal cancer organoid data (n = 15 samples), as in a. c, scHCL results for liver bud organoid data (n = 465 cells), as in a except that each column represents a single cell in the customer data set. d, scHCL results for cerebral organoid data (n = 508 cells), as in c. e, t-SNE map of single-cell data for day 20 embryoid bodies differentiated from human iPS cells (n = 9,140 cells). Cells are coloured according to cell-type cluster. f, scHCL results obtained using the data set of day 20 embryoid bodies differentiated from human iPS cells (n = 9,140 cells), as in c. g, Cell–cell correlation network for embryoid body. Each node represents a pseudo-cell in each cell type.

Source Data

Extended Data Fig. 10 Comparison of human and mouse tissues.

a, t-SNE analysis of 333,778 single cells from the MCA data, with 104 main cell-type clusters labelled. b, t-SNE analysis of 333,778 single cells from the MCA data, with tissue types labelled. c, t-SNE map of pseudo-cell 20 data for mouse (n = 16,740 pseudo-cells). Pseudo-cells are coloured according to cell-type cluster. d, Heat map showing the conserved cell types in human and mouse adult kidney. AUROC scores were calculated from the Spearman correlation between human and mouse adult kidney pseudo-cells (n = 1,197 pseudo-cells). e, Heat map showing conserved cell types in human and mouse fetal kidney. AUROC scores were calculated from the Spearman correlation between human and mouse fetal kidney pseudo-cells (n = 1,459 pseudo-cells).

Source Data

Extended Data Fig. 11 Comparison of human and mouse regulons.

a, b, Binary regulon activity t-SNE maps for human and mouse based on 259 human regulons (a) and 248 mouse regulons (b), created with R package SCENIC. Each dot represents a pseudo-cell of 20 in the HCL or MCA cell clusters. The t-SNE maps were created using binary regulon activity matrices from 17,028 human pseudo-cells and 16,740 mouse pseudo-cells. c, Binary RASs for human special regulon BHLHE41 and mouse special regulon Olig1 in the regulon activity t-SNE maps (n = 17,028 for human; n = 16,740 for mouse). d, Binary RASs for human special regulon FOXO1 and mouse special regulon Dlx5 in the regulon activity t-SNE maps (n = 17,028 for human; n = 16,740 for mouse). e, Binary RASs for human special regulon ZNF230 and mouse special regulon Mlxipl in the regulon activity t-SNE maps (n = 17,028 for human; n = 16,740 for mouse). f, Binary RASs for regulons IRF8/Irf8 and GATA1/Gata1 in the regulon activity t-SNE maps (n = 17,028 for human; n = 16,740 for mouse). Note that in the regulation of antigen-presenting endothelial cells, the IRF8/Irf8 regulon is conserved. In the regulation of erythroid cells, the GATA1/Gata1 regulon is conserved.

Source Data

Supplementary information

Reporting Summary

Supplementary Table 1 | Basic information about samples processed in the HCL

The table contains the age (gestational age for fetal tissues), gender, cause of death, medical history of the donors, sample location, source, dissociation, concentration and digestion time of the tissues. Information about sequencing titration experiment, the analysed cell number, experimental batches, and average transcript number (UMI) are also provided. All used 0ligonucleotide sequences are listed in the last sheet of the table.

Supplementary Table 2 | Differentially expressed genes detected in 102 human cell types from HCL and 104 mouse cell types from MCA

P values were calculated by the Wilcoxon rank-sum test (n = gene name, l = logFc, s = score, p = P value). Statistical tests were two-sided. Tissue contribution for each of the HCL clusters and HCL TF-TF correlation data matrix are also provided.

Supplementary Table 3 | Differentially expressed genes detected in each cell type for all tissues or cell lines in HCL datasets

Genes were selected by log foldchange > 0.25, Bonferroni-adjusted P < 0.1, expressed in at least 15% of cells in either population (Seurat FindAllMarkers). Log foldchange (avg_logFC) was calculated as the arithmetic mean of ln cpm values of one population minus the arithmetic mean of ln cpm values of the other populations. P value (p_val) was calculated by the Wilcoxon rank-sum test. Statistical tests were two-sided. Sample size for each tissue type was listed in Supplementary Table 1.

Supplementary Table 4 | Sheet 1: List of novel cell populations revealed from the HCL study. Sheets 2 and 3: Differentially expressed genes detected in the cross-tissue comparison of stromal (n=9,452) and endothelial (n=7,140) cells

Genes are selected by log foldchange > 0.25, Bonferroni-adjusted P < 0.1, expressed in at least 15% of cells in either population (Seurat FindAllMarkers). Log foldchange (avg_logFC) was calculated as the arithmetic mean of ln cpm values of one population minus the arithmetic mean of ln cpm values of the other populations. P value (p_val) was calculated by the Wilcoxon rank-sum test. Statistical tests were two-sided. Sheet 4 and 5: Differentially expressed genes between fetal and adult stage for 85 pairs of fetal-to-adult cell types. Genes are ranked by the number of times they appear as fetal or adult differentially expressed genes. Statistical tests were two-sided. Commonly enriched genes for fetal cells are listed on sheet 4; commonly enriched genes for adult cells are listed on sheet 5.

Supplementary Table 5 | External data references and cross-species comparison

The sources of published datasets used in scHCL are listed. The relationship of the corresponding conserved cell types among human and mouse are measured using AUROC scores in MetaNeighbor. The TFs are grouped into 140 orthologous regulons and then clustered into 15 orthologous TF-TF regulon modules using clustering method ‘complete’. The TF module activity in each cluster is listed; a comparison between human and mouse regulons is also provided.

Source data

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Han, X., Zhou, Z., Fei, L. et al. Construction of a human cell landscape at single-cell level. Nature 581, 303–309 (2020). https://doi.org/10.1038/s41586-020-2157-4

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41586-020-2157-4

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: Translational Research

Sign up for the Nature Briefing: Translational Research newsletter — top stories in biotechnology, drug discovery and pharma.

Get what matters in translational research, free to your inbox weekly. Sign up for Nature Briefing: Translational Research