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:

Spatial charting of single-cell transcriptomes in tissues

Abstract

Single-cell RNA sequencing methods can profile the transcriptomes of single cells but cannot preserve spatial information. Conversely, spatial transcriptomics assays can profile spatial regions in tissue sections, but do not have single-cell resolution. Here, we developed a computational method called CellTrek that combines these two datasets to achieve single-cell spatial mapping through coembedding and metric learning approaches. We benchmarked CellTrek using simulation and in situ hybridization datasets, which demonstrated its accuracy and robustness. We then applied CellTrek to existing mouse brain and kidney datasets and showed that CellTrek can detect topological patterns of different cell types and cell states. We performed single-cell RNA sequencing and spatial transcriptomics experiments on two ductal carcinoma in situ tissues and applied CellTrek to identify tumor subclones that were restricted to different ducts, and specific T cell states adjacent to the tumor areas. Our data show that CellTrek can accurately map single cells in diverse tissue types to resolve their spatial organization.

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

Access options

Buy this article

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

Fig. 1: Overview of the CellTrek workflow.
Fig. 2: CellTrek reconstructs spatial organization in a mouse brain tissue.
Fig. 3: CellTrek reconstructs spatial organization in a mouse kidney tissue.
Fig. 4: CellTrek identifies the spatial subclone heterogeneity in DCIS1.
Fig. 5: CellTrek displays the spatial tumor-immune microenvironment in DCIS2.

Similar content being viewed by others

Data availability

The scRNA-seq and ST data were submitted to the Gene Expression Omnibus (GEO): GSE181254.

Code availability

The CellTrek software toolkit is available at GitHub: https://github.com/navinlabcode/CellTrek.

References

  1. Lim, B., Lin, Y. & Navin, N. Advancing cancer research and medicine with single-cell genomics. Cancer Cell 37, 456–470 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Regev, A. et al. The human cell atlas. eLife 6, e27041 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  4. Wang, Y. & Navin, N. E. Advances and applications of single-cell sequencing technologies. Mol. Cell 58, 598–609 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Papalexi, E. & Satija, R. Single-cell RNA sequencing to explore immune cell heterogeneity. Nat. Rev. Immunol. 18, 35–45 (2018).

    Article  CAS  PubMed  Google Scholar 

  6. Longo, S. K., Guo, M. G., Ji, A. L., Khavari, P. A. Integrating single-cell and spatial transcriptomics to elucidate intercellular tissue dynamics. Nat. Rev. Genet. 22, 627–644 (2021).

    Article  CAS  PubMed  Google Scholar 

  7. Lee, J. H. Quantitative approaches for investigating the spatial context of gene expression. Wiley Interdiscip. Rev. Syst. Biol. Med. 9, e1369 (2017).

    Article  CAS  Google Scholar 

  8. Janiszewska, M. The microcosmos of intratumor heterogeneity: the space-time of cancer evolution. Oncogene 39, 2031–2039 (2020).

    Article  CAS  PubMed  Google Scholar 

  9. Stahl, P. L. et al. Visualization and analysis of gene expression in tissue sections by spatial transcriptomics. Science 353, 78–82 (2016).

    Article  CAS  PubMed  Google Scholar 

  10. Rodriques, S. G. et al. Slide-seq: a scalable technology for measuring genome-wide expression at high spatial resolution. Science 363, 1463–1467 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Cable, D. M. et al. Robust decomposition of cell type mixtures in spatial transcriptomics. Nat. Biotechnol. https://doi.org/10.1038/s41587-021-00830-w (2021).

  12. Maaskola, J. et al. Charting tissue expression anatomy by spatial transcriptome decomposition. Preprint at bioRxiv, 362624 (2018).

  13. Andersson, A. et al. Spatial mapping of cell types by integration of transcriptomics data. Preprint at bioRxiv, 2019.2012.2013.874495 (2019).

  14. Andersson, A. et al. Single-cell and spatial transcriptomics enables probabilistic inference of cell type topography. Commun Biol. 3, 565 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  15. Kleshchevnikov, V. et al. Comprehensive mapping of tissue cell architecture via integrated single cell and spatial transcriptomics. Preprint at bioRxiv, 2020.2011.2015.378125 (2020).

  16. Moncada, R. et al. Integrating microarray-based spatial transcriptomics and single-cell RNA-seq reveals tissue architecture in pancreatic ductal adenocarcinomas. Nat. Biotechnol. 38, 333–342 (2020).

    Article  CAS  PubMed  Google Scholar 

  17. Tasic, B. et al. Adult mouse cortical cell taxonomy revealed by single cell transcriptomics. Nat. Neurosci. 19, 335–346 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Ransick, A. et al. Single-cell profiling reveals sex, lineage, and regional diversity in the mouse kidney. Dev. Cell 51, 399–413 e397 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Breiman, L. Random forests. Mach. Learn. 45, 5–32 (2001).

    Article  Google Scholar 

  20. Nitzan, M., Karaiskos, N., Friedman, N. & Rajewsky, N. Gene expression cartography. Nature 576, 132 (2019).

    Article  CAS  PubMed  Google Scholar 

  21. Wilkerson, M. D. & Hayes, D. N. ConsensusClusterPlus: a class discovery tool with confidence assessments and item tracking. Bioinformatics 26, 1572–1573 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Langfelder, P. & Horvath, S. WGCNA: an R package for weighted correlation network analysis. BMC Bioinformatics 9, 559 (2008).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  23. Lohoff, T. Integration of spatial and single-cell transcriptomic data elucidates mouse organogenesis. Nat. Biotechnol. 40, 74–85 (2022).

    Article  CAS  PubMed  Google Scholar 

  24. Stuart, T. et al. Comprehensive integration of single-cell data. Cell 177, 1888 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Trapnell, C. et al. The dynamics and regulators of cell fate decisions are revealed by pseudotemporal ordering of single cells. Nat Biotechnol 32, 381–386 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Qiu, X. J. et al. Reversed graph embedding resolves complex single-cell trajectories. Nat. Methods 14, 979 (2017) .

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Fawkner-Corbett, D. et al. Spatiotemporal analysis of human intestinal development at single-cell resolution. Cell 184, 810–826. e823 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Tasic, B. et al. Shared and distinct transcriptomic cell types across neocortical areas. Nature 563, 72–78 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Stickels, R. R. et al. Highly sensitive spatial transcriptomics at near-cellular resolution with Slide-seqV2. Nat. Biotechnol. 39, 313–319 (2021).

    Article  CAS  PubMed  Google Scholar 

  30. Saunders, A. et al. Molecular diversity and specializations among the cells of the adult mouse brain. Cell 174, 1015–1030.e1016 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Lake, B. B. et al. A single-nucleus RNA-sequencing pipeline to decipher the molecular anatomy and pathophysiology of human kidneys. Nat. Commun. 10, 2832 (2019).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  32. Janosevic, D. et al. The orchestrated cellular and molecular responses of the kidney to endotoxin define a precise sepsis timeline. eLife 10, e62270 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Naray-Fejes-Toth, A., Snyder, P. M. & Fejes-Toth, G. The kidney-specific WNK1 isoform is induced by aldosterone and stimulates epithelial sodium channel-mediated Na+ transport. Proc. Natl Acad. Sci. USA 101, 17434–17439 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Park, J. et al. Single-cell transcriptomics of the mouse kidney reveals potential cellular targets of kidney disease. Science 360, 758–763 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Zhao, Y. et al. Isolation and epithelial co-culture of mouse renal peritubular endothelial cells. BMC Cell Biol. 15, 40 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  37. Miao, Z. et al. Single cell regulatory landscape of the mouse kidney highlights cellular differentiation programs and disease targets. Nat. Commun. 12, 2277 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Mantovani, A. et al. The chemokine system in diverse forms of macrophage activation and polarization. Trends Immunol. 25, 677–686 (2004).

    Article  CAS  PubMed  Google Scholar 

  39. Quinn, K. E., Mackie, D. I. & Caron, K. M. Emerging roles of atypical chemokine receptor 3 (ACKR3) in normal development and physiology. Cytokine 109, 17–23 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Gao, R. et al. Delineating copy number and clonal substructure in human tumors from single-cell transcriptomes. Nat. Biotechnol. 39, 599–608 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Korotkevich, G. et al. Fast gene set enrichment analysis. Preprint at bioRxiv, 060012 (2021).

  42. Casasent, A. K. et al. Multiclonal invasion in breast tumors identified by topographic single cell sequencing. Cell 172, 205 (2018) .

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Prabhakaran, S. et al. Evaluation of invasive breast cancer samples using a 12-chemokine gene expression score: correlation with clinical outcomes. Breast Cancer Res. 19, 71 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  44. Sautes-Fridman, C., Petitprez, F., Calderaro, J. & Fridman, W. H. Tertiary lymphoid structures in the era of cancer immunotherapy. Nat. Rev. Cancer 19, 307–325 (2019).

    Article  CAS  PubMed  Google Scholar 

  45. Sinha, V. C. & Piwnica-Worms, H. Intratumoral heterogeneity in ductal carcinoma in situ: chaos and consequence. J Mammary Gland Biol. Neoplasia 23, 191–205 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  46. Marusyk, A., Janiszewska, M. & Polyak, K. Intratumor heterogeneity: the Rosetta Stone of therapy resistance. Cancer Cell 37, 471–484 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Lennington, W. J., Jensen, R. A., Dalton, L. W. & Page, D. L. Ductal carcinoma in-situ of the breast—heterogeneity of individual lesions. Cancer 73, 118–124 (1994).

    Article  CAS  PubMed  Google Scholar 

  48. Ishwaran, H., Kogalur, U. B., Blackstone, E. H. & Lauer, M. S. Random survival forests. Ann. Appl. Stat. 2, 841–860 (2008).

    Article  Google Scholar 

  49. Hahsler, M., Piekenbrock, M. & Doran, D. dbscan: fast density-based clustering with R. J. Stat. Softw. 91, 1–30 (2019).

    Article  Google Scholar 

  50. Gu, Z., Eils, R. & Schlesner, M. Complex heatmaps reveal patterns and correlations in multidimensional genomic data. Bioinformatics 32, 2847–2849 (2016).

    Article  CAS  PubMed  Google Scholar 

  51. Paradis, E. & Schliep, K. ape 5.0: an environment for modern phylogenetics and evolutionary analyses in R. Bioinformatics 35, 526–528 (2019).

    Article  CAS  PubMed  Google Scholar 

  52. Zappia, L., Phipson, B. & Oshlack, A. Splatter: simulation of single-cell RNA sequencing data. Genome Biol. 18, 174 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  53. Schuhmacher, D. et al. transport: computation of optimal transport plans and Wasserstein distances. R package v.0.12-2, https://cran.r-project.org/package=transport (2020).

  54. 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 

  55. Scrucca, L., Fop, M., Murphy, T. B. & Raftery, A. E. mclust 5: clustering, classification and density estimation using Gaussian finite mixture models. R. J. 8, 289–317 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

This work was supported by grants to N.E.N. from the NIH National Cancer Institute (RO1CA240526, RO1CA236864), the CPRIT Single Cell Genomics Center (RP180684), the Chan-Zuckerberg Initiative (CZI) SEED Network Grant (CZF2019-002432) and the PRECISION Cancer Grand Challenge Grant. N.E.N. is an AAAS Fellow, AAAS Wachtel Scholar, Damon-Runyon Rachleff Innovator, Andrew Sabin Fellow, and Jack & Beverly Randall Innovator. This study was supported by the MD Anderson Sequencing Core Facility Grant (CA016672). R.W. is a Damon Runyon Fellow supported by the Damon Runyon Cancer Research Foundation (DRQ-08-20). We thank J. Wang, X. He and J. Wei for their unwavering support. We also thank D. Minussi and Y. Lin for their valuable suggestions.

Author information

Authors and Affiliations

Authors

Contributions

R.W. and S.H. developed the CellTrek method, analyzed data, prepared figures and wrote the manuscript. S.B. and E.S. performed single-cell and ST experiments. M.H. processed data. A.T. and S.K. collected the DCIS tissue samples, interpreted data and managed the IRB protocols. K.C. provided input on the CellTrek method and manuscript. N.E.N. managed the project and wrote the manuscript.

Corresponding author

Correspondence to Nicholas E. Navin.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature Biotechnology thanks the anonymous reviewers 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.

Supplementary information

Supplementary Information

Supplementary Figs. 1–11, Tables 1–4 and Data 1–4.

Reporting Summary

Supplementary Data 1

The CellTrek map for mouse brain.

Supplementary Data 2

The CellTrek map for mouse brain.

Supplementary Data 3

The CellTrek map for DCIS1.

Supplementary Data 4

The CellTrek map for DCIS2.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Wei, R., He, S., Bai, S. et al. Spatial charting of single-cell transcriptomes in tissues. Nat Biotechnol 40, 1190–1199 (2022). https://doi.org/10.1038/s41587-022-01233-1

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41587-022-01233-1

This article is cited by

Search

Quick links

Nature Briefing: Cancer

Sign up for the Nature Briefing: Cancer newsletter — what matters in cancer research, free to your inbox weekly.

Get what matters in cancer research, free to your inbox weekly. Sign up for Nature Briefing: Cancer