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:

Delineating copy number and clonal substructure in human tumors from single-cell transcriptomes

Abstract

Single-cell transcriptomic analysis is widely used to study human tumors. However, it remains challenging to distinguish normal cell types in the tumor microenvironment from malignant cells and to resolve clonal substructure within the tumor. To address these challenges, we developed an integrative Bayesian segmentation approach called copy number karyotyping of aneuploid tumors (CopyKAT) to estimate genomic copy number profiles at an average genomic resolution of 5 Mb from read depth in high-throughput single-cell RNA sequencing (scRNA-seq) data. We applied CopyKAT to analyze 46,501 single cells from 21 tumors, including triple-negative breast cancer, pancreatic ductal adenocarcinoma, anaplastic thyroid cancer, invasive ductal carcinoma and glioblastoma, to accurately (98%) distinguish cancer cells from normal cell types. In three breast tumors, CopyKAT resolved clonal subpopulations that differed in the expression of cancer genes, such as KRAS, and signatures, including epithelial-to-mesenchymal transition, DNA repair, apoptosis and hypoxia. These data show that CopyKAT can aid in the analysis of scRNA-seq data in a variety of solid human tumors.

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 CopyKAT analysis workflow.
Fig. 2: Comparison of bulk DNA and single-cell RNA copy number profiles.
Fig. 3: Classification of cancer cells and normal cells in human tumors.
Fig. 4: Classification of tumor and normal cells sequenced by different scRNA-seq technologies.
Fig. 5: Clonal substructure of three triple-negative breast tumors.

Similar content being viewed by others

Data availability

scRNA-seq data from this study were deposited in the Gene Expression Omnibus (GEO; GSE148673).

Code availability

Software is available at GitHub (https://github.com/navinlabcode/copykat).

References

  1. Peng, J. et al. Single-cell RNA-seq highlights intra-tumoral heterogeneity and malignant progression in pancreatic ductal adenocarcinoma. Cell Res. 29, 725–738 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Ma, L. et al. Tumor cell biodiversity drives microenvironmental reprogramming in liver cancer. Cancer Cell 36, 418–430 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Patel, A. P. et al. Single-cell RNA-seq highlights intratumoral heterogeneity in primary glioblastoma. Science 344, 1396–1401 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. 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 

  5. 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 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Gao, R. et al. Nanogrid single-nucleus RNA sequencing reveals phenotypic diversity in breast cancer. Nat. Commun. 8, 228 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  8. Gierahn, T. M. et al. Seq-Well: portable, low-cost RNA sequencing of single cells at high throughput. Nat. Methods 14, 395–398 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Taylor, A. M. et al. Genomic and functional approaches to understanding cancer aneuploidy. Cancer Cell 33, 676–689 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Fan, J. et al. Linking transcriptional and genetic tumor heterogeneity through allele analysis of single-cell RNA-seq data. Genome Res. 28, 1217–1227 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Freeman, M. F. & Tukey, J. W. Transformations related to the angular and the square root. Ann. Math. Stat. 21, 607–611 (1950).

    Article  Google Scholar 

  12. Petris, G. An R package for dynamic linear models. J. Stat. Softw. 36, 1–16 (2010).

    Article  Google Scholar 

  13. Baslan, T. et al. Genome-wide copy number analysis of single cells. Nat. Protoc. 7, 1024–1041 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Harada, T. et al. Genome-wide DNA copy number analysis in pancreatic cancer using high-density single nucleotide polymorphism arrays. Oncogene 27, 1951–1960 (2008).

    Article  CAS  PubMed  Google Scholar 

  15. Samuel, N. et al. Integrated genomic, transcriptomic, and RNA-interference analysis of genes in somatic copy number gains in pancreatic ductal adenocarcinoma. Pancreas 42, 1016–1026 (2013).

    Article  CAS  PubMed  Google Scholar 

  16. Cancer Genome Atlas Research Network. Integrated genomic characterization of pancreatic ductal adenocarcinoma. Cancer Cell 32, 185–203 (2017).

    Article  Google Scholar 

  17. Yao, H. et al. Glypican-3 and KRT19 are markers associating with metastasis and poor prognosis of pancreatic ductal adenocarcinoma. Cancer Biomark. 17, 397–404 (2016).

    Article  CAS  PubMed  Google Scholar 

  18. Girgis, A. H., Bui, A., White, N. M. & Yousef, G. M. Integrated genomic characterization of the kallikrein gene locus in cancer. Anticancer Res. 32, 957–963 (2012).

    PubMed  Google Scholar 

  19. Dijk, F. et al. Unsupervised class discovery in pancreatic ductal adenocarcinoma reveals cell-intrinsic mesenchymal features and high concordance between existing classification systems. Sci. Rep. 10, 337 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Heid, I. et al. Co-clinical assessment of tumor cellularity in pancreatic cancer. Clin. Cancer Res. 23, 1461–1470 (2017).

    Article  CAS  PubMed  Google Scholar 

  21. Ravi, N. et al. Identification of targetable lesions in anaplastic thyroid cancer by genome profiling. Cancers 11, 402 (2019).

    Article  CAS  PubMed Central  Google Scholar 

  22. Ribeiro, F. R., Meireles, A. M., Rocha, A. S. & Teixeira, M. R. Conventional and molecular cytogenetics of human non-medullary thyroid carcinoma: characterization of eight cell line models and review of the literature on clinical samples. BMC Cancer 8, 371 (2008).

    Article  PubMed  PubMed Central  Google Scholar 

  23. Guo, D. et al. Cytokeratin-8 in anaplastic thyroid carcinoma: more than a simple structural cytoskeletal protein. Int. J. Mol. Sci. 19, 577 (2018).

  24. Hunt, J. L. Molecular Pathology of Endocrine Diseases (Springer, 2010).

  25. Barletta, J. A. Endocrine pathology: advances, updates, and diagnostic pearls. Surg. Pathol. Clin. 12, xi–xii (2019).

    Article  PubMed  Google Scholar 

  26. Asa, S. L. & LiVolsi, V. A. New diagnostic and management approaches in endocrine pathology. Arch. Pathol. Lab. Med. 132, 1228–1230 (2008).

    Article  PubMed  Google Scholar 

  27. Turner, N. et al. Integrative molecular profiling of triple negative breast cancers identifies amplicon drivers and potential therapeutic targets. Oncogene 29, 2013–2023 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Gao, R. et al. Punctuated copy number evolution and clonal stasis in triple-negative breast cancer. Nat. Genet. 48, 1119–1130 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Andre, F. et al. Molecular characterization of breast cancer with high-resolution oligonucleotide comparative genomic hybridization array. Clin. Cancer Res. 15, 441–451 (2009).

    Article  CAS  PubMed  Google Scholar 

  30. Neftel, C. et al. An integrative model of cellular states, plasticity, and genetics for glioblastoma. Cell 178, 835–849 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Brennan, C. W. et al. The somatic genomic landscape of glioblastoma. Cell 155, 462–477 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Cancer Genome Atlas Network. Comprehensive molecular portraits of human breast tumours. Nature 490, 61–70 (2012).

    Article  Google Scholar 

  33. Forbes, S. A. et al. COSMIC: somatic cancer genetics at high-resolution. Nucleic Acids Res. 45, D777–D783 (2017).

    Article  CAS  PubMed  Google Scholar 

  34. Hanzelmann, S., Castelo, R. & Guinney, J. GSVA: gene set variation analysis for microarray and RNA-seq data. BMC Bioinformatics 14, 7 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  35. Xin, Y. et al. Use of the Fluidigm C1 platform for RNA sequencing of single mouse pancreatic islet cells. Proc. Natl Acad. Sci. USA 113, 3293–3298 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Picelli, S. et al. Smart-seq2 for sensitive full-length transcriptome profiling in single cells. Nat. Methods 10, 1096–1098 (2013).

    Article  CAS  PubMed  Google Scholar 

  37. Liberzon, A. et al. Molecular signatures database (MSigDB) 3.0. Bioinformatics 27, 1739–1740 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Martin, A. D., Quinn, K. M. & Park, J. H. MCMCpack: Markov chain Monte Carlo in R. J. Stat. Softw. 42, 1–21 (2011).

    Article  Google Scholar 

  39. Kim, C. et al. Chemoresistance evolution in triple-negative breast cancer delineated by single-cell sequencing. Cell 173, 879–893 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Olshen, A. B., Venkatraman, E. S., Lucito, R. & Wigler, M. Circular binary segmentation for the analysis of array-based DNA copy number data. Biostatistics 5, 557–572 (2004).

    Article  PubMed  Google Scholar 

  41. Willenbrock, H. & Fridlyand, J. A comparison study: applying segmentation to array CGH data for downstream analyses. Bioinformatics 21, 4084–4091 (2005).

    Article  CAS  PubMed  Google Scholar 

  42. Stuart, T. et al. Comprehensive integration of single-cell data. Cell 177, 1888–1902 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Finak, G. et al. MAST: a flexible statistical framework for assessing transcriptional changes and characterizing heterogeneity in single-cell RNA sequencing data. Genome Biol. 16, 278 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  44. Durinck, S., Spellman, P. T., Birney, E. & Huber, W. Mapping identifiers for the integration of genomic datasets with the R/Bioconductor package biomaRt. Nat. Protoc. 4, 1184–1191 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Liberzon, A. et al. The Molecular Signatures Database (MSigDB) hallmark gene set collection. Cell Syst. 1, 417–425 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Ritchie, M. E. et al. limma powers differential expression analyses for RNA-sequencing and microarray studies. Nucleic Acids Res. 43, e47 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  47. 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 

Download references

Acknowledgements

This work was supported by grants to N.E.N. from the American Cancer Society (129098-RSG-16-092-01-TBG), the National Cancer Institute (RO1CA240526, RO1CA236864), the Emerson Collective Cancer Research Fund (20200619153514) and the CPRIT Single Cell Genomics Center (RP180684). N.E.N. is an AAAS Wachtel Scholar, AAAS Fellow, Andrew Sabin Family Fellow and Jack & Beverly Randall Innovator. This study was supported by the MD Anderson Breast Cancer Moonshot Program. This study was supported by the MD Anderson Sequencing Core Facility Grant (CA016672). This project was also supported by a Susan Komen Postdoctoral Fellowship to R.G. (PDF17487910). Other grant support includes the Anaplastic Thyroid Cancer Research Fund (S.Y.L. and J.R.W.) and an institutional multi-investigator research program grant to S.Y.L.

Author information

Authors and Affiliations

Authors

Contributions

R.G. and N.E.N. designed the research project. R.G. developed and implemented the computational methods with contributions from N.E.N., Y.Y., A.D., F.W. and K.C. M.H. preprocessed the data. S.F.S. and S.M. provided clinical samples. J.R.W. and S.Y.L. collected thyroid tumor samples. S.B., Y.C.H., Y.L., A.S., T.K. and E.S. performed single-cell sequencing experiments. R.G. and N.E.N. wrote the manuscript with input from all authors.

Corresponding author

Correspondence to Nicholas E. Navin.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Peer review information Nature Biotechnology thanks Elana Fertig, Jan Korbel 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.

Supplementary information

Supplementary Information

Supplementary Figs. 1–4 and Tables 1 and 2.

Reporting Summary

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Gao, R., Bai, S., Henderson, Y.C. et al. Delineating copy number and clonal substructure in human tumors from single-cell transcriptomes. Nat Biotechnol 39, 599–608 (2021). https://doi.org/10.1038/s41587-020-00795-2

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41587-020-00795-2

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