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:

Single-cell analysis of structural variations and complex rearrangements with tri-channel processing

Abstract

Structural variation (SV), involving deletions, duplications, inversions and translocations of DNA segments, is a major source of genetic variability in somatic cells and can dysregulate cancer-related pathways. However, discovering somatic SVs in single cells has been challenging, with copy-number-neutral and complex variants typically escaping detection. Here we describe single-cell tri-channel processing (scTRIP), a computational framework that integrates read depth, template strand and haplotype phase to comprehensively discover SVs in individual cells. We surveyed SV landscapes of 565 single cells, including transformed epithelial cells and patient-derived leukemic samples, to discover abundant SV classes, including inversions, translocations and complex DNA rearrangements. Analysis of the leukemic samples revealed four times more somatic SVs than cytogenetic karyotyping, submicroscopic copy-number alterations, oncogenic copy-neutral rearrangements and a subclonal chromothripsis event. Advancing current methods, single-cell tri-channel processing can directly measure SV mutational processes in individual cells, such as breakage–fusion–bridge cycles, facilitating studies of clonal evolution, genetic mosaicism and SV formation mechanisms, which could improve disease classification for precision medicine.

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: Haplotype-aware discovery of SVs in single cells by scTRIP.
Fig. 2: Analysis pipeline for predicting somatic SVs in individual cells.
Fig. 3: Unbiased translocation discovery based on correlated segregation.
Fig. 4: Analysis of complex and ongoing DNA rearrangement processes.
Fig. 5: Haplotype-resolved karyotypes and subclonal heterogeneity of T-ALL relapses.
Fig. 6: Locating previously unrecognized SVs in a T-ALL relapse sample.

Similar content being viewed by others

Data availability

Sequencing data from this study can be retrieved from the European Genome-phenome Archive (EGA) and the European Nucleotide Archive (accession codes: PRJEB30027, PRJEB30059, PRJEB8037, PRJEB33731, EGAS00001003248, EGAS00001003365). Access to human patient data is governed by the EGA Data Access Committee.

Code availability

The computational code of our analytical framework is hosted on GitHub (see https://github.com/friendsofstrandseq/mosaicatcher-pipeline, https://github.com/friendsofstrandseq/TranslocatoR and https://github.com/friendsofstrandseq/mosaicatcher). All code is available freely for academic research.

References

  1. Ciriello, G. et al. Emerging landscape of oncogenic signatures across human cancers. Nat. Genet. 45, 1127–1133 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Mertens, F., Johansson, B., Fioretos, T. & Mitelman, F. The emerging complexity of gene fusions in cancer. Nat. Rev. Cancer 15, 371–381 (2015).

    Article  CAS  PubMed  Google Scholar 

  3. Northcott, P. A. et al. The whole-genome landscape of medulloblastoma subtypes. Nature 547, 311–317 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Beroukhim, R., Zhang, X. & Meyerson, M. Copy number alterations unmasked as enhancer hijackers. Nat. Genet. 49, 5–6 (2016).

    Article  PubMed  CAS  Google Scholar 

  5. Northcott, P. A. et al. Enhancer hijacking activates GFI1 family oncogenes in medulloblastoma. Nature 511, 428–434 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. 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 

  7. Turajlic, S. et al. Tracking cancer evolution reveals constrained routes to metastases: TRACERx renal. Cell 173, 581–594 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Sottoriva, A. et al. A Big Bang model of human colorectal tumor growth. Nat. Genet. 47, 209–216 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Aparicio, S. & Caldas, C. The implications of clonal genome evolution for cancer medicine. N. Engl. J. Med. 368, 842–851 (2013).

    Article  CAS  PubMed  Google Scholar 

  10. Forsberg, L. A., Gisselsson, D. & Dumanski, J. P. Mosaicism in health and disease - clones picking up speed. Nat. Rev. Genet. 18, 128–142 (2017).

    Article  CAS  PubMed  Google Scholar 

  11. Stratton, M. R. Exploring the genomes of cancer cells: progress and promise. Science 331, 1553–1558 (2011).

    Article  CAS  PubMed  Google Scholar 

  12. Korbel, J. O. et al. Paired-end mapping reveals extensive structural variation in the human genome. Science 318, 420–426 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Layer, R. M., Chiang, C., Quinlan, A. R. & Hall, I. M. LUMPY: a probabilistic framework for structural variant discovery. Genome Biol. 15, R84 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  14. Leibowitz, M. L., Zhang, C.-Z. & Pellman, D. Chromothripsis: a new mechanism for rapid karyotype evolution. Annu. Rev. Genet. 49, 183–211 (2015).

    Article  CAS  PubMed  Google Scholar 

  15. Navin, N. E. Cancer genomics: one cell at a time. Genome Biol. 15, 452 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  16. Zahn, H. et al. Scalable whole-genome single-cell library preparation without preamplification. Nat. Methods 14, 167–173 (2017).

    Article  CAS  PubMed  Google Scholar 

  17. Gawad, C., Koh, W. & Quake, S. R. Single-cell genome sequencing: current state of the science. Nat. Rev. Genet. 17, 175–188 (2016).

    Article  CAS  PubMed  Google Scholar 

  18. Bakker, B. et al. Single-cell sequencing reveals karyotype heterogeneity in murine and human malignancies. Genome Biol. 17, 115 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  19. Voet, T. et al. Single-cell paired-end genome sequencing reveals structural variation per cell cycle. Nucleic Acids Res. 41, 6119–6138 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Zhang, C. Z. et al. Chromothripsis from DNA damage in micronuclei. Nature 522, 179–184 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Falconer, E. et al. DNA template strand sequencing of single cells maps genomic rearrangements at high resolution. Nat. Methods 9, 1107–1112 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Porubsky, D. et al. Dense and accurate whole-chromosome haplotyping of individual genomes. Nat. Commun. 8, 1293 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  23. Sanders, A. D. et al. Characterizing polymorphic inversions in human genomes by single-cell sequencing. Genome Res. 26, 1575–1587 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Chaisson, M. J. P. et al. Multi-platform discovery of haplotype-resolved structural variation in human genomes. Nat. Commun. 10, 1784 (2019).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  25. Sanders, A. D., Falconer, E., Hills, M., Spierings, D. C. J. & Lansdorp, P. M. Single-cell template strand sequencing by Strand-seq enables the characterization of individual homologs. Nat. Protoc. 12, 1151–1176 (2017).

    Article  CAS  PubMed  Google Scholar 

  26. Yang, L. et al. Diverse mechanisms of somatic structural variations in human cancer genomes. Cell 153, 919–929 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Li, Y. et al. Patterns of structural variation in human cancer. Preprint at bioRxiv https://doi.org/10.1101/181339 (2017).

  28. Janssen, A., van der Burg, M., Szuhai, K., Kops, G. J. & Medema, R. H. Chromosome segregation errors as a cause of DNA damage and structural chromosome aberrations. Science 333, 1895–1898 (2011).

    Article  CAS  PubMed  Google Scholar 

  29. Mardin, B. R. et al. A cell-based model system links chromothripsis with hyperploidy. Mol. Syst. Biol. 11, 828 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  30. Maciejowski, J., Li, Y., Bosco, N., Campbell, P. J. & de Lange, T. Chromothripsis and kataegis induced by telomere crisis. Cell 163, 1641–1654 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Riches, A. et al. Neoplastic transformation and cytogenetic changes after Gamma irradiation of human epithelial cells expressing telomerase. Radiat. Res. 155, 222–229 (2001).

    Article  CAS  PubMed  Google Scholar 

  32. Rausch, T. et al. DELLY: structural variant discovery by integrated paired-end and split-read analysis. Bioinformatics 28, i333–i339 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Hills, M., O’Neill, K., Falconer, E., Brinkman, R. & Lansdorp, P. M. BAIT: Organizing genomes and mapping rearrangements in single cells. Genome Med. 5, 82 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  34. Amatu, A., Sartore-Bianchi, A. & Siena, S. NTRK gene fusions as novel targets of cancer therapy across multiple tumour types. ESMO Open 1, e000023 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  35. Zhang, C.-Z., Leibowitz, M. L. & Pellman, D. Chromothripsis and beyond: rapid genome evolution from complex chromosomal rearrangements. Genes Dev. 27, 2513–2530 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Campbell, P. J. et al. The patterns and dynamics of genomic instability in metastatic pancreatic cancer. Nature 467, 1109–1113 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Rode, A., Maass, K. K., Willmund, K. V., Lichter, P. & Ernst, A. Chromothripsis in cancer cells: An update. Int. J. Cancer 138, 2322–2333 (2016).

    Article  CAS  PubMed  Google Scholar 

  38. Selvarajah, S. et al. The breakage–fusion–bridge (BFB) cycle as a mechanism for generating genetic heterogeneity in osteosarcoma. Chromosoma 115, 459–467 (2006).

    Article  CAS  PubMed  Google Scholar 

  39. Li, Y. et al. Constitutional and somatic rearrangement of chromosome 21 in acute lymphoblastic leukaemia. Nature 508, 98–102 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. McClintock, B. The stability of broken ends of chromosomes in Zea mays. Genetics 26, 234–282 (1941).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Gisselsson, D. et al. Chromosomal breakage-fusion-bridge events cause genetic intratumor heterogeneity. Proc. Natl Acad. Sci. USA 97, 5357–5362 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Thompson, S. L., Bakhoum, S. F. & Compton, D. A. Mechanisms of chromosomal instability. Curr. Biol. 20, R285–R295 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Stephens, P. J. et al. Massive genomic rearrangement acquired in a single catastrophic event during cancer development. Cell 144, 27–40 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Korbel, J. O. & Campbell, P. J. Criteria for inference of chromothripsis in cancer genomes. Cell 152, 1226–1236 (2013).

    Article  CAS  PubMed  Google Scholar 

  45. Girardi, T., Vicente, C., Cools, J. & De Keersmaecker, K. The genetics and molecular biology of T-ALL. Blood 129, 1113–1123 (2017).

    Article  CAS  PubMed  Google Scholar 

  46. Richter-Pechańska, P. et al. PDX models recapitulate the genetic and epigenetic landscape of pediatric T cell leukemia. EMBO Mol. Med. 10, e9443 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  47. Liu, Y. et al. The genomic landscape of pediatric and young adult T-lineage acute lymphoblastic leukemia. Nat. Genet. 49, 1211–1218 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Wang, Q. et al. Mutations of PHF6 are associated with mutations of NOTCH1, JAK1 and rearrangement of SET-NUP214 in T cell acute lymphoblastic leukemia. Haematologica 96, 1808–1814 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Rao, S. et al. Inactivation of ribosomal protein L22 promotes transformation by induction of the stemness factor, Lin28B. Blood 120, 3764–3773 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Nagel, S. et al. Activation of TLX3 and NKX2-5 in t(5;14)(q35;q32) T cell acute lymphoblastic leukemia by remote 3′-BCL11B enhancers and coregulation by PU.1 and HMGA1. Cancer Res. 67, 1461–1471 (2007).

    Article  CAS  PubMed  Google Scholar 

  51. Bernard, O. A. et al. A new recurrent and specific cryptic translocation, t(5;14)(q35;q32), is associated with expression of the Hox11L2 gene in T acute lymphoblastic leukemia. Leukemia 15, 1495–1504 (2001).

    Article  CAS  PubMed  Google Scholar 

  52. Kunz, J. B. et al. Pediatric T cell lymphoblastic leukemia evolves into relapse by clonal selection, acquisition of mutations and promoter hypomethylation. Haematologica 100, 1442–1450 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Li, L. et al. A far downstream enhancer for murine Bcl11b controls its T cell-specific expression. Blood 122, 902–911 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Sugimoto, K.-J. et al. T cell lymphoblastic leukemia/lymphoma with t(7;14)(p15;q32) [TCRγ-TCL1A translocation]: a case report and a review of the literature. Int. J. Clin. Exp. Pathol. 7, 2615–2623 (2014).

    PubMed  PubMed Central  Google Scholar 

  55. Virgilio, L. et al. Deregulated expression of TCL1 causes T cell leukemia in mice. Proc. Natl Acad. Sci. USA 95, 3885–3889 (1998).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Alkan, C., Coe, B. P. & Eichler, E. E. Genome structural variation discovery and genotyping. Nat. Rev. Genet. 12, 363–376 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Campbell, I. M., Shaw, C. A., Stankiewicz, P. & Lupski, J. R. Somatic mosaicism: implications for disease and transmission genetics. Trends Genet. 31, 382–392 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Dou, Y., Gold, H. D., Luquette, L. J. & Park, P. J. Detecting somatic mutations in normal cells. Trends Genet. 34, 545–557 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Voet, T. et al. Breakage-fusion-bridge cycles leading to inv dup del occur in human cleavage stage embryos. Hum. Mutat. 32, 783–793 (2011).

    Article  CAS  PubMed  Google Scholar 

  60. Bakhoum, S. F. et al. The mitotic origin of chromosomal instability. Curr. Biol. 24, R148–R149 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Wang, Y. K. et al. Genomic consequences of aberrant DNA repair mechanisms stratify ovarian cancer histotypes. Nat. Genet. 49, 856–865 (2017).

    Article  CAS  PubMed  Google Scholar 

  62. Rücker, F. G. et al. Chromothripsis is linked to TP53 alteration, cell cycle impairment, and dismal outcome in acute myeloid leukemia with complex karyotype. Haematologica 103, e17–e20 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  63. Navin, N. E. & Hicks, J. Tracing the tumor lineage. Mol. Oncol. 4, 267–283 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

  64. Lee, H. & Kim, J.-S. Unexpected CRISPR on-target effects. Nat. Biotechnol. 36, 703–704 (2018).

    Article  CAS  PubMed  Google Scholar 

  65. Yoshihara, M., Hayashizaki, Y. & Murakawa, Y. Genomic instability of iPSCs: challenges towards their clinical applications. Stem Cell Rev. 13, 7–16 (2017).

    Article  CAS  Google Scholar 

  66. Rausch, T. et al. Genome sequencing of pediatric medulloblastoma links catastrophic DNA rearrangements with TP53 mutations. Cell 148, 59–71 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. 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 

  68. Lapunzina, P. & Monk, D. The consequences of uniparental disomy and copy number neutral loss-of-heterozygosity during human development and cancer. Biol. Cell 103, 303–317 (2011).

    Article  PubMed  Google Scholar 

  69. Frismantas, V. et al. Ex vivo drug response profiling detects recurrent sensitivity patterns in drug-resistant acute lymphoblastic leukemia. Blood 129, e26–e37 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. van Wietmarschen, N. & Lansdorp, P. M. Bromodeoxyuridine does not contribute to sister chromatid exchange events in normal or Bloom syndrome cells. Nucleic Acids Res. 44, 6787–6793 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  71. 1000 Genomes Project Consortiumet al. A global reference for human genetic variation. Nature 526, 68–74 (2015).

    Article  CAS  Google Scholar 

  72. Garrison, E. & Marth, G. Haplotype-based variant detection from short-read sequencing. Preprint at https://arxiv.org/abs/1207.3907 (2012).

  73. Huber, W., Toedling, J. & Steinmetz, L. M. Transcript mapping with high-density oligonucleotide tiling arrays. Bioinformatics 22, 1963–1970 (2006).

    Article  CAS  PubMed  Google Scholar 

  74. Claussin, C. et al. Genome-wide mapping of sister chromatid exchange events in single yeast cells using Strand-seq. eLife 6, e30560 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  75. Porubsky, D. et al. Direct chromosome-length haplotyping by single-cell sequencing. Genome Res. 26, 1565–1574 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  76. 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 

  77. Benjamini, Y. & Hochberg, Y. Controlling the false discovery rate: a practical and powerful approach to multiple testing. J. R. Stat. Soc. Series B Stat. Methodol. 57, 289–300 (1995).

    Google Scholar 

  78. Klambauer, G. et al. cn.MOPS: mixture of Poissons for discovering copy number variations in next-generation sequencing data with a low false discovery rate. Nucleic Acids Res. 40, e69 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank W. Huber, O. Stegle, F. Marass and P. Lansdorp for discussions and T. Christiansen for software documentation. We thank M. Paulsen (Flow Cytometry Core Facility) for assistance in sorting and C. Eckert for primary T-ALL samples for engraftment and N. Habermann for project support. J.O.K. acknowledges funding from European Research Council Starting (grant no. 336045) and Consolidator (grant no. 773026) grants and the National Institutes of Health (grant no. 3U41HG007497-04S1). Funding also came from the German Research Foundation (grant nos. 391137747 and 395192176) to T.M., the José Carreras Foundation (grant no. DJCLS 06R/2016) to J.O.K., A.E.K. and J.B.K., the Baden-Württemberg Stiftung (grant no. ID16) to A.E.K. and the Iten-Kohaut Stiftung to J.P.B. A.D.S. and H.Y. received postdoctoral fellowships through the Alexander von Humboldt Foundation.

Author information

Authors and Affiliations

Authors

Contributions

A.D.S., T.M. and J.O.K. conceived the study. A.D.S., S.M., M.G., D.P., T.M. and J.O.K. described the SV footprints. The Strand-seq library preparation workflow was created by A.D.S., B.R., G.M.C.L., J.Z. and V.B. BM510 was generated by B.R.M. and J.O.K. T-ALL samples were processed by A.D.S., S.J., B.R., B.B. and J.-P.B. The MosaiCatcher tool for scTRIP data analysis was developed by S.M., M.G., D.P., A.D.S., T.R., T.M. and J.O.K. The Bayesian framework was created by M.G., S.M., D.P., A.D.S., T.R., J.O.K. and T.M. Cell mixing and simulations experiments were performed by S.M., T.R., D.B. and T.M. SCE detection was developed by S.M., V.K. T.M. and A.D.S. Translocations were discovered and validated by A.v.V., A.D.S., D.P. and J.O.K. Clustered rearrangement analyses were done by A.D.S., D.P., T.R., T.M. and J.O.K. CNN-LOHs were detected by D.P., A.D.S. and T.M. Haplotagging was done by M.G., D.P., A.D.S. and T.M. The bulk DNA sequencing was done by T.R. and B.R. T-ALL clinical/cytogenetic data were collected by P.R.-P., J.B.K., M.S., A.K., B.B. and J.-P.B. T-ALL expression was analysed by H.J., P.R.-P., J.B.K., S.J., B.B., B.R., J.-P.B. and A.K. The manuscript was written by A.D.S., T.M. and J.O.K., which was edited and approved by all authors.

Corresponding authors

Correspondence to Tobias Marschall or Jan O. Korbel.

Ethics declarations

Competing interests

The following authors have disclosed a patent application (no. EP19169090): A.D.S., J.O.K., T.M., D.P., S.M. and M.G.

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. 1–24, Tables 1,2,6,8 and 9 and Notes

Reporting Summary

Supplementary Table 3

Overview of Strand-seq libraries included in the study. Metrics of the single cell sequencing data for RPE-1, C7, BM510, P33 and P1 samples, with total number of high-quality mapped fragments per library listed.

Supplementary Table 4

SV calls generated with our framework and using external methodologies. Overview of the single cell SV calls generated for RPE-1, C7, BM510, P33 and P1 samples, with variant allele frequencies and orthogonal validation notes included.

Supplementary Table 5

Presumed clonal CNA events in RPE cells detected by genomic sequencing. Data are shown for RPE-1, C7 and BM510. WGS, whole-genome sequencing; MP, mate-pair sequencing. Only regions of 200 kb and longer are reported.

Supplementary Table 7

Copy-number of BFB segments in single C7 cells. Description of the 10p BFB locus for every C7 cell with the CN estimate provided for each stepwise segment used to predict the BFB cycle number for that cell.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Sanders, A.D., Meiers, S., Ghareghani, M. et al. Single-cell analysis of structural variations and complex rearrangements with tri-channel processing. Nat Biotechnol 38, 343–354 (2020). https://doi.org/10.1038/s41587-019-0366-x

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41587-019-0366-x

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