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.

  • Protocol
  • Published:

Simultaneous quantification of protein–DNA interactions and transcriptomes in single cells with scDam&T-seq

Abstract

Protein–DNA interactions are essential for establishing cell type–specific chromatin architecture and gene expression. We recently developed scDam&T-seq, a multi-omics method that can simultaneously quantify protein–DNA interactions and the transcriptome in single cells. The method effectively combines two existing methods: DNA adenine methyltransferase identification (DamID) and CEL-Seq2. DamID works through the tethering of a protein of interest (POI) to the Escherichia coli DNA adenine methyltransferase (Dam). Upon expression of this fusion protein, DNA in proximity to the POI is methylated by Dam and can be selectively digested and amplified. CEL-Seq2, in contrast, makes use of poly-dT primers to reverse transcribe mRNA, followed by linear amplification through in vitro transcription. scDam&T-seq is the first technique capable of providing a combined readout of protein–DNA contact and transcription from single-cell samples. Once suitable cell lines have been established, the protocol can be completed in 5 d, with a throughput of hundreds to thousands of cells. The processing of raw sequencing data takes an additional 1–2 d. Our method can be used to understand the transcriptional changes a cell undergoes upon the DNA binding of a POI. It can be performed in any laboratory with access to FACS, robotic and high-throughput-sequencing facilities.

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 method.
Fig. 2: CEL-Seq2 primer and DamID adapter structure.
Fig. 3: Bioinformatics workflow.
Fig. 4: Examples of aRNA bioanalyzer plots.
Fig. 5: Technical statistics of a scDam&T-seq run.
Fig. 6: Examples of DNA bioanalyzer plots.

Similar content being viewed by others

Data availability

A test dataset is available from GitHub (https://github.com/KindLab/scDamAndTools).

Code availability

All codes are available from GitHub (https://github.com/KindLab/scDamAndTools). The code in this manuscript has been peer reviewed.

References

  1. Johnson, D. S., Mortazavi, A., Myers, R. M. & Wold, B. Genome-wide mapping of in vivo protein-DNA interactions. Science 316, 1497–1502 (2007).

    Article  CAS  PubMed  Google Scholar 

  2. Vogel, M. J., Peric-Hupkes, D. & van Steensel, B. Detection of in vivo protein-DNA interactions using DamID in mammalian cells. Nat. Protoc. 2, 1467–1478 (2007).

    Article  CAS  PubMed  Google Scholar 

  3. Crawford, G. E. et al. Genome-wide mapping of DNase hypersensitive sites using massively parallel signature sequencing (MPSS). Genome Res. 16, 123–131 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Lieberman-Aiden, E. et al. Comprehensive mapping of long-range interactions reveals folding principles of the human genome. Science 326, 289–293 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Nagano, T. et al. Single-cell Hi-C reveals cell-to-cell variability in chromosome structure. Nature 502, 59–64 (2013).

    Article  CAS  PubMed  Google Scholar 

  6. Kind, J. et al. Genome-wide maps of nuclear lamina interactions in single human cells. Cell 163, 134–147 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Flyamer, I. M. et al. Single-nucleus Hi-C reveals unique chromatin reorganization at oocyte-to-zygote transition. Nature 544, 110–114 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Stevens, T. J. et al. 3D structures of individual mammalian genomes studied by single-cell Hi-C. Nature 544, 59–64 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Buenrostro, J. D. et al. Single-cell chromatin accessibility reveals principles of regulatory variation. Nature 523, 486–490 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Cusanovich, D. A. et al. Multiplex single cell profiling of chromatin accessibility by combinatorial cellular indexing. Science 348, 910–914 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Jin, W. et al. Genome-wide detection of DNase I hypersensitive sites in single cells and FFPE tissue samples. Nature 528, 142–146 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Guo, H. et al. Single-cell methylome landscapes of mouse embryonic stem cells and early embryos analyzed using reduced representation bisulfite sequencing. Genome Res. 23, 2126–2135 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Smallwood, S. A. et al. Single-cell genome-wide bisulfite sequencing for assessing epigenetic heterogeneity. Nat. Methods 11, 817–820 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Farlik, M. et al. Single-cell DNA methylome sequencing and bioinformatic inference of epigenomic cell-state dynamics. Cell Rep. 10, 1386–1397 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Mooijman, D., Dey, S. S., Boisset, J. C., Crosetto, N. & van Oudenaarden, A. Single-cell 5hmC sequencing reveals chromosome-wide cell-to-cell variability and enables lineage reconstruction. Nat. Biotechnol. 34, 852–856 (2016).

    Article  CAS  PubMed  Google Scholar 

  16. Wu, X., Inoue, A., Suzuki, T. & Zhang, Y. Simultaneous mapping of active DNA demethylation and sister chromatid exchange in single cells. Genes Dev. 31, 511–523 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Zhu, C. et al. Single-cell 5-formylcytosine landscapes of mammalian early embryos and ESCs at single-base resolution. Cell Stem Cell 20, 720–731.e5 (2017).

    Article  CAS  PubMed  Google Scholar 

  18. Rotem, A. et al. Single-cell ChIP-seq reveals cell subpopulations defined by chromatin state. Nat. Biotechnol. 33, 1165–1172 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Harada, A. et al. A chromatin integration labelling method enables epigenomic profiling with lower input. Nat. Cell Biol. 21, 287–296 (2019).

    Article  CAS  PubMed  Google Scholar 

  20. Hainer, S. J., Boskovic, A., McCannell, K. N., Rando, O. J. & Fazzio, T. G. Profiling of pluripotency factors in single cells and early embryos. Cell 177, 1319–1329.e11 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Ku, W. L. et al. Single-cell chromatin immunocleavage sequencing (scChIC-seq) to profile histone modification. Nat. Methods 16, 323–325 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Angermueller, C. et al. Parallel single-cell sequencing links transcriptional and epigenetic heterogeneity. Nat. Methods 13, 229–232 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Hou, Y. et al. Single-cell triple omics sequencing reveals genetic, epigenetic, and transcriptomic heterogeneity in hepatocellular carcinomas. Cell Res. 26, 304–319 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Clark, S. J. et al. scNMT-seq enables joint profiling of chromatin accessibility DNA methylation and transcription in single cells. Nat. Commun. 9, 781 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  25. Rooijers, K. et al. Simultaneous quantification of protein-DNA contacts and transcriptomes in single cells. Nat. Biotechnol. 37, 766–772 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Hashimshony, T., Wagner, F., Sher, N. & Yanai, I. CEL-Seq: single-cell RNA-Seq by multiplexed linear amplification. Cell Rep. 2, 666–673 (2012).

    Article  CAS  PubMed  Google Scholar 

  27. Hashimshony, T. et al. CEL-Seq2: sensitive highly-multiplexed single-cell RNA-Seq. Genome Biol. 17, 77 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  28. Nishimura, K., Fukagawa, T., Takisawa, H., Kakimoto, T. & Kanemaki, M. An auxin-based degron system for the rapid depletion of proteins in nonplant cells. Nat. Methods 6, 917–922 (2009).

    Article  CAS  PubMed  Google Scholar 

  29. Boers, R. et al. Genome-wide DNA methylation profiling using the methylation-dependent restriction enzyme LpnPI. Genome Res. 28, 88–99 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Sen, M. et al. Strand-specific single-cell methylomics reveals distinct modes of DNA demethylation dynamics during early mammalian development. Preprint at bioRxiv https://doi.org/10.1101/804526 (2019).

  31. Borsos, M. et al. Genome-lamina interactions are established de novo in the early mouse embryo. Nature 569, 729–733 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Liu, C. L., Schreiber, S. L. & Bernstein, B. E. Development and validation of a T7 based linear amplification for genomic DNA. BMC Genomics 4, 19 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Grosselin, K. et al. High-throughput single-cell ChIP-seq identifies heterogeneity of chromatin states in breast cancer. Nat. Genet. 51, 1060–1066 (2019).

    Article  CAS  PubMed  Google Scholar 

  34. Kaya-Okur, H. S. et al. CUT&Tag for efficient epigenomic profiling of small samples and single cells. Nat. Commun. 10, 1930 (2019).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  35. Schmid, M., Durussel, T. & Laemmli, U. K. ChIC and ChEC: genomic mapping of chromatin proteins. Mol. Cell 16, 147–157 (2004).

    CAS  PubMed  Google Scholar 

  36. Skene, P. J. & Henikoff, S. An efficient targeted nuclease strategy for high-resolution mapping of DNA binding sites. eLife 6, e21856 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  37. Sirunyan, A. M. et al. Search for rare decays of Z and Higgs bosons to J/Ψ and a photon in proton-proton collisions at √s = 13 TeV. Eur. Phys. J. C. Part. Fields 79, 94 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Tosti, L. et al. Mapping transcription factor occupancy using minimal numbers of cells in vitro and in vivo. Genome Res. 28, 592–605 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Monkhorst, K., Jonkers, I., Rentmeester, E., Grosveld, F. & Gribnau, J. X inactivation counting and choice is a stochastic process: evidence for involvement of an X-linked activator. Cell 132, 410–421 (2008).

    Article  CAS  PubMed  Google Scholar 

  40. Kim, D., Langmead, B. & Salzberg, S. L. HISAT: a fast spliced aligner with low memory requirements. Nat. Methods 12, 357–360 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Li, H. et al. The Sequence Alignment/Map format and SAMtools. Bioinformatics 25, 2078–2079 (2009).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  42. Ditzel, M. et al. Biologic meshes are not superior to synthetic meshes in ventral hernia repair: an experimental study with long-term follow-up evaluation. Surg. Endosc. 27, 3654–3662 (2013).

    Article  CAS  PubMed  Google Scholar 

  43. Aad, G. et al. Observation of associated near-side and away-side long-range correlations in √s NN = 5.02 TeV proton-lead collisions with the ATLAS detector. Phys. Rev. Lett. 110, 182302 (2013).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank the members of the Kind laboratory for their comments on the manuscript. S.S.D. acknowledges support from the Center for Scientific Computing at UCSB: an NSF MRSEC (DMR-1720256) and NSF CNS-1725797. S.S.D. was also supported by the NIH grant R01HG011013. This work was funded by a European Research Council Starting grant (ERC-StG 678423-EpiID), a Nederlandse Organisatie voor Wetenschappelijk Onderzoek37 Open (824.15.019) and ALW/VENI grant (016.181.013). The Oncode Institute is supported by KWF Dutch Cancer Society.

Author information

Authors and Affiliations

Authors

Contributions

K.R., S.S.D. and J.K. designed the study. S.S.D. developed the method with input and assistance from D.M. K.R. supervised and performed bioinformatic analyses and developed the scDam&T computational pipeline. F.J.R. performed cloning and bioinformatic analyses on mESC scDam&T data and developed the clonal selection strategy. C.M.M. optimized the method, performed experiments, generated cell lines and designed the protocol for scDamID2. S.S.d.V. generated cell lines, assisted with experiments and designed the protocol for bulk DamID2. S.J.A.L. generated cell lines. K.L.d.L. assisted with experiments. A.C. assisted with analyses. J.K. and S.S.D. conceived and supervised the study. C.M.M. and F.J.R. wrote the manuscript with input from J.K.

Corresponding authors

Correspondence to Siddharth S. Dey or Jop Kind.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Peer review information Nature Protocols thanks John Arne Dahl, Madeleine Fosslie, Tamar Hashimshony 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.

Related links

Key reference using this protocol

Rooijers, K. et al. Nat. Biotechnol. 37, 766–772 (2019): https://doi.org/10.1038/s41587-019-0150-y

Integrated supplementary information

Supplementary Fig. 1 Gating strategy for FACS.

a, Dot plot of ungated mESCs showing gating strategy to exclude debris in FSC (forward scatter) versus SSC (side scatter). The percentage of events in Gate 1 is indicated. b, Dot plot of mESCs passing Gate 1, showing the gating strategy to exclude dead cells in propidium iodide versus FSC. The percentage of events in Gate 2 is indicated. c, Dot plot of mESCs passing Gate 2, showing the gating strategy to exclude duplet cells in Hoechst versus Hoechst area. The percentage of events in Gate 3 is indicated. d, Dot plot of mESCs passing Gate 3 were gated for DNA content in G1 and G2/M phase of the cell cycle. The gate for the G2/M population was defined by doubling the intensity value of the G1 peak maximum. e, DNA content histogram events in Gate 3, showing counted events versus Hoechst area. Only cells passing gate G2/M were sorted. f, Table indicating gate hierarchy, percentage of events in each gate relative to parent population and total numbers of events within each gate. g, Table indicating the mean value for the G1 and G2/M populations. All measurements were done on the BD FACSJazz and analyzed with FlowJo software, version 10.1r5.

Supplementary information

Supplementary Information

Supplementary Fig. 1, Supplementary Methods, Supplementary Manual and Supplementary 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

Markodimitraki, C.M., Rang, F.J., Rooijers, K. et al. Simultaneous quantification of protein–DNA interactions and transcriptomes in single cells with scDam&T-seq. Nat Protoc 15, 1922–1953 (2020). https://doi.org/10.1038/s41596-020-0314-8

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41596-020-0314-8

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