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.

  • Letter
  • Published:

Simultaneous quantification of protein–DNA contacts and transcriptomes in single cells

Abstract

Protein–DNA interactions are critical to the regulation of gene expression, but it remains challenging to define how cell-to-cell heterogeneity in protein–DNA binding influences gene expression variability. Here we report a method for the simultaneous quantification of protein–DNA contacts by combining single-cell DNA adenine methyltransferase identification (DamID) with messenger RNA sequencing of the same cell (scDam&T-seq). We apply scDam&T-seq to reveal how genome–lamina contacts or chromatin accessibility correlate with gene expression in individual cells. Furthermore, we provide single-cell genome-wide interaction data on a polycomb-group protein, RING1B, and the associated transcriptome. Our results show that scDam&T-seq is sensitive enough to distinguish mouse embryonic stem cells cultured under different conditions and their different chromatin landscapes. Our method will enable the analysis of protein-mediated mechanisms that regulate cell-type-specific transcriptional programs in heterogeneous tissues.

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: Quantitative comparison of scDamID, CEL-Seq and scDam&T-seq applied to KBM7 cells.
Fig. 2: Untethered Dam marks accessible chromatin in single cells.
Fig. 3: Parallel transcriptomic and DamID measurements link transcriptional dependencies to heterogeneity in DamID contacts.
Fig. 4: scDam&T-seq enables in silico cell sorting and reconstruction of corresponding cell-type-specific gene-regulatory landscapes.

Similar content being viewed by others

Data availability

The sequencing data from this study are available from the Gene Expression Omnibus, accession number GSE108639.

Code availability

All computational code used for this study is available upon request.

References

  1. 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 

  2. 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 

  3. 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 

  4. 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 

  5. 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 

  6. 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 

  7. 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 

  8. 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 

  9. 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 

  10. 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 

  11. Mooijman, D. et al. 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 

  12. 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 (2017).

    Article  CAS  PubMed  Google Scholar 

  13. Wu, X. et al. 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 

  14. 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 

  15. Dey, S. et al. Integrated genome and transcriptome sequencing of the same cell. Nat. Biotechnol. 33, 285–289 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Macaulay, I. C. et al. G&T-seq: parallel sequencing of single-cell genomes and transcriptomes. Nat. Methods 12, 519–522 (2015).

    Article  CAS  PubMed  Google Scholar 

  17. 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 

  18. 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 

  19. 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  Google Scholar 

  20. Steensel van, B. et al. Chromatin profiling using targeted DNA adenine methyltransferase. Nat. Genet. 27, 304–308 (2001).

    Article  Google Scholar 

  21. Vogel, M. J. et al. Detection of in vivo protein–DNA interactions using DamID in mammalian cells. Nat. Protoc. 2, 1467–1478 (2007).

    Article  CAS  PubMed  Google Scholar 

  22. Kind, J. et al. Single-cell dynamics of genome–nuclear lamina interactions. Cell 153, 178–192 (2013).

    Article  CAS  PubMed  Google Scholar 

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

    Article  PubMed  PubMed Central  Google Scholar 

  24. Meuleman, W. et al. Constitutive nuclear lamina–genome interactions are highly conserved and associated with A/T-rich sequence. Genome Res. 23, 270–281 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Monkhorst, K. et al. 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 

  26. Nishimura, K. et al. 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 

  27. Peric-Hupkes, D. et al. Molecular maps of the reorganization of genome–nuclear lamina interactions during differentiation. Mol. Cell 38, 603–613 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Aughey, G. N. et al. CATaDa reveals global remodelling of chromatin accessibility during stem cell differentiation in vivo. eLife 7, e32341 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  29. Schones, D. E. et al. Dynamic regulation of nucleosome positioning in the human genome. Cell 132, 887–898 (2008).

    Article  CAS  PubMed  Google Scholar 

  30. Valouev, A. et al. Determinants of nucleosome organization in primary human cells. Nature 474, 516–520 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Boyle, A. P. et al. High-resolution mapping and characterization of open chromatin across the genome. Cell 132, 311–322 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Wang, H. et al. Role of histone H2A ubiquitination in polycomb silencing. Nature 431, 873–878 (2004).

    Article  CAS  PubMed  Google Scholar 

  33. Zylicz, J. J. et al. The implication of early chromatin changes in X chromosome inactivation. Cell 176, 182–197 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Bonev, B. et al. Multiscale 3D genome rewiring during mouse neural development. Cell 171, 557–572.e524 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Wang, Y. et al. The 3D Genome Browser: a web-based browser for visualizing 3D genome organization and long-range chromatin interactions. Genome Biol. 19, 151 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  36. Sakaue-Sawano, A. et al. Visualizing spatiotemporal dynamics of multicellular cell cycle progression. Cell 132, 487–498 (2008).

    Article  CAS  PubMed  Google Scholar 

  37. Keane, T. M. et al. Mouse genomic variation and its effect on phenotypes and gene regulation. Nature 477, 289–294 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Lun, A. T. et al. Pooling across cells to normalize single-cell RNA sequencing data with many zero counts. Genome Biol. 17, 75 (2016).

    Article  PubMed  Google Scholar 

  39. Lun, A. T. et al. A step-by-step workflow for low-level analysis of single-cell RNA-seq data with Bioconductor. F1000Res 5, 2122 (2016).

    PubMed  PubMed Central  Google Scholar 

  40. Boedigheimer, M. J. et al. Sources of variation in baseline gene expression levels from toxicogenomics study control animals across multiple laboratories. BMC Genomics 9, 285 (2008).

    Article  PubMed  PubMed Central  Google Scholar 

  41. Johnson, W. E. et al. Adjusting batch effects in microarray expression data using empirical Bayes methods. Biostatistics 1, 118–127 (2006).

    Google Scholar 

  42. Kent, W. J. et al. The human genome browser at UCSC. Genome Res. 12, 996–1006 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Ernst, J. et al. Mapping and analysis of chromatin state dynamics in nine human cell types. Nature 473, 43–49 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Knijnenburg, T. A. et al. Multiscale representation of genomic signals. Nat. Methods 11, 689–694 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Robinson, M. D. et al. edgeR: a Bioconductor package for differential expression analysis of digital gene expression data. Bioinformatics 26, 139–140 (2010).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We would like to thank the members of the Kind, Dey and van Oudenaarden laboratories for their comments on the manuscript and J. Gribnau (Erasmus UMC) for kindly providing the 129/Sv:CAST/EiJ mESCs and for advice on differentiation. We would like to thank B. de Barbanson and J. Yeung for suggestions regarding computational analyses and statistics, R. van der Linden for FACS and M. Muraro and L. Kester for input on the scDam&T-seq technique. S.S.D and A.C. received computational support from the Center of Scientific Computing at UCSB based on funding from NSF MRSEC (DMR-1720256) and NSF CNS-1725797. This work was funded by a European Research Council Starting grant (no. ERC-StG 678423-EpiID) and a Nederlandse Organisatie voor Wetenschappelijk Onderzoek (NWO) Open grant (no. 824.15.019) and ALW/VENI grant (no. 016.Veni.181.013). The Oncode Institute is supported by the KWF Dutch Cancer Society.

Author information

Authors and Affiliations

Authors

Contributions

K.R. and C.M.M. contributed equally as first authors. F.J.R. and S.S.d.V. contributed equally as second authors. K.R., S.S.D. and J.K. designed the study and wrote the manuscript. S.S.D. developed the method. C.M.M. optimized the method and performed all the experiments unless stated otherwise. S.S.d.V. and K.L.d.L. assisted with experiments. S.S.d.V. created the mESC lines. K.R. performed all analyses except when stated otherwise. F.J.R. performed cloning and all analyses pertaining to the RING1B data. A.C. performed the analysis of Supplementary Fig. 4d and exploratory analyses together with S.S.D. D.M. provided input during initial technology development. J.K. and S.S.D conceived and supervised the study.

Corresponding authors

Correspondence to Siddharth S. Dey or Jop Kind.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Publisher’s note: Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Integrated supplementary information

Supplementary Figure 1 Quantitative comparison between scDamID and scDam&T-seq.

a) Comparison between the binarized (OE >= 1) single cell (horizontal tracks) maps for scDamID and scDam&T-seq (horizontal tracks; both panels show 75 single-cell samples with highest sample depths). Each row represents a single cell; each column a 100-kb bin along the genome. Unmappable genomic regions are indicated in red along the top of the track. b) Comparison of scDamID and scDam&T-seq CFs. CF distributions are depicted in the margins. Pearson’s r and p-value are indicated. P-value indicates the result of a two-sided test (0 indicates a value smaller than 32-bit floating point precision, that is 1.18e-38) c) Raw signal (RPKM values) on LAD-boundaries, for both scDamID and scDam&T-seq. LAD positions were defined independently based on HT1080 cells (Genome Research 23, 270–281, 2013). d) Run-length frequencies of uninterrupted ‘OE >= 1’ runs for two Dam-LMNB1 clones (#14 and #5-5) and one Dam clone (#5–8) for both scDam&T-seq (top) and scDamID (bottom). Run-length frequencies of randomized matrices with preserved marginals (Nature communications 5, 4114, 2014) are shown in light colors. e) Pearson autocorrelation of raw signal (y-axis) vs genomic distance (x-axis) of in silico population samples for two Dam-LMNB1 clones and one Dam clone, measured with scDam&T-seq (top) and scDamID (bottom). f) Comparison of sample complexities obtained with scDam&T-seq (dark markers) and scDamID (light markers) for Dam-LMNB1 clones and one Dam clone. Unique detected GATCs are depicted on the y-axis vs. GATC-aligning reads (including duplicates) on the x-axis. g) Overview of losses during processing of raw sequencing data in scDamID and scDam&T-seq. Bars from left-to-right follow the order of the processing pipeline, where raw reads are first filtered on the correct adapter structure, then aligned to the human genome, where reads not yielding a unique alignment are filtered out, as well as reads not aligning immediately adjacent to GATCs. Finally, duplicate reads are removed, on account of the haploid nature of the KBM7 cell line.

Supplementary Figure 2 Quantitative comparison between CEL-Seq and scDam&T-seq.

a) Distributions of the number of unique transcripts detected using CEL-Seq (Cell 163, 134–147, 2015) and scDam&T-seq. b) Correlation in expression values (TPM, transcripts per million reads) between scDam&T-seq and CEL-Seq (left panel) and two scDam&T-seq libraries processed in parallel (right panel). Pearson’s correlation coefficient is indicated. P-value indicate the result of a two-sided test (0 indicates a value smaller than 32-bit floating point precision, ie. 1.18e-38). c) Correlation of fraction of cells (passing cutoffs) in which a gene was detected between scDam&T-seq and CEL-Seq (left panel) and two scDam&T-seq libraries processed in parallel (right panel). Pearson’s correlation. P-value indicates the result of a two-sided test (0 indicates a value smaller than 32-bit floating point precision, ie. 1.18e-38). d) Coefficient of variation (CV) of gene expression (y-axis) vs, mean expression values (x-axis), as measured by scDam&T-seq (4 libraries across 2 KBM7 clones) and CEL-Seq. The dotted line indicates the CV of Poisson-distributed data (CV = λ−1/2) e) Principal component analysis on normalized expression data obtained from CEL-Seq and scDam&T-seq (Dam-LMNB1 clone #14), where the first three principal components are shown, as well as correlation of PC1 with sample depth (number of unique transcripts detected). Numbers in parentheses indicate the fraction of data variance explained by the principal component. f) Overview of losses during processing of transcriptomic data obtained with CEL-Seq and scDam&T-seq. Bars from left-to-right follow the order of the processing pipeline, where raw reads are aligned to the human genome, reads that do not yield unique alignments are filtered, as well as reads that do not match exons. Finally, duplicate reads are removed based on the UMIs. g) Fraction of transcriptomic reads mapping uniquely to either gene introns, exons or to intergenic loci for scDam&T-seq (top) and CEL-Seq (bottom) in KBM7 samples. Error bars indicate a 95% confidence interval for the mean. Error bars indicate a 95% confidence interval of the mean (calculated by bootstrap procedure). n=315 for scDam&T-seq, n=87 for CELseq. h) Relation between number of unique transcripts detected (y-axis) and number of unique GATCs detected (x-axis) with scDam&T-seq for two DamID adapter concentrations. Pearson’s r and p-values (two-sided test) are indicated. The dotted line indicates a linear regression estimate, the shaded area indicates a 95% confidence interval of regression estimates (determined by bootstrap procedure).

Supplementary Figure 3 scDam&T-seq in hybrid mESCs.

a) Auxin-mediated control of AID-Dam (clone #c8) and AID-Dam-LMNB1(clone #b2) cell lines. DamID PCR products of cells 24 and 48 hours after auxin washout (left). Time course and quantitative PCR analysis of auxin induction for a locus within a LAD, 0-, 8-, 10-, 12- and 24 hours after auxin washout (right). Quantification of the m6A levels as described for the DpnII assay (Cell 153, 178–192, 2013). Dot-plot depicts the mean value of n = 2 independent experiments. b) mESC in silico population Dam-LMNB1 RPKM values projected on the starts and ends of LAD boundaries defined previously (Molecular cell 38, 603–613, 2010), for two different Dam-LMNB1 clones (#b2 and #e6) and a total of 166 single-cell samples.

Supplementary Figure 4 Untethered Dam enzyme marks accessible chromatin in single cells.

a) 10 bp resolution frequency spectrum of in silico population Dam signal stratified in four regimes of increasing CTCF binding activities (corresponding to Fig. 2d). The black vertical lines indicate 174 bp. b) Distribution of 20-kb bins as a function of bulk H3K4me3 (y-axis, left) or bulk H3K36me3 (y-axis, right) ChIP-seq and in silico population Dam data (x-axis). Pearson’s r and p-values are indicated. c) Percentage of methylation (for scNMT-seq) and RPKM values (for scDam&T-seq Dam signal) at DNaseI hypersensitivity sites, relates to Fig. 1d from Clark et al. (Nature communications 9, 781, 2018). Solid lines indicate the mean across single-cell samples while the shaded areas indicate the standard deviation of signals observed across single-cell samples. n=95 Dam-only single-cell samples, n=72 scNMT-seq single-samples. d) Number of unique genes observed with scNMT-seq and scDam&T-seq, using single-cell samples down sampled to 150,000 reads. Samples below cutoff were not considered for this analysis.

Supplementary Figure 5 Single-cell associations between transcription levels and variance, and Dam or Dam-LMNB1 contacts.

a) Relation between expression log2FC values and mean expression levels for Dam-LMNB1. See Fig. 3a for analysis of expression log2FC values. Solid line indicates the mean, shaded area indicates 1.96 times the standard deviation around the mean. b) As (a), but for Dam. c) Relation between expression log2FC values and expression variance-to-mean ratio for Dam-LMNB1. The variance-to-mean ratios were adjusted by controlling for mean expression levels, since the raw variance-to-mean values were not constant (nor linearly correlating) with mean expression levels (see methods for details). Solid line indicates the mean, shaded area indicates 1.96 times the standard deviation around the mean. d) As (c), but for Dam. e) Relative enrichment (min-max normalized) of several histone post-translational modifications (PTMs) in genomic regions with different CF values. f) The fraction of the genome coverage for (constitutive) cLADs, (facultative) fLADs, ciLADs and fiLADs over a range of CF values.

Supplementary Figure 6 Allelic associations between single-cell transcription and Dam contacts.

a) AluI signal obtained from 129/Sv:CAST/EiJ mESCs. Each row represents a single cell; each column a 100-kb bin along the genome. Red colors indicate an enrichment of signal compared to expected (OE, based on AluI-motif density) whereas blue colors indicate a depletion. The top cells with exclusive 129/Sv genomic annotations are likely a contamination of feeder cells (mouse embryonic fibroblasts). b) Dam signal from the same clone as in (a), on the maternal (129/Sv) and paternal (CAST/EiJ) alleles, and for 2i and serum, respectively. c) Example of a cell (marked with a blue X) which has no duplication of the paternal chromosome 12 (unlike the majority of the population), but harbors a duplication of the maternal chromosome 12 instead, observable in the Dam signals. This reciprocally corresponds to allele-specific transcription with approximately double the maternal level and half the paternal level, compared to the majority. d) Relation between allelic imbalance (‘bias’) of Dam signals (x-axis) and transcription (y-axis). Note that chromosomes 5, 8 and 12 (and sex chromosomes) seem frequently (partially) duplicated and were excluded from this analysis, as well as single-cell samples for which there was evidence of any CNV on any autosome (see methods). Pearson’s correlation is indicated. P-value indicate the result of a two-sided test. The solid line indicates a linear regression estimate, the shaded area indicates a 95% confidence interval of regression estimates (determined by bootstrap procedure).

Supplementary Figure 7 In silico identification of cell identities and corresponding regulatory landscapes with scDam&T-seq.

a) Normalized expression values for the top five down-regulated (left) and up-regulated (right) genes in 2i compared to serum. Box plots indicate the 25th and 75th percentile (box), and the median (line). Data points are overlaid as circles. n=61 and n=71 for serum and 2i conditions, respectively. b) Distribution of variances of differential 2i/serum accessibility between 100-kb bins within TADs (Cell 171, 557–572.e524, 2017, black line) and within randomly reordered TADs (50 iterations, green lines). P-values between variance distributions of the original and randomized TADs were calculated using a two-sided Mann-Whitney U test. Box plots indicate the 25th and 75th percentile (box), median (line) and 1.5 times the inter-quartile range (IQR) past the 25th and 75th percentiles (whiskers). c) Relation between log2 fold-change in Dam signal (RPM) values of in silico population samples (y-axis) and normalized transcription (x-axis) between 2i and serum conditions. Pearson’s correlation coefficient and p-value (two-sided test) are indicated. The solid line indicates a linear regression estimate, the shaded area indicates a 95% confidence interval of regression estimates (determined by bootstrap procedure). d) Dam OE values of in silico population samples at TSSs of down-regulated (left), upregulated (middle), or unaffected (right) genes in 2i (orange) compared to serum (blue). e) Dam OE values measured in single cells at TSSs of the top 5 down-regulated (left) and up-regulated (right) genes in 2i compared to serum. Box plots indicate the 25th and 75th percentile (box), and the median (line). Data points are overlaid as circles. f) Genome-wide correlation between RING1B ChIP-seq (bulk, input normalized) and DamID (62 cells, Dam normalized) in 100-kb bins. Spearman’s rho and p-value (two-sided test, determined by bootstrap) are indicated. Chromosomes 12, X and Y were excluded from the analysis. g) Transcriptional allelic bias on chromosome X (blue) compared to the allelic bias on the somatic chromosomes (green, downsampled to chromosome X coverage). Box plots indicate the 25th and 75th percentile (box), median (line) and 1.5 times the inter-quartile range (IQR) past the 25th and 75th percentiles (whiskers). Data points are overlaid as circles. h) Average allelic profiles of RING1B scDam&T-seq signal on chromosome 6 for cells that show a transcriptional bias on chromosome X towards neither allele (top), towards 129/Sv (middle), or towards CAST/EiJ (bottom). i) H2AK119Ub ChIP-seq data from GSM3267034 (Cell 176, 182–197, 2019) showing allele-specific signals upon X-inactivation (left panel). Comparison of allelic imbalance (ratio maternal over total) between DamID signals measured in cells showing either maternal or paternal X-inactivation, to H2AK119Ub allelic imbalance, per 100-kb bin. Pearson’s correlation coefficient and p-value (two-sided test) are indicated in the right panels. The solid line indicates a linear regression estimate, the shaded area indicates a 99% confidence interval of regression estimates (determined by bootstrap procedure).

Supplementary information

Supplementary information

Supplementary Figs. 1–7 and Supplementary Tables 1–4

Reporting Summary

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Rooijers, K., Markodimitraki, C.M., Rang, F.J. et al. Simultaneous quantification of protein–DNA contacts and transcriptomes in single cells. Nat Biotechnol 37, 766–772 (2019). https://doi.org/10.1038/s41587-019-0150-y

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41587-019-0150-y

This article is cited by

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