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:

ISSAAC-seq enables sensitive and flexible multimodal profiling of chromatin accessibility and gene expression in single cells

Abstract

Joint profiling of chromatin accessibility and gene expression from the same single cell provides critical information about cell types in a tissue and cell states during a dynamic process. Here, we develop in situ sequencing hetero RNA–DNA-hybrid after assay for transposase-accessible chromatin-sequencing (ISSAAC-seq), a highly sensitive and flexible single-cell multi-omics method to interrogate chromatin accessibility and gene expression from the same single nucleus. We demonstrated that ISSAAC-seq is sensitive and provides high quality data with orders of magnitude more features than existing methods. Using the joint profiles from over 10,000 nuclei from the mouse cerebral cortex, we uncovered major and rare cell types and cell-type specific regulatory elements and identified heterogeneity at the chromatin level within established cell types defined by gene expression. Finally, we revealed distinct dynamics and relationships of gene expression and chromatin accessibility during an oligodendrocyte maturation trajectory.

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: Joint profiling of chromatin accessibility and gene expression using ISSAAC-seq.
Fig. 2: Simultaneous investigation of chromatin accessibility and gene expression from the mouse cerebral cortex using ISSAAC-seq.
Fig. 3: Joint analysis of RNA and ATAC from the same cell in the mouse cortex.

Similar content being viewed by others

Data availability

The raw sequencing data generated in this study has been deposited at ArrayExpress under the accession number E-MTAB-11264. External data from previously published studies are available from the Gene Expression Omnibus: sci-CAR-seq (GSE117089), SNARE-seq (GSE126074), Paired-seq (GSE130399) and SHARE-seq (GSE140203). Source data are provided with this paper.

Code availability

The code used for the data processing and analysis mentioned in the method section is available on the GitHub repository https://github.com/dbrg77/ISSAAC-seq.

References

  1. Tang, F. et al. mRNA-seq whole-transcriptome analysis of a single cell. Nat. Methods 6, 377–382 (2009).

    Article  CAS  PubMed  Google Scholar 

  2. 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 

  3. 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 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Cao, J. et al. The single-cell transcriptional landscape of mammalian organogenesis. Nature 566, 496–502 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Hagemann-Jensen, M. et al. Single-cell RNA counting at allele and isoform resolution using Smart-seq3. Nat. Biotechnol. 38, 708–714 (2020).

    Article  CAS  PubMed  Google Scholar 

  7. 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 

  8. 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 

  9. Chen, X., Miragaia, R. J., Natarajan, K. N. & Teichmann, S. A. A rapid and robust method for single cell chromatin accessibility profiling. Nat. Commun. 9, 5345 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Chen, X. et al. Joint single-cell DNA accessibility and protein epitope profiling reveals environmental regulation of epigenomic heterogeneity. Nat. Commun. 9, 4590 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  11. Lake, B. B. et al. Integrative single-cell analysis of transcriptional and epigenetic states in the human adult brain. Nat. Biotechnol. 36, 70–80 (2018).

    Article  CAS  PubMed  Google Scholar 

  12. Satpathy, A. T. et al. Massively parallel single-cell chromatin landscapes of human immune cell development and intratumoral T cell exhaustion. Nat. Biotechnol. 37, 925–936 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Lareau, C. A. et al. Droplet-based combinatorial indexing for massive-scale single-cell chromatin accessibility. Nat. Biotechnol. 37, 916–924 (2019).

    Article  CAS  PubMed  Google Scholar 

  14. Stoeckius, M. et al. Simultaneous epitope and transcriptome measurement in single cells. Nat. Methods 14, 865–868 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Peterson, V. M. et al. Multiplexed quantification of proteins and transcripts in single cells. Nat. Biotechnol. 35, 936–939 (2017).

    Article  CAS  PubMed  Google Scholar 

  16. Stuart, T. & Satija, R. Integrative single-cell analysis. Nat. Rev. Genet. 20, 257–272 (2019).

    Article  CAS  PubMed  Google Scholar 

  17. Hao, Y. et al. Integrated analysis of multimodal single-cell data. Cell 184, 3573–3587.e29 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Argelaguet, R., Cuomo, A. S. E., Stegle, O. & Marioni, J. C. Computational principles and challenges in single-cell data integration. Nat. Biotechnol. 39, 1202–1215 (2021).

    Article  CAS  PubMed  Google Scholar 

  19. Zhu, C. et al. An ultra high-throughput method for single-cell joint analysis of open chromatin and transcriptome. Nat. Struct. Mol. Biol. 26, 1063–1070 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Ma, S. et al. Chromatin potential identified by shared single-cell profiling of RNA and chromatin. Cell 183, 1103–1116.e20 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Liu, L. et al. Deconvolution of single-cell multi-omics layers reveals regulatory heterogeneity. Nat. Commun. 10, 470 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Chen, S., Lake, B. B. & Zhang, K. High-throughput sequencing of the transcriptome and chromatin accessibility in the same cell. Nat. Biotechnol. 37, 1452–1457 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Cao, J. et al. Joint profiling of chromatin accessibility and gene expression in thousands of single cells. Science 361, eaau0730 (2018).

    Article  Google Scholar 

  24. Zhu, C. et al. Joint profiling of histone modifications and transcriptome in single cells from mouse brain. Nat. Methods 18, 283–292 (2021).

  25. Mimitou, E. P. et al. Scalable, multimodal profiling of chromatin accessibility, gene expression and protein levels in single cells. Nat. Biotechnol. 39, 1246–1258 (2021).

  26. 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 

  27. Swanson, E. et al. Simultaneous trimodal single-cell measurement of transcripts, epitopes, and chromatin accessibility using TEA-seq. eLife 10, e63632 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Xiong, H., Luo, Y., Wang, Q., Yu, X. & He, A. Single-cell joint detection of chromatin occupancy and transcriptome enables higher-dimensional epigenomic reconstructions. Nat. Methods 18, 652–660 (2021).

    Article  CAS  PubMed  Google Scholar 

  29. Guo, F. et al. Single-cell multi-omics sequencing of mouse early embryos and embryonic stem cells. Cell Res. 27, 967–988 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Rooijers, K. et al. scDam&T‐seq combines DNA adenine methyltransferase-based labeling of protein-DNA contact sites with transcriptome sequencing to analyze regulatory programs in single cells. Nat. Biotechnol. 37, 766–772 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Xing, Q. R. et al. Parallel bimodal single-cell sequencing of transcriptome and chromatin accessibility. Genome Res 30, 1027–1039 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Di, L. et al. RNA sequencing by direct tagmentation of RNA/DNA hybrids. Proc. Natl Acad. Sci. USA 117, 2886–2893 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Lu, B. et al. Transposase assisted tagmentation of RNA/DNA hybrid duplexes. eLife 9, e54919 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Xu, W. et al. A plate-based single-cell ATAC-seq workflow for fast and robust profiling of chromatin accessibility. Nat. Protoc. 16, 4084–4107 (2021).

    Article  CAS  PubMed  Google Scholar 

  35. Rosenberg, A. B. et al. Single-cell profiling of the developing mouse brain and spinal cord with split-pool barcoding. Science 360, eaam8999 (2018).

    Article  Google Scholar 

  36. Ding, J. et al. Systematic comparison of single-cell and single-nucleus RNA-sequencing methods. Nat. Biotechnol. 38, 737–746 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Becht, E. et al. Dimensionality reduction for visualizing single-cell data using UMAP. Nat. Biotechnol. 37, 38–44 (2019).

    Article  CAS  Google Scholar 

  38. 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 

  39. Stuart, T. et al. Comprehensive integration of single-cell data. Cell 177, 1888–1902.e21 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. 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 

  41. Aibar, S. et al. SCENIC: single-cell regulatory network inference and clustering. Nat. Methods 14, 1083–1086 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Factor, D. C. et al. Epigenomic comparison reveals activation of ‘seed’ enhancers during transition from naive to primed pluripotency. Cell Stem Cell 14, 854–863 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Bevington, S. L. et al. Inducible chromatin priming is associated with the establishment of immunological memory in T cells. EMBO J. 35, 515–535 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Ziffra, R. S. et al. Single-cell epigenomics reveals mechanisms of human cortical development. Nature 598, 205–213 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Qiu, X. et al. Reversed graph embedding resolves complex single-cell trajectories. Nat. Methods 14, 979–982 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Rop, F. V. D. et al. Hydrop enables droplet-based single-cell ATAC-seq and single-cell RNA-seq using dissolvable hydrogel beads. eLife 11, e73971 (2022).

    Article  PubMed  PubMed Central  Google Scholar 

  47. Chen, A. F. et al. NEAT-seq: simultaneous profiling of intra-nuclear proteins, chromatin accessibility and gene expression in single cells. Nat. Methods 19, 547–553 (2022).

    Article  CAS  PubMed  Google Scholar 

  48. Renaud, G., Stenzel, U., Maricic, T., Wiebe, V. & Kelso, J. deML: robust demultiplexing of Illumina sequences using a likelihood-based approach. Bioinformatics 31, 770–772 (2015).

    Article  CAS  PubMed  Google Scholar 

  49. Köster, J. & Rahmann, S. Snakemake—a scalable bioinformatics workflow engine. Bioinformatics 28, 2520–2522 (2012).

    Article  PubMed  Google Scholar 

  50. Li, H. & Durbin, R. Fast and accurate short read alignment with Burrows–Wheeler transform. Bioinformatics 25, 1754–1760 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Kaminow, B., Yunusov, D. & Dobin, A. STARsolo: accurate, fast and versatile mapping/quantification of single-cell and single-nucleus RNA-seq data. Preprint at bioRxiv https://doi.org/10.1101/2021.05.05.442755 (2021).

  52. Zhang, H. et al. Fast alignment and preprocessing of chromatin profiles with Chromap. Nat. Commun. 12, 6566 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Waltman, L. & Eck, N. Jvan A smart local moving algorithm for large-scale modularity-based community detection. Eur. Phys. J. B. 86, 471 (2013).

    Article  Google Scholar 

  54. Wolf, F. A., Angerer, P. & Theis, F. J. SCANPY: large-scale single-cell gene expression data analysis. Genome Biol. 19, 15 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  55. Stuart, T., Srivastava, A., Madad, S., Lareau, C. A. & Satija, R. Single-cell chromatin state analysis with Signac. Nat. Methods 18, 1333–1341 (2021).

  56. Zhang, Y. et al. Model-based analysis of ChIP-seq (MACS). Genome Biol. 9, R137 (2008).

    Article  PubMed  PubMed Central  Google Scholar 

  57. Heinz, S. et al. Simple combinations of lineage-determining transcription factors prime cis-regulatory elements required for macrophage and B cell identities. Mol. Cell 38, 576–589 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank all members from the Chen and Jin laboratories for the helpful discussion of the project. We also thank W. Chen and H. Cui from SUSTech for the helpful advice on the experiments. We thank X. Lu from SUSTech Core Research Facilities for the excellent support of FACS. The computational work was supported by Center for Computational Science and Engineering at Southern University of Science and Technology. Figures 1a and 2a were created with BioRender.com. This study was supported by National Key R&D Program of China (grant no. 2021YFF1200900 to W.J., X.C. and N.H.), National Natural Science Foundation of China (grant nos. 32170646 and 81872330 to W.J.), Guangdong Basic and Applied Basic Research Foundation (grant no. 2021B1515120070 to X.C.), Shenzhen Innovation Committee of Science and Technology (grant no. ZDSYS20200811144002008 to Shenzhen Key Laboratory of Gene Regulation and Systems Biology, to K.S., W.J. and X.C.), Shenzhen Science and Technology Program (grant no. KQTD20180411143432337 to W.J.), Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions (grant no. 2021SHIBS0002 to K.S.) and Cambridge-SU Joint Research Center for Genomic Resources (grant no. 2017B01012 to X.C.). The funders had no role in study design, data collection and analysis, decision to publish or preparation of the manuscript.

Author information

Authors and Affiliations

Authors

Contributions

X.C. and W.J. conceived the project. X.C., W.X. and W.J. designed the protocol and experiments. W.X. performed the experiments with help from Y.C., N.H., Q.Z., X.W. and Y.H. W.Y. carried out the computational analysis with help from Y.Z., W.X., X.C., W.J. and K.S. X.C., W.J., K.S. and N.H. supervised the entire project. All authors contributed to the writing.

Corresponding authors

Correspondence to Wenfei Jin or Xi Chen.

Ethics declarations

Competing interests

W.X., N.H., W.J. and X.C. have filed a patent application related to this work. The remaining authors declare no competing interests.

Peer review

Peer review information

Nature Methods thanks Ansuman Satpathy, Caleb Lareau, Kun Zhang and the other, anonymous, reviewer(s) for their contribution to the peer review of this work. Peer reviewer reports are available. Primary Handling Editor: Lei Tang, in collaboration with the Nature Methods team.

Additional information

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

Extended data

Extended Data Fig. 1 The plate-based (FACS) ISSAAC-seq workflow.

Schematic view of the plate-based (FACS) ISSAAC-seq workflow and adaptor configurations.

Extended Data Fig. 2 The droplet-based ISSAAC-seq workflow.

Schematic view of the droplet-based ISSAAC-seq workflow and adaptor configurations using 10x Chromium.

Extended Data Fig. 3 Experimental tests and benchmarking of ISSAAC-seq.

a and b, Assessment of ISSAAC-seq RNA data. Distributions of total UMIs (a) and numbers of detected genes (b) from indicated conditions performed on the E14 mouse embryonic stem cells. Pro, Protector RNase inhibitor. Ribo, RiboLock RNase inhibitors. c-e, Assessment of ISSAAC-seq ATAC data. Distributions of unique nuclear fragments (c), fraction of reads in peaks (FRiP) (d) and mitochondrial content (e) from indicated conditions performed on the E14 mouse embryonic stem cells. Boxplots show median with the top and bottom edges of the box representing the 75th and 25th percentiles, respectively. The whiskers represent the most extreme values that are within 1.5 times the interquartile range of the 25th and 75th percentiles. Outliers outside the whiskers are not shown. f, UCSC genome browser tracks of aggregated scATAC-seq signal from indicated conditions. g, Species mixing experiments of ISSAAC-seq using HEK293T and NIH3T3 cells.

Extended Data Fig. 4 ISSAAC-seq experimental results from different cell lines and PBMCs.

a, Examples of a typical FACS based workflow using K562 cells. FSC-A and SSC-A were used to select intact nuclei (P1). FSC-A and FSC-W were used to select singlets (P2). DAPI-A was used to select DAPI positive nuclei (P3). b, DNA electrophoresis of typical ISSAAC-seq libraries from the FACS (left) and droplet (right) workflow using K562 cells. c and d, Fragment size (c) and Tn5 insertion frequency (d) of ATAC data using different methods in the indicated cell lines. e, Quality metrics of the ISSAAC-seq PBMC data. f, Genomic distribution of reads from RNA data using different methods. g, Distributions of the numbers of UMIs using different methods performed in the indicated cell lines and PBMCs. Boxplots show median with the top and bottom edges of the box representing the 75th and 25th percentiles, respectively. The whiskers represent the most extreme values that are within 1.5 times the interquartile range of the 25th and 75th percentiles. Outliers outside the whiskers are not shown. The exact number of cells can be found in the Source Data.

Extended Data Fig. 5 UCSC genome browser tracks of single cell aggregates from ISSAAC-seq.

a, Visual comparisons of SHARE-seq, ISSAAC-seq and the 10x Multiome kit from K562 cells. Cell numbers are the same as in Fig. 1b. b, Visualisation of ATAC and RNA signals from single cell aggregates of ISSAAC-seq (FACS) in H1 (Day 3 upon mesoderm differentiation) and E14 (Day 4 upon EB differentiation) cells around the BRACHYURY and MIXL1 genes. c, UMAP projection of ISSAAC-seq PBMCs.

Extended Data Fig. 6 Computational pipeline for the data processing of droplet-based ISSAAC-seq.

a, Flow chart of the computational pipeline to process mouse cortex data. b, Cell barcode qualities and the number of nuclei returned by the pipeline in each step.

Extended Data Fig. 7 ISSAAC-seq ATAC and RNA data comparison.

a, UMAP projection of mouse cortex cells with ISSAAC-seq ATAC data colored by ATAC clusters (left) and batches (right). b, Ribbon plot showed the relationship between all RNA clusters and all ATAC clusters. c, UMAP projection of mouse cortex cells with ISSAAC-seq RNA data, highlighted by RNA cluster R2 Ex-L4/5 IT neurons (left) and ATAC cluster A2, A3 and A9 (right). d, UMAP projection of Ex-L4/5 IT neurons (RNA cluster R2) colored by RNA subclusters (left) and ATAC clusters. e, Gene ontology of the top 50 differentially expressed genes among A2, A3 and A9 clusters within the Ex-L4/5 IT neurons. Hypergeometric tests were used to derive the p-values, and the Benjamini-Hochberg Procedure was used to adjust multiple comparisons. f, SCENIC analysis of regulon (TF) activity in A2, A3 and A9 cells.

Extended Data Fig. 8 The prediction probability of each single cell based on the Allen Brain reference data using the label transfer function from Seurat.

Each row is a predicted label. Each column is a cell. The cluster identify of each cell is indicated at the bottom using the same colour code as in Fig. 2c.

Extended Data Fig. 9 UMAP projection of ISSAAC-seq RNA data coloured by the expression levels of indicated genes or batches.

A total of 24 marker genes are shown. Batch (biological replicates) information is shown in the bottom right plot.

Supplementary information

Supplementary Information

Supplementary Tables 1 and 2, Notes 1–4, References and Protocols 1 and 2.

Reporting Summary

Peer Review File

Supplementary Tables

Supplementary Tables 3 and 4.

Source data

Source Data Fig. 1

Source data for the boxplot and the violinplot in Fig. 1c,d.

Source Data Fig. 2

Source data for the boxplot and the violinplot in Fig. 2b.

Rights and permissions

Springer Nature or its licensor holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Xu, W., Yang, W., Zhang, Y. et al. ISSAAC-seq enables sensitive and flexible multimodal profiling of chromatin accessibility and gene expression in single cells. Nat Methods 19, 1243–1249 (2022). https://doi.org/10.1038/s41592-022-01601-4

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41592-022-01601-4

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