Abstract
Technologies that profile chromatin modifications at single-cell resolution offer enormous promise for functional genomic characterization, but the sparsity of the measurements and integrating multiple binding maps represent substantial challenges. Here we introduce single-cell (sc)CUT&Tag-pro, a multimodal assay for profiling protein–DNA interactions coupled with the abundance of surface proteins in single cells. In addition, we introduce single-cell ChromHMM, which integrates data from multiple experiments to infer and annotate chromatin states based on combinatorial histone modification patterns. We apply these tools to perform an integrated analysis across nine different molecular modalities in circulating human immune cells. We demonstrate how these two approaches can characterize dynamic changes in the function of individual genomic elements across both discrete cell states and continuous developmental trajectories, nominate associated motifs and regulators that establish chromatin states and identify extensive and cell-type-specific regulatory priming. Finally, we demonstrate how our integrated reference can serve as a scaffold to map and improve the interpretation of additional scCUT&Tag datasets.
This is a preview of subscription content, access via your institution
Relevant articles
Open Access articles citing this article.
-
Multimodal chromatin profiling using nanobody-based single-cell CUT&Tag
Nature Biotechnology Open Access 19 December 2022
-
Multifactorial profiling of epigenetic landscapes at single-cell resolution using MulTI-Tag
Nature Biotechnology Open Access 31 October 2022
Access options
Access Nature and 54 other Nature Portfolio journals
Get Nature+, our best-value online-access subscription
$29.99 / 30 days
cancel any time
Subscribe to this journal
Receive 12 print issues and online access
$209.00 per year
only $17.42 per issue
Rent or buy this article
Get just this article for as long as you need it
$39.95
Prices may be subject to local taxes which are calculated during checkout





Data availability
Data generated for this manuscript have been deposited in the Gene Expression Omnibus with accession code GSE195725. The processed datasets are available as open-access downloads at https://zenodo.org/record/5504061. The following public datasets were used in this study: GSM4732109 and GSE164378 (Figs. 2,4 and Supplementary Fig. 3); GSM4732123 (Fig. 1d); GSM1220567, GSM1220569, GSM1027296 and GSM1102793 (Fig. 1g); and GSM5034342 and GSM5034344 (Supplementary Figs. 1c and Fig. 5). Datasets used in differential expression analysis with the bulk total RNA-seq were http://r2platform.com/rna_atlas and https://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=GSE138734.
Code availability
Seurat, Signac and scChromHMM are freely available as open-source software packages at https://github.com/satijalab/seurat, https://github.com/timoast/signac and https://github.com/satijalab/scchromhmm, respectively.
References
Tang, F. et al. mRNA-seq whole-transcriptome analysis of a single cell. Nat. Methods 6, 377–382 (2009).
Jaitin, D. A. et al. Massively parallel single-cell RNA-seq for marker-free decomposition of tissues into cell types. Science 343, 776–779 (2014).
Macosko, E. Z. et al. Highly parallel genome-wide expression profiling of individual cells using nanoliter droplets. Cell 161, 1202–1214 (2015).
Hashimshony, T., Wagner, F., Sher, N. & Yanai, I. CEL-Seq: single-cell RNA-seq by multiplexed linear amplification. Cell Rep. 2, 666–673 (2012).
Picelli, S. et al. Smart-seq2 for sensitive full-length transcriptome profiling in single cells. Nat. Methods 10, 1096–1098 (2013).
Shalek, A. K. et al. Single-cell transcriptomics reveals bimodality in expression and splicing in immune cells. Nature 498, 236–240 (2013).
Buenrostro, J. D. et al. Single-cell chromatin accessibility reveals principles of regulatory variation. Nature 523, 486–490 (2015).
Cusanovich, D. A. et al. Multiplex single cell profiling of chromatin accessibility by combinatorial cellular indexing. Science 348, 910–914 (2015).
ENCODE Project Consortium. A user’s guide to the encyclopedia of DNA elements (ENCODE). PLoS Biol. 9, e1001046 (2011).
Hoffman, M. M. et al. Unsupervised pattern discovery in human chromatin structure through genomic segmentation. Nat. Methods 9, 473–476 (2012).
Bannister, A. J. & Kouzarides, T. Regulation of chromatin by histone modifications. Cell Res. 21, 381–395 (2011).
Ernst, J. & Kellis, M. Discovery and characterization of chromatin states for systematic annotation of the human genome. Nat. Biotechnol. 28, 817–825 (2010).
Strahl, B. D. & Allis, C. D. The language of covalent histone modifications. Nature 403, 41–45 (2000).
Jenuwein, T. & Allis, C. D. Translating the histone code. Science 293, 1074–1080 (2001).
Park, P. J. ChIP-seq: advantages and challenges of a maturing technology. Nat. Rev. Genet. 10, 669–680 (2009).
Kaya-Okur, H. S. et al. CUT&Tag for efficient epigenomic profiling of small samples and single cells. Nat. Commun. 10, 1930 (2019).
Ernst, J. & Kellis, M. ChromHMM: automating chromatin-state discovery and characterization. Nat. Methods 9, 215–216 (2012).
Bartosovic, M., Kabbe, M. & Castelo-Branco, G. Single-cell CUT&Tag profiles histone modifications and transcription factors in complex tissues. Nat. Biotechnol. 39, 825–835 (2021).
Wu, S. J. et al. Single-cell CUT&Tag analysis of chromatin modifications in differentiation and tumor progression. Nat. Biotechnol. 39, 819–824 (2021).
Wang, Q. et al. CoBATCH for high-throughput single-cell epigenomic profiling. Mol. Cell 76, 206–216 (2019).
Carter, B. et al. Mapping histone modifications in low cell number and single cells using antibody-guided chromatin tagmentation (ACT-seq). Nat. Commun. 10, 3747 (2019).
Zhu, C. et al. Joint profiling of histone modifications and transcriptome in single cells from mouse brain. Nat. Methods 18, 283–292 (2021).
Xiong, H. et al. Single-cell joint detection of chromatin occupancy and transcriptome enables higher-dimensional epigenomic reconstructions. Nat. Methods 18, 652–660 (2021).
Zhou, V. W., Goren, A. & Bernstein, B. E. Charting histone modifications and the functional organization of mammalian genomes. Nat. Rev. Genet. 12, 7–18 (2011).
Suganuma, T. & Workman, J. L. Signals and combinatorial functions of histone modifications. Annu. Rev. Biochem. 80, 473–499 (2011).
Stoeckius, M. et al. Simultaneous epitope and transcriptome measurement in single cells. Nat. Methods 14, 865–868 (2017).
Peterson, V. M. et al. Multiplexed quantification of proteins and transcripts in single cells. Nat. Biotechnol. 35, 936–939 (2017).
Mimitou, P. et al. Scalable, multimodal profiling of chromatin accessibility, gene expression and protein levels in single cells. Nat. Biotechnol. 39, 1246–1258 (2021).
Swanson, E. et al. Simultaneous trimodal single-cell measurement of transcripts, epitopes, and chromatin accessibility using TEA-seq. Elife 10, e63632 (2021).
Hao, Y. et al. Integrated analysis of multimodal single-cell data. Cell 184, 3573–3587 (2021).
Nathan, A. et al. Multimodally profiling memory T cells from a tuberculosis cohort identifies cell state associations with demographics, environment and disease. Nat. Immunol. 22, 781–793 (2021).
Pombo Antunes, A. R. et al. Single-cell profiling of myeloid cells in glioblastoma across species and disease stage reveals macrophage competition and specialization. Nat. Neurosci. 24, 595–610 (2021).
Lareau, C. A. et al. Droplet-based combinatorial indexing for massive-scale single-cell chromatin accessibility. Nat. Biotechnol. 37, 916–924 (2019).
Stoeckius, M. et al. Cell hashing with barcoded antibodies enables multiplexing and doublet detection for single cell genomics. Genome Biol. 19, 224 (2018).
Bernstein, B. E. et al. The NIH Roadmap Epigenomics Mapping Consortium. Nat. Biotechnol. 28, 1045–1048 (2010).
Durbin, R., Eddy, S. R., Krogh, A. & Mitchison, G. Biological Sequence Analysis: Probabilistic Models of Proteins and Nucleic Acids (Cambridge Univ. Press, 1998).
Dann, E., Henderson, N. C., Teichmann, S. A., Morgan, M. D. & Marioni, J. C. Differential abundance testing on single-cell data using k-nearest neighbor graphs. Nat. Biotechnol. 40, 245–253 (2022).
Stuart, T. et al. Comprehensive inteÿgration of single-cell data. Cell 177, 1888–1902 (2019).
Smit, A. F. A., Hubley, R. & Green, P. RepeatMasker Open-4.0 (ISB, 2013); http://www.repeatmasker.org
Schep, A. N., Wu, B., Buenrostro, J. D. & Greenleaf, W. J. chromVAR: inferring transcription-factor-associated accessibility from single-cell epigenomic data. Nat. Methods 14, 975–978 (2017).
Szabo, S. J. et al. A novel transcription factor, T-bet, directs Th1 lineage commitment. Cell 100, 655–669 (2000).
Kallies, A. & Good-Jacobson, K. L. Transcription factor T-bet orchestrates lineage development and function in the immune system. Trends Immunol. 38, 287–297 (2017).
Zhao, X., Shan, Q. & Xue, H.-H. TCF1 in T cell immunity: a broadened frontier. Nat. Rev. Immunol. https://doi.org/10.1038/s41577-021-00563-6 (2021).
Lorenzi, L. et al. The RNA Atlas expands the catalog of human non-coding RNAs. Nat. Biotechnol. 39, 1453–1465 (2021).
Ashburner, M. et al. Gene Ontology: tool for the unification of biology. Nat. Genet. 25, 25–29 (2000).
Roh, T.-Y., Cuddapah, S., Cui, K. & Zhao, K. The genomic landscape of histone modifications in human T cells. Proc. Natl Acad. Sci. USA 103, 15782–15787 (2006).
Wei, G. et al. Global mapping of H3K4me3 and H3K27me3 reveals specificity and plasticity in lineage fate determination of differentiating CD4+ T cells. Immunity 30, 155–167 (2009).
Schoenborn, J. R. et al. Comprehensive epigenetic profiling identifies multiple distal regulatory elements directing transcription of the gene encoding interferon-gamma. Nat. Immunol. 8, 732–742 (2007).
Cui, K. et al. Chromatin signatures in multipotent human hematopoietic stem cells indicate the fate of bivalent genes during differentiation. Cell Stem Cell 4, 80–93 (2009).
Barshan, E., Ghodsi, A., Azimifar, Z. & Zolghadri Jahromi, M. Supervised principal component analysis: visualization, classification and regression on subspaces and submanifolds. Pattern Recognit. 44, 1357–1371 (2011).
Gopalan, S. et al. Simultaneous profiling of multiple chromatin proteins in the same cells. Mol. Cell 81, 4736–4746 (2021).
Meers, M. P., Janssens, D. H. & Henikoff, S. Multifactorial chromatin regulatory landscapes at single cell resolution. Preprint at bioRxiv https://doi.org/10.1101/2021.07.08.451691 (2021).
Mellis, I. A. et al. Responsiveness to perturbations is a hallmark of transcription factors that maintain cell identity in vitro.Cell Syst. 12, 885–899 (2021).
Shim, W. J. et al. Conserved epigenetic regulatory logic infers genes governing cell identity. Cell Syst. 11, 625–639 (2020).
La Manno, G. et al. RNA velocity of single cells. Nature 560, 494–498 (2018).
Srivastava, A., Malik, L., Smith, T., Sudbery, I. & Patro, R. Alevin efficiently estimates accurate gene abundances from dscRNA-seq data. Genome Biol. 20, 65 (2019).
Stuart, A. et al. Multimodal single-cell chromatin analysis with Signac. Nat. Methods 18, 1333–1341 (2021).
Cao, J. et al. The single-cell transcriptional landscape of mammalian organogenesis. Nature 566, 496–502 (2019).
Hafemeister, C. & Satija, R. Normalization and variance stabilization of single-cell RNA-seq data using regularized negative binomial regression. Genome Biol. 20, 296 (2019).
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).
Acknowledgements
We thank all the members of the Satija Lab for thoughtful discussions related to this work. B.Z. is a postdoctoral fellow of the Jane Coffin Childs Memorial Fund for Medical Research. This investigation has been aided by a grant from the Jane Coffin Childs Memorial Fund for Medical Research. This work was supported by the Chan Zuckerberg Initiative (grants EOSS-0000000082 and HCA-A-1704-01895 to R.S.) and the National Institutes of Health (grant K99CA267677-01 to A.S.; grant K99HG011489-01 to T.S.; and grants RM1HG011014-02, 1OT2OD026673-01 and DP2HG009623-01 to R.S.).
Author information
Authors and Affiliations
Contributions
B.Z., A.S. and R.S. conceived the study. A.S., B.Z., T.S. and Y.H. performed computational work supervised by R.S.; B.Z., E.M. and I.R. performed experimental work supervised by R.S. All authors participated in interpretation and writing the manuscript.
Corresponding author
Ethics declarations
Competing interests
In the past three years, R.S. has worked as a consultant for Bristol Myers Squibb, Regeneron and Kallyope and served as scientific advisory board member for ImmunAI, Resolve Biosciences, NanoString and the NYC Pandemic Response Lab. P.S. is co-inventor of a patent related to this work. The other authors declare no competing interests.
Peer review
Peer review information
Nature Biotechnology thanks Barbara Treutlein, Goncalo Castelo-Branco, Anoop Patel and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.
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. 1–6 and Tables 1–3.
Rights and permissions
About this article
Cite this article
Zhang, B., Srivastava, A., Mimitou, E. et al. Characterizing cellular heterogeneity in chromatin state with scCUT&Tag-pro. Nat Biotechnol 40, 1220–1230 (2022). https://doi.org/10.1038/s41587-022-01250-0
Received:
Accepted:
Published:
Issue Date:
DOI: https://doi.org/10.1038/s41587-022-01250-0