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:

Joint profiling of histone modifications and transcriptome in single cells from mouse brain

Abstract

Genome-wide profiling of histone modifications can reveal not only the location and activity state of regulatory elements, but also the regulatory mechanisms involved in cell-type-specific gene expression during development and disease pathology. Conventional assays to profile histone modifications in bulk tissues lack single-cell resolution. Here we describe an ultra-high-throughput method, Paired-Tag, for joint profiling of histone modifications and transcriptome in single cells to produce cell-type-resolved maps of chromatin state and transcriptome in complex tissues. We used this method to profile five histone modifications jointly with transcriptome in the adult mouse frontal cortex and hippocampus. Integrative analysis of the resulting maps identified distinct groups of genes subject to divergent epigenetic regulatory mechanisms. Our single-cell multiomics approach enables comprehensive analysis of chromatin state and gene regulation in complex tissues and characterization of gene regulatory programs in the constituent cell types.

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 Paired-Tag.
Fig. 2: Histone modification-based cell clustering recapitulates transcriptomic-based cell clustering with varying degrees of success.
Fig. 3: Integrative analysis of chromatin states at promoters and gene expression across mouse brain cell types.
Fig. 4: Characterization of chromatin state at distal candidate cis-regulatory elements across brain cell types.
Fig. 5: Correlative analysis of chromatin state and gene expression links distal candidate cis-regulatory elements to putative target genes.

Similar content being viewed by others

Data availability

The sequencing data obtained in this study have been deposited at the NCBI Gene Expression Omnibus (GEO) (http://www.ncbi.nlm.nih.gov/geo/) under accession number GSE152020. The processed data can be accessed from the web portal (http://catlas.org/pairedTag). All other data are available upon request. CEMBA datasets are available from NEMO with the accession number RRID SCR_016152 (http://data.nemoarchive.org/biccn/grant/cemba/ecker/chromatin/scell/raw/). ENCODE (https://www.encodeproject.org/) datasets were downloaded with the accession numbers: H3K4me1 (ENCSR000APW), H3K27ac (ENCSR000AOC), H3K27me3 (ENCSR000DTY), H3K9me3 (ENCSR000AQO) and DNase-seq (ENCSR959ZXU). The other external datasets were downloaded from NCBI Gene Expression Omnibus (GEO) (http://www.ncbi.nlm.nih.gov/geo/), with the accession numbers: SPLiT-seq (GSE110823), CoBATCH (GSE129335), itChIP (GSE109762) and HT-scChIP-seq (GSE117309). 10x scRNA-seq datasets were downloaded from the 10x Genomics website (https://www.10xgenomics.com/). Source data are provided with this paper.

Code availability

Custom scripts used in this study are available from https://github.com/cxzhu/Paired-Tag.

References

  1. Stadhouders, R., Filion, G. J. & Graf, T. Transcription factors and 3D genome conformation in cell-fate decisions. Nature 569, 345–354 (2019).

    Article  CAS  PubMed  Google Scholar 

  2. 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 

  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. Buenrostro, J. D., Giresi, P. G., Zaba, L. C., Chang, H. Y. & Greenleaf, W. J. Transposition of native chromatin for fast and sensitive epigenomic profiling of open chromatin, DNA-binding proteins and nucleosome position. Nat. Methods 10, 1213–1218 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Consortium, E. P. et al. Expanded encyclopaedias of DNA elements in the human and mouse genomes. Nature 583, 699–710 (2020).

    Article  CAS  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  7. 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 

  8. 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 

  9. 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 

  10. 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 

  11. 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 

  12. 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 e1311 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. 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 

  14. 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 

  15. 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).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  16. 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 

  17. Wang, Q. et al. CoBATCH for high-throughput single-cell epigenomic profiling. Mol. Cell 76, 206–216 e207 (2019).

    Article  CAS  PubMed  Google Scholar 

  18. Ai, S. et al. Profiling chromatin states using single-cell itChIP-seq. Nat. Cell Biol. 21, 1164–1172 (2019).

    Article  CAS  PubMed  Google Scholar 

  19. 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 

  20. 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 

  21. 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 

  22. 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 e725 (2017).

    Article  CAS  PubMed  Google Scholar 

  23. 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 

  24. 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 

  25. 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 

  26. Preissl, S. et al. Single-nucleus analysis of accessible chromatin in developing mouse forebrain reveals cell-type-specific transcriptional regulation. Nat. Neurosci. 21, 432–439 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. 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 

  28. Luo, C. et al. Single-cell methylomes identify neuronal subtypes and regulatory elements in mammalian cortex. Science 357, 600–604 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Cao, J. et al. Joint profiling of chromatin accessibility and gene expression in thousands of single cells. Science 361, 1380–1385 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. 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 

  31. 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 

  32. Luo, C. et al. Single nucleus multi-omics links human cortical cell regulatory genome diversity to disease risk variants. Preprint at bioRxiv https://doi.org/10.1101/2019.12.11.873398 (2019).

  33. Zhu, C., Preissl, S. & Ren, B. Single-cell multimodal omics: the power of many. Nat. Methods 17, 11–14 (2020).

    Article  CAS  PubMed  Google Scholar 

  34. Rosenberg, A. B. et al. Single-cell profiling of the developing mouse brain and spinal cord with split-pool barcoding. Science 360, 176–182 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. 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 

  36. Zeisel, A. et al. Molecular architecture of the mouse nervous system. Cell 174, 999–1014 e1022 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Magklara, A. et al. An epigenetic signature for monoallelic olfactory receptor expression. Cell 145, 555–570 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Baroti, T. et al. Transcription factors Sox5 and Sox6 exert direct and indirect influences on oligodendroglial migration in spinal cord and forebrain. Glia 64, 122–138 (2016).

    Article  PubMed  Google Scholar 

  39. Li, Y. E. et al. An atlas of gene regulatory elements in adult mouse cerebrum. Preprint at bioRxiv https://doi.org/10.1101/2020.05.10.087585 (2020).

  40. Briata, P. et al. Transcriptional repression by the human homeobox protein EVX1 in transfected mammalian cells. J. Biol. Chem. 270, 27695–27701 (1995).

    Article  CAS  PubMed  Google Scholar 

  41. Katsuoka, F. et al. Small Maf compound mutants display central nervous system neuronal degeneration, aberrant transcription, and Bach protein mislocalization coincident with myoclonus and abnormal startle response. Mol. Cell. Biol. 23, 1163–1174 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Park, N. I. et al. ASCL1 reorganizes chromatin to direct neuronal fate and suppress tumorigenicity of glioblastoma stem cells. Cell Stem Cell 21, 209–224 e207 (2017).

    Article  CAS  PubMed  Google Scholar 

  43. Yang, N. et al. Generation of pure GABAergic neurons by transcription factor programming. Nat. Methods 14, 621–628 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Lennard Richard, M. L., Nowling, T. K., Brandon, D., Watson, D. K. & Zhang, X. K. Fli-1 controls transcription from the MCP-1 gene promoter, which may provide a novel mechanism for chemokine and cytokine activation. Mol. Immunol. 63, 566–573 (2015).

    Article  CAS  PubMed  Google Scholar 

  45. Meng, G. & Mei, H. Transcriptional dysregulation study reveals a core network involving the progression of Alzheimer’s disease. Front. Aging Neurosci. 11, 101 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Yu, M. & Ren, B. The three-dimensional organization of mammalian genomes. Annu. Rev. Cell Dev. Biol. 33, 265–289 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Pliner, H. A. et al. Cicero predicts cis-regulatory DNA interactions from single-cell chromatin accessibility data. Mol. Cell 71, 858–871 e858 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Ngan, C. Y. et al. Chromatin interaction analyses elucidate the roles of PRC2-bound silencers in mouse development. Nat. Genet. 52, 264–272 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Pang, B. & Snyder, M. P. Systematic identification of silencers in human cells. Nat. Genet. 52, 254–263 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Wang, Y., Lin, L., Lai, H., Parada, L. F. & Lei, L. Transcription factor Sox11 is essential for both embryonic and adult neurogenesis. Dev. Dyn. 242, 638–653 (2013).

    Article  CAS  PubMed  Google Scholar 

  51. Zhu, C. et al. High-throughput single-cell joint analysis of histone modifications and gene expression by Paired-Tag. Preprint at Protocol Exchange https://doi.org/10.21203/rs.3.pex-1301/v1 (2021).

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Mulqueen, R. M. et al. Improved single-cell ATAC-seq reveals chromatin dynamics of in vitro corticogenesis. Preprint at bioRxiv https://doi.org/10.1101/637256 (2019).

  54. Langmead, B. & Salzberg, S. L. Fast gapped-read alignment with Bowtie 2. Nat. Methods 9, 357–359 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Langmead, B., Trapnell, C., Pop, M. & Salzberg, S. L. Ultrafast and memory-efficient alignment of short DNA sequences to the human genome. Genome Biol. 10, R25 (2009).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  56. Dobin, A. & Gingeras, T. R. Mapping RNA-seq reads with STAR. Curr. Protoc. Bioinformatics 51, 11 14 11-19 (2015).

    Google Scholar 

  57. Stuart, T. et al. Comprehensive integration of single-cell data. Cell 177, 1888–1902 e1821 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Korsunsky, I. et al. Fast, sensitive and accurate integration of single-cell data with Harmony. Nat. Methods 16, 1289–1296 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. McInnes, L., Healy, J., Saul, N. & Großberger, L. UMAP: Uniform manifold approximation and projection. J. Open Source Softw. 3, 861 (2018).

    Article  Google Scholar 

  60. Fang, R. et al. SnapATAC: a comprehensive analysis package for single cell ATAC-seq. Preprint at bioRxiv https://doi.org/10.1101/615179 (2020).

  61. 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).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. 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).

    CAS  PubMed  PubMed Central  Google Scholar 

  63. Khan, A. et al. JASPAR 2018: update of the open-access database of transcription factor binding profiles and its web framework. Nucleic Acids Res. 46, D260–D266 (2018).

    Article  CAS  PubMed  Google Scholar 

  64. Robinson, M. D., McCarthy, D. J. & Smyth, G. K. edgeR: a bioconductor package for differential expression analysis of digital gene expression data. Bioinformatics 26, 139–140 (2010).

    Article  CAS  PubMed  Google Scholar 

  65. Ernst, J. & Kellis, M. Chromatin-state discovery and genome annotation with ChromHMM. Nat. Protoc. 12, 2478–2492 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank B. Li for bioinformatic support, and S. Kuan for assistance with DNA sequencing. We thank QB3 MacroLab for the protein A-Tn5 enzyme. We thank S. Preissl, X. Hou, H. Huang, M. Yu and J. Song for discussion. This study was funded by grant nos. 1U19 MH114831-02, U01MH121282 and R01AG066018 and the Ludwig Institute for Cancer Research (to B.R.); grant no. 1K99HG011483-01 (to C.Z.); grant no. 1K99CA252020-01 (to Y.Z.); and grant no. R01MH112763 (to M.M.B.). This publication includes data generated at the UC San Diego IGM Genomics Center utilizing an Illumina NovaSeq 6000 that was purchased with funding from a National Institutes of Health SIG grant (no. S10 OD026929).

Author information

Authors and Affiliations

Authors

Contributions

B.R. and C.Z. conceived and designed the study and wrote the manuscript. C.Z. performed the Paired-Tag experiments. C.Z., Y.Z. and Y.E.L. performed the data analysis. Y.E.L. set up the web portal. J.L. and M.M.B. harvested and dissected the mouse brain tissues. All authors discussed results and edited the manuscript. B.R. supervised the research.

Corresponding author

Correspondence to Bing Ren.

Ethics declarations

Competing interests

B.R. is a co-founder and consultant for Arima Genomics, Inc., and a co-founder of Epigenome Technologies, Inc. B.R. and C.Z. are listed as inventors of a provisional patent titled ‘Parallel analysis of individual cells for RNA expression and DNA from targeted tagmentation by sequencing’.

Additional information

Peer review information Nature Methods thanks Andrew Adey, Steven Henikoff and the other, anonymous, reviewer(s) for their contribution to the peer review of this work. Lei Tang was the primary editor on this article and managed its editorial process and peer review in collaboration with the rest of the editorial team.

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

Extended data

Extended Data Fig. 1 Overview of Paired-Tag.

a, Schematics for 2nd adaptor tagging of DNA and RNA libraries. For DNA libraries, amplified products were digested with a type IIS restriction enzyme – FokI, and the cohesive end was then used to ligate the P5 adaptor. For RNA libraries, N5 adaptor was added by tagmentation. b, Table showing the numbers of overlapped peaks across different histone marks between Paired-Tag and ENCODE ChIP-seq or DNase-seq datasets. The total numbers of peaks identified for each dataset are also indicated. c, d, Heatmaps showing the Pearson’s Correlation Coefficients of genome-wide reads distribution (in 10-kb bins) (c) between Paired-Tag datasets and ENCODE ChIP-seq or DNase-seq datasets, and (d) between replicates of Paired-Tag datasets and ENCODE ChIP-seq datasets from HeLa cells. e, Scatter plot showing the Pearson’s correlation coefficients of Paired-Tag RNA dataset and in-house generated nuclei RNA-seq from HeLa cells.

Extended Data Fig. 2 Performances of Paired-Tag in single-nucleus analysis from mouse brain.

a, Schematics showing the sample multiplexing strategy in this study. Different samples or replicates were labeled by the 1st round of Paired-Tag cellular barcode (Sample Barcode) located in reverse transcription primers and transposome oligos. b, Heatmap showing the pair-wise Pearson’s correlation coefficients of genome-wide reads distribution for different histone marks from single-cell Paired-Tag datasets (indicated with ‘sc’, aggregated from all cells shown in Fig. 2a) and bulk datasets. c, Boxplots showing the mapping rates (upper panels) and the fraction of reads uniquely mapped to the reference genome (bottom panels) of DNA profiles of different histone marks and RNA profiles in frontal cortex and hippocampus. The boxes were drawn from lower quartile (Q1) to upper quartile (Q3) with the middle line denote the median, whiskers with maximum 1.5 IQR, outliers were indicated with dots. For Frontal Cortex, n = 7,781 (H3K4me1), 3,509 (H3K4me3), 7,584 (H3K27ac), 3,891 (H3K27me3), 6,560 (H3K9me3), 6,551 (ChromAcc) from dissections of 2 different mice s and n = 35,876 (RNA) from dissections of 4 different mice; for Hippocampus, n = 5,181 (H3K4me1), 3,956 (H3K4me3), 4,165 (H3K27ac), 2,643 (H3K27me3), 5,484 (H3K9me3), 7,544 (ChromAcc) from dissections of 2 different mice and n = 28,973 (RNA) from dissections of 4 different mice. d, Scatter plots showing the proportion of human and mouse RNA reads in each cell (left panel) and the fraction of human reads in DNA and RNA libraries for each cell (right panel) in the species-mixing experiment. Barcodes with less than 80% reads from the same species were identified as mixed cells, the 230 mixed cells of RNA profiles (left panel) were excluded from plotting of the right panel. e, Numbers of unique loci per nucleus for deeply sequenced H3K4me1, H3K4me3, H3K27ac, H3K27me3 and H3K9me3 DNA profiles down-sampled to different levels. f, Numbers of unique loci per nucleus for deeply sequenced Paired-seq DNA profiles down-sampled to different levels. g, Numbers of UMI per nucleus for the deeply sequenced RNA sub-library down-sampled to different levels. For comparison, the numbers of unique loci per cell from the stand-alone high-throughput scChIP-seq assays and the numbers of UMI per cell from scRNA-seq assays were also shown, indicated by dots with labels. h, Violin plots showing the numbers of unique loci mapped per nucleus for all sequenced DNA libraries (average 35k sequenced reads/nuclei with ~40–60% PCR duplication rates). Median numbers, H3K4me1: 5,770 and 5,443, H3K4me3: 1,392 and 1,081, H3K27ac: 1,842 and 1,803, H3K27me3: 904 and 925, H3K9me3: 6,563 and 7,182, chromatin accessibility: 3,170 and 4,381, for frontal cortex and hippocampus, respectively. i, Violin plots showing the fraction of reads inside peaks for different histone marks and brain regions. For Frontal Cortex, n = 7,781 (H3K4me1), 3,509 (H3K4me3), 7,584 (H3K27ac), 3,891 (H3K27me3), 6,560 (H3K9me3), 6,551 (ChromAcc) from dissections of 2 different mice; for Hippocampus, n = 5,181 (H3K4me1), 3,956 (H3K4me3), 4,165 (H3K27ac), 2,643 (H3K27me3), 5,484 (H3K9me3), 7,544 (ChromAcc) from dissections of 2 different mice. j, Violin plots showing the numbers of UMI and genes detected per nucleus for all sequenced RNA libraries (average 30k sequenced reads/nuclei with ~40–60% PCR duplication rates). Median numbers, 4,215 and 3,568 RNA UMI per nucleus for frontal and hippocampus, respectively. k,l, Violin plots showing the (k) fraction of reads mapped to annotated gene regions (GENCODE GRCm38.p6) and (l) fraction of intronic reads for Paired-Tag RNA datasets and 10X scRNA-seq datasets (10k Brain Cells from an E18 Mouse, V3). n = 35,876 (Frontal Cortex) and 28,973 (Hippocampus) from dissections of 4 different mice. For (h-l), the violin plots were drawn from lower quartile (Q1) to upper quartile (Q3) with the middle line denote the median, whiskers with maximum 1.5 IQR, outliers were indicated with dots.

Source data

Extended Data Fig. 3 Annotation of cell types by Paired-Tag transcriptomic profiles.

a, UMAP embedding of Paired-Tag transcriptomic profiles and stacked bar plots showing the fraction of cells from different regions or replicates (dissections from different mice) in each cell type. b, UMAP embedding of transcriptomic profiles from individual Paired-Tag and Paired-seq datasets. The color of cell types was the same as in Fig.1d. c, Dot plots showing the expression of marker genes for each mouse brain cell type measured from Paired-Tag RNA profiles. The size of the dots represents the fraction of cells positively detect the transcripts and the color of the dots represents the average. d, UMAPs showing the co-embedding of single-nucleus gene expression from Paired-Tag RNA profiles and the previously published scRNA-seq datasets of the same tissues. e, Heatmaps showing the confusion matrices of the overlap between cell type annotations based on Paired-Tag RNA profiles and from the previously published scRNA-seq datasets. The circles left side indicating RNA clusters and were colored according to Fig.1d. f, Boxplots showing the Pearson’s correlation coefficients for all genes, variable genes and invariable genes for matched and non-matched cell types between Paired-Tag RNA profiles and the previously published scRNA-seq. The boxes were drawn from lower quartile (Q1) to upper quartile (Q3) with the middle line denote the median, whiskers with maximum 1.5 IQR, outliers were indicated with dots. n = 22 cell types. g, Scatter plot showing the expression levels of variable genes in Astrocytes measured by Paired-Tag RNA profiles and the published scRNA-seq datasets.

Source data

Extended Data Fig. 4 Histone marks-based single-cell clustering.

a-e, UMAP embeddings based on (a) H3K4me1, (b) H3K4me3, (c) H3K27ac, (d) H3K27me3 and (e) H3K9me3 DNA profiles and stacked bar plots showing the fraction of cells from each region or replicate. f, UMAP embeddings based on Paired-Tag H3K27ac DNA profiles down-sampled to different sequencing depth (11,749 nuclei, 100–1,500 loci/nuclei). g, UMAP embeddings based on Paired-Tag H3K27ac DNA profiles of different numbers of sub-sampled nuclei (median 1,826 loci/nuclei, 200–10,000 nuclei). For visualization, cells were colored according to clustering results from Fig. 1d.

Source data

Extended Data Fig. 5 Gene expression and promoter epigenetic states.

a, Violin plots showing reads densities of the five histone marks in Group II-a and III-b promoters. Colors represent cell types the same as in (e). Group II-a promoters were repressed by H3K27me3 in all cell types; genes in III-b were activated by H3K27ac in neuron cells, with comparable H3K27me3 levels in all cell groups. b, Boxplots showing the expression levels of genes grouped by their promoter DNA reads densities for different histone marks. The boxes were drawn from lower quartile (Q1) to upper quartile (Q3) with the middle line denote the median, whiskers with maximum 1.5 IQR, outliers were indicated with dots. n = 2,900 genes for the first 5 groups and n = 2,898 for the 6th group of each histone modification. c, Heatmap showing the Spearman’s correlation coefficients of gene expression and promoter histone modification levels within each cell type. d, Genome browser view of aggregated Paired-Tag profiles showing the three Olfr gene clusters in chr7 was silenced by H3K9me3. e, 3D-scatter plot showing the PCA embedding of aggregated RNA profiles. PC1 differs neuron cells from glial cells and PC2 mainly separates different non-neuron cell types. f, Scatter plot showing the loadings of the first 2 PCs for each gene. Genes from group II-b and III-d were colored in brown and blue, respectively. g, UMAP embedding of 4,659 OPC and Oligodendrocytes nuclei used for pseudotime analysis. h, Expression of marker genes alone the pseudotime.

Source data

Extended Data Fig. 6 Histone modification states in mouse neuron cell types.

a, Stacked bar plots showing the fraction of genomic regions for CREs of each group in Fig. 4a and b. b, Line plots showing the densities of CREs from different groups around CpG islands. c, Scatter plot showing the Spearman’s correlation coefficients of TF motif enrichment and TF gene expression across cell types. TFs with significant positive correlations (FDR < 0.05) between expression and motif enrichment for both H3K4me1 and H3K27ac were colored in red. d, Heatmap showing the emission probability of each histone mark across the 8 chromatin states identified by chromHMM. e, Heatmap showing the fold enrichment of the 8 chromatin states around transcription start sites of FC L2/3 cell cluster.

Source data

Extended Data Fig. 7 Identification of putative CRE-gene pairs.

a, Bar charts showing the fraction of predicted H3K27ac- and H3K27me3- associated cCRE-gene pairs supported by the CEMBA datasets. P-value, two-tailed Fisher’s exact test. be, Bar charts showing the numbers of cCREs per targeted genes for (b) H3K27ac- and (c) H3K27me3- associated cCRE-gene pairs, and the numbers of predicted targeted genes per cCRE for (d) H3K27ac- and (e) H3K27me3- associated cCRE-gene pairs. f, Representative genome browser view of Gad2 gene locus, both H3K27ac- and H3K27me3-associated cCREs were shown. TSS-proximal region is indicated with green box and cCREs are marked with blue (H3K27ac-specific), brown (H3K27me3-specific) or purple (shared) boxes. g, Top enriched de novo TF motifs and GO terms of cCREs in H3K27ac-specific, shared and H3K27me3-specific pairs. h, Stacked bar plots showing the fraction of genomic regions for cCREs with potential active and repressive functions. P-value, two-tailed Fisher’s exact test. i,j, Bar charts showing the distribution of the distance between cCRE and TSS of predicted target genes from (i) H3K27ac- and (j) H3K27me3- associated cCRE-gene pairs. k, Spearman’s correlations coefficients between reads densities of cCREs and promoters of putative target genes across cell types for H3K4me1 and H3K9me3. P-value, two-tailed Wilcoxon signed-rank test. The boxes were drawn from lower quartile (Q1) to upper quartile (Q3) with the middle line denote the median, whiskers with maximum 1.5 IQR. n = 22 cell types. l, Heatmap showing the histone modification levels at cCREs with potential repressive roles in expression of putative target gene. cCRE were grouped using K-means clustering based on histone modification levels. m, Heatmap showing the expression levels of corresponding putative target genes of cCREs in (l). n, Top enriched Gene Ontology terms for genes in (m) and the top enriched de novo motifs for cCREs from each group in (l).

Source data

Supplementary information

Supplementary Information

Supplementary Notes.

Reporting Summary

Supplementary Table 1

Paired-Tag primer sequences. This table shows the primer and barcode sequences used in Paired-Tag experiments.

Supplementary Table 2

Nuclei metadata. This table shows the sequencing quality, mapping status, clustering and annotation information of single nuclei in this study.

Supplementary Table 3

Marker genes by clusters. This table shows the differentially expressed genes between the clusters obtained from Paired-Tag RNA profiles. P value: two-sided Wilcoxon rank sum test and adjusted by Bonferroni correction using all features in the dataset.

Supplementary Table 4

Promoters by groups. This table lists the genes of different groups classified by epigenetic states of the corresponding promoters described in Fig. 3.

Supplementary Table 5

Gene Ontology analysis of genes from different groups. This table summarized the Gene Ontology analysis results for genes from each group in Fig. 3. P value: one-sided binomial test.

Supplementary Table 6

cis-Regulatory elements by groups. This table lists the CREs of different groups classified by their epigenetic states described in Fig. 4.

Supplementary Table 7

Known Motifs Enrichment analysis of CREs from different groups. This table summarizes the enrichment of known motifs for CREs from each group in Fig. 4. P value: one-sided binomial test.

Supplementary Table 8

TF motif enrichment—gene expression correlation. This table lists Spearman’s correlation coefficients between motif enrichment (chromVAR deviations) and gene expression (RPKM) across clusters for 342 transcription factors.

Supplementary Table 9

Predicted cCRE–gene pairs. This table lists the predicted H3K27ac- and H3K27me3-associated cCRE–gene pairs in Fig. 5.

Supplementary Table 10

Predicted target genes by groups. This table lists the predicted target genes from both H3K27ac- and H3K27me3-associated pairs for each group in Fig. 5.

Source data

Source Data Fig. 1

Statistical source data.

Source Data Fig. 2

Statistical source data.

Source Data Fig. 3

Statistical source data.

Source Data Fig. 4

Statistical source data.

Source Data Fig. 5

Statistical source data.

Source Data Extended Data Fig. 2

Statistical source data.

Source Data Extended Data Fig. 3

Statistical source data.

Source Data Extended Data Fig. 4

Statistical source data.

Source Data Extended Data Fig. 5

Statistical source data.

Source Data Extended Data Fig. 6

Statistical source data.

Source Data Extended Data Fig. 7

Statistical source data.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Zhu, C., Zhang, Y., Li, Y.E. et al. Joint profiling of histone modifications and transcriptome in single cells from mouse brain. Nat Methods 18, 283–292 (2021). https://doi.org/10.1038/s41592-021-01060-3

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41592-021-01060-3

This article is cited by

Search

Quick links

Nature Briefing: Translational Research

Sign up for the Nature Briefing: Translational Research newsletter — top stories in biotechnology, drug discovery and pharma.

Get what matters in translational research, free to your inbox weekly. Sign up for Nature Briefing: Translational Research