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:

Single-cell joint detection of chromatin occupancy and transcriptome enables higher-dimensional epigenomic reconstructions

Abstract

Deciphering mechanisms in cell-fate decisions requires single-cell holistic reconstructions of multidimensional epigenomic states in transcriptional regulation. Here we develop CoTECH, a combinatorial barcoding method allowing high-throughput single-cell joint detection of chromatin occupancy and transcriptome. We used CoTECH to examine bivalent histone marks (H3K4me3 and H3K27me3) with transcription from naive to primed mouse embryonic stem cells. We also derived concurrent bivalent marks in pseudosingle cells using transcriptome as an anchor for resolving pseudotemporal bivalency trajectories and disentangling a context-specific interplay between H3K4me3/H3K27me3 and transcription level. Next, we revealed the regulatory basis of endothelial-to-hematopoietic transition in two waves of hematopoietic cells and distinctive enhancer-gene-linking schemes guiding hemogenic endothelial cell emergence, indicating a unique epigenetic control of transcriptional regulation for hematopoietic stem cell priming. CoTECH provides an efficient framework for single-cell coassay of chromatin occupancy and transcription, thus enabling higher-dimensional epigenomic reconstructions.

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: Schematic design of CoTECH.
Fig. 2: Assessing the ability of CoTECH to capture histone mark and transcription in single cells.
Fig. 3: Single-cell joint detection of H3K4me3/H3K27me3 and transcription in mouse embryonic stem cells.
Fig. 4: CoTECH reveals both coupling and uncoupling of dynamic histone modifications and transcription from naive to primed mESCs.
Fig. 5: Single-cell reconstructions of dynamic trajectories of chromatin bivalency and transcription from naive to primed mESCs.
Fig. 6: Linking enhancers to regulated genes reveals mechanisms underlying EHT in YS and AGM.

Similar content being viewed by others

Data availability

Sequencing data have been deposited at the NCBI GEO with accession number GSE158435. Other public datasets used in this study were downloaded from NCBI GEO with accession numbers as follows: scCC (GSE148448), sci-CAR (GSE117089), Paired-seq (GSE130399), SNARE-seq (GSE126074), bulk H3K4me3 ChIP-seq in NIH 3T3 (GSM1544520), bulk RNA-seq in NIH 3T3 (GSM970853), bulk H3K4me3 ChIP-seq in mESC (GSM1000124), bulk H3K27me3 ChIP-seq in mESC (GSM1000089), bulk RNA-seq in mESC (GSM723776), bulk H3K27ac ChIP-seq in NIH 3T3 (GSM801538), bulk RNA-seq in 293T (GSM2258993) and scATAC-seq in H1ESC (GSE65360). Source data are provided with this paper.

Code availability

Custom scripts used in this study are available from https://github.com/Helab-bioinformatics/CoTECH.

References

  1. 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 

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

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  4. 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 

  5. 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  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. Lai, B. et al. Principles of nucleosome organization revealed by single-cell micrococcal nuclease sequencing. Nature 562, 281–285 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Guo, F. et al. The transcriptome and DNA methylome landscapes of human primordial germ cells. Cell 161, 1437–1452 (2015).

    Article  CAS  PubMed  Google Scholar 

  9. 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 

  10. Efremova, M. & Teichmann, S. A. Computational methods for single-cell omics across modalities. Nat. Methods 17, 14–17 (2020).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. 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 

  13. 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 

  14. 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 

  15. 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 

  16. Park, P. J. ChIP-seq: advantages and challenges of a maturing technology. Nat. Rev. Genet. 10, 669–680 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Moudgil, A. et al. Self-reporting transposons enable simultaneous readout of gene expression and transcription factor binding in single cells. Cell 182, 992–1008 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. 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 

  20. Picelli, S. et al. Full-length RNA-seq from single cells using Smart-seq2. Nat. Protoc. 9, 171–181 (2014).

    Article  CAS  PubMed  Google Scholar 

  21. Srivatsan, S. R. et al. Massively multiplex chemical transcriptomics at single-cell resolution. Science 367, 45–51 (2020).

    Article  CAS  PubMed  Google Scholar 

  22. Yang, S. et al. Decontamination of ambient RNA in single-cell RNA-seq with DecontX. Genome Biol. 21, 57 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  23. Bravo Gonzalez-Blas, C. et al. cisTopic: cis-regulatory topic modeling on single-cell ATAC-seq data. Nat. Methods 16, 397–400 (2019).

    Article  CAS  PubMed  Google Scholar 

  24. Benayoun, B. A. et al. H3K4me3 breadth is linked to cell identity and transcriptional consistency. Cell 163, 1281–1286 (2015).

    Article  CAS  PubMed  Google Scholar 

  25. Callis, T. E. et al. MicroRNA-208a is a regulator of cardiac hypertrophy and conduction in mice. J. Clin. Invest. 119, 2772–2786 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Howe, F. S., Fischl, H., Murray, S. C. & Mellor, J. Is H3K4me3 instructive for transcription activation? Bioessays 39, 1–12 (2017).

    Article  CAS  PubMed  Google Scholar 

  27. Ruthenburg, A. J., Allis, C. D. & Wysocka, J. Methylation of lysine 4 on histone H3: intricacy of writing and reading a single epigenetic mark. Mol. Cell 25, 15–30 (2007).

    Article  CAS  PubMed  Google Scholar 

  28. Wang, Z. et al. Combinatorial patterns of histone acetylations and methylations in the human genome. Nat. Genet. 40, 897–903 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Barski, A. et al. High-resolution profiling of histone methylations in the human genome. Cell 129, 823–837 (2007).

    Article  CAS  PubMed  Google Scholar 

  30. Santos-Rosa, H. et al. Active genes are tri-methylated at K4 of histone H3. Nature 419, 407–411 (2002).

    Article  CAS  PubMed  Google Scholar 

  31. Fortier, S., MacRae, T., Bilodeau, M., Sargeant, T. & Sauvageau, G. Haploinsufficiency screen highlights two distinct groups of ribosomal protein genes essential for embryonic stem cell fate. Proc. Natl Acad. Sci. USA 112, 2127–2132 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Bernstein, B. E. et al. A bivalent chromatin structure marks key developmental genes in embryonic stem cells. Cell 125, 315–326 (2006).

    Article  CAS  PubMed  Google Scholar 

  33. Harikumar, A. & Meshorer, E. Chromatin remodeling and bivalent histone modifications in embryonic stem cells. EMBO Rep. 16, 1609–1619 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Dainese, R. et al. A parallelized, automated platform enabling individual or sequential ChIP of histone marks and transcription factors. Proc. Natl Acad. Sci. USA 117, 13828–13838 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Dzierzak, E. & Bigas, A. Blood development: hematopoietic stem cell dependence and independence. Cell Stem Cell 22, 639–651 (2018).

    Article  CAS  PubMed  Google Scholar 

  36. Hou, S. et al. Embryonic endothelial evolution towards first hematopoietic stem cells revealed by single-cell transcriptomic and functional analyses. Cell Res. 30, 376–392 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  37. Palis, J. Hematopoietic stem cell-independent hematopoiesis: emergence of erythroid, megakaryocyte, and myeloid potential in the mammalian embryo. FEBS Lett. 590, 3965–3974 (2016).

    Article  CAS  PubMed  Google Scholar 

  38. Lilly, A. J. et al. Interplay between SOX7 and RUNX1 regulates hemogenic endothelial fate in the yolk sac. Development 143, 4341–4351 (2016).

    CAS  PubMed  Google Scholar 

  39. Creyghton, M. P. et al. Histone H3K27ac separates active from poised enhancers and predicts developmental state. Proc. Natl Acad. Sci. USA 107, 21931–21936 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Argelaguet, R. et al. Multi-omics factor analysis-a framework for unsupervised integration of multi-omics data sets. Mol. Syst. Biol. 14, e8124 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  41. 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 

  42. Teichweyde, N. et al. HOXB4 promotes hemogenic endothelium formation without perturbing endothelial cell development. Stem Cell Rep. 10, 875–889 (2018).

    Article  CAS  Google Scholar 

  43. McGrath, K. E. & Palis, J. Expression of homeobox genes, including an insulin promoting factor, in the murine yolk sac at the time of hematopoietic initiation. Mol. Reprod. Dev. 48, 145–153 (1997).

    Article  CAS  PubMed  Google Scholar 

  44. Gao, L. et al. RUNX1 and the endothelial origin of blood. Exp. Hematol. 68, 2–9 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Yzaguirre, A. D., Howell, E. D., Li, Y., Liu, Z. & Speck, N. A. Runx1 is sufficient for blood cell formation from non-hemogenic endothelial cells in vivo only during early embryogenesis. Development 145, dev158162 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  46. Kelsey, G., Stegle, O. & Reik, W. Single-cell epigenomics: recording the past and predicting the future. Science 358, 69–75 (2017).

    Article  CAS  PubMed  Google Scholar 

  47. Shema, E., Bernstein, B. E. & Buenrostro, J. D. Single-cell and single-molecule epigenomics to uncover genome regulation at unprecedented resolution. Nat. Genet. 51, 19–25 (2019).

    Article  CAS  PubMed  Google Scholar 

  48. 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 

  49. Schier, A. F. Single-cell biology: beyond the sum of its parts. Nat. Methods 17, 17–20 (2020).

    Article  CAS  PubMed  Google Scholar 

  50. Eisenstein, M. The secret life of cells. Nat. Methods 17, 7–10 (2020).

    Article  CAS  PubMed  Google Scholar 

  51. Zeng, Y. et al. Tracing the first hematopoietic stem cell generation in human embryo by single-cell RNA sequencing. Cell Res. 29, 881–894 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Picelli, S. et al. Tn5 transposase and tagmentation procedures for massively scaled sequencing projects. Genome Res. 24, 2033–2040 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Luo, Y. et al. CoTECH for single-cell joint detection of transcriptome and chromatin occupancy. Protoc. Exch. https://doi.org/10.21203/rs.3.pex-1300/v1 (2021).

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Anders, S., Pyl, P. T. & Huber, W. HTSeq–a Python framework to work with high-throughput sequencing data. Bioinformatics 31, 166–169 (2015).

    Article  CAS  PubMed  Google Scholar 

  56. Langdon, W. B. Performance of genetic programming optimised Bowtie2 on genome comparison and analytic testing (GCAT) benchmarks. BioData Min. 8, 1 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  PubMed  PubMed Central  Google Scholar 

  58. Ramírez, F., Dündar, F., Diehl, S., Grüning, B. A. & Manke, T. deepTools: a flexible platform for exploring deep-sequencing data. Nucleic Acids Res. 42, W187–191 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  59. Thorvaldsdóttir, H., Robinson, J. T. & Mesirov, J. P. Integrative Genomics Viewer (IGV): high-performance genomics data visualization and exploration. Brief. Bioinform. 14, 178–192 (2013).

    Article  PubMed  Google Scholar 

  60. Fan, X. et al. Spatial transcriptomic survey of human embryonic cerebral cortex by single-cell RNA-seq analysis. Cell Res. 28, 730–745 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Satija, R., Farrell, J. A., Gennert, D., Schier, A. F. & Regev, A. Spatial reconstruction of single-cell gene expression data. Nat. Biotechnol. 33, 495–502 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. 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 

  64. Huang, D. W., Sherman, B. T. & Lempicki, R. A. Bioinformatics enrichment tools: paths toward the comprehensive functional analysis of large gene lists. Nucleic Acids Res. 37, 1–13 (2009).

    Article  Google Scholar 

  65. Huang, D. W., Sherman, B. T. & Lempicki, R. A. Systematic and integrative analysis of large gene lists using DAVID bioinformatics resources. Nat. Protoc. 4, 44–57 (2009).

    Article  CAS  Google Scholar 

  66. Granja, J. M. et al. Single-cell multiomic analysis identifies regulatory programs in mixed-phenotype acute leukemia. Nat. Biotechnol. 37, 1458–1465 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank all members of the He laboratory for critical comments on this manuscript. Part of the analyses was performed on the High-Performance Computing Platform of the Center for Life Sciences, Peking University. We thank the flow cytometry core at the National Center for Protein Sciences at Peking University, particularly L. Du and H. Yang, for technical help. A.H. was supported by the National Key Research and Development Program of China (2019YFA0801802 and 2017YFA0103402), the National Natural Science Foundation of China (32025015 and 31771607), the Peking-Tsinghua Center for Life Sciences and the 1000 Youth Talents Program of China.

Author information

Authors and Affiliations

Authors

Contributions

A.H. conceived and designed the study. Y.L. and Q.W. designed and performed all experiments, assisted by X.Y. H.X. performed the computational analyses, supervised by A.H. H.X., Y.L. and A.H. wrote the paper with input from all other authors. All authors participated in data discussion and interpretation.

Corresponding author

Correspondence to Aibin He.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Peer review information Nature Methods thanks Andrew Adey 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 the DNA- and RNA-devoted library preparation of CoTECH.

a, Molecular structure of the CoTECH DNA-partition library. Following tagmentation and pre-amplification, the DNA-partition library was prepared by the 2-round PCR enrichment, resulting in the molecular constructs ready for sequencing using the standard Illumina Truseq recipe for paired-end 150-bp reads on Hiseq X-ten or Novaseq 6000 platform. b, Molecular structure of the CoTECH RNA-partition library. The mRNA was captured and reverse transcribed as described in a modified Smart-seq2 protocol. The reverse-transcribed cDNA was amplified by IS + 3’P2 primers and tagmented by Tn5-MEA. The resulting library was enriched by Nextera i5 and Truseq i7 primers and sequenced for paired-end 150-bp reads by loading both Nextera and Truseq read primers on the Novaseq 6000 platform.

Extended Data Fig. 2 Data quality of DNA and RNA profiles of CoTECH data.

a, Track view displaying both H3K27ac ChIP and RNA signal at representative locus in NIH 3T3 (left) and 293T (right) cells. Bulk H3K27ac ChIP-seq data in NIH 3T3 and 293T cells were downloaded from GSM801538 and ENCODE, respectively. Bulk RNA data in NIH 3T3 and 293T cells were downloaded from GSM970853 and GSM2258993. b-c, Species-mixing experiment of H3K27ac-RNA CoTECH in NIH 3T3 and 293T cells. Scatter plots showing the unique mapped reads (b) and UMI (c) rate for cells in which DNA and RNA profiles were obtained. d, Cross-cell contamination in the species-mixing dataset (n = 180 and 158 for mouse and human cells. The contamination was accurately quantified and estimated by DecontX. e, Venn diagram showing peak overlapping between CoTECH-H3K4me3 and bulk ChIP-seq data. Bulk data was from GSM1544520. f-g, Heatmap showing CoTECH-H3K4me3 and bulk H3K4me3 ChIP signals at 10-kb peak regions. The rows were sorted by the descending signals in 25,311 bulk peaks (f) and 23,149 CoTECH-H3K4me3 peaks (g). h-i, Violin plot showing the FRiP (h) and sensitivity (i) for single cells of CoTECH data (n = 844), scATAC-seq data (H1ESC, GSE65360) (n = 96) and simulated random genomic regions. The 25,311 bulk ChIP peaks were used to calculate the FRiP and sensitivity. j, Comparison of two CoTECH experiments with respect to mapping rate and exonic rate in NIH 3T3 cells (n = 844). k, Characterization of CoTECH, scDam&T-seq and scCC. l, Violin plot showing the distribution of non-duplicated reads (DNA), gene number (RNA), and UMI (RNA) in different datasets across SNARE-seq (n = 1,047, 10,309, and 5,081 for CellLineMixture, AdBrainCortex and P0_BrainCortex dataset, respectively), Paired-seq (n = 3,359, 25,845 and 15,191 for Cell_Mix, Fetal_Forebrain and Adult_Cerebrail_Cortex dataset, respectively), sci-CAR (n = 6,085 and 13,395 for A549 and mouse_kidney dataset, respectively), and CoTECH (n = 844 and 6,993 for NIH 3T3 and mESC dataset, respectively). Processed data were downloaded from GSE126074, GSE130399, and GSE117089, respectively. The boxes in violin plots of Extended Data Fig. 2d, h-j and l indicate upper and lower quartiles (25th and 75th percentiles).

Source data

Extended Data Fig. 3 Optimization of read number with/without pre-amplification of DNA fragments and detection of gene numbers.

a, Distribution of non-duplicated reads (DNA), gene number (RNA), and UMI (RNA) of 96 single cells in K562 between two independent CoTECH libraries. Red line indicates the median. For DNA pre-amplification, primer pairs Connector A/B (Supplementary Table 1) were added to amplify CoTECH DNA fragments for another 5 cycles following 4+7 cycles for full-length cDNA pre-amplification by primers IS and P2. For RNA partition, reads mapped to gene exons were used for calculation here. b, Violin plot showing the distribution of gene number calculated using reads mapping to exons (spliced mRNAs) and reads mapped to both exons and introns (precursor and spliced mRNAs) detected in RNA partition of CoTECH data in K562 cells without (left) or with pre-amplification (right) (n = 96 for each condition). The boxes in violin plots indicate upper and lower quartiles (25th and 75th percentiles).

Source data

Extended Data Fig. 4 CoTECH Data quality and relationship between H3K4me3 and RNA in mESCs.

a, Hierarchical clustering of DNA profiles of different groups by using the genome-wide signals in non-overlapping 5-kb windows. b, Pearson correlation of gene expression (n=24,413 genes) between four CoTECH assays. Aggregated transcript reads were log10 normalized. Two-sided correlation coefficients, p value < 2.2e-16. c, Comparison of CoTECH assays with respect to mapping rate and exonic rate (n = 3,907 and 3,086 cells for CoTECH-RNA (H3K4me3) and CoTECH-RNA (H3K27me3), respectively). The boxes in violin plots indicate upper and lower quartiles (25th and 75th percentiles). d, Dimensionality reduction for visualizing 6,993 single cells (both DNA and RNA detected) from different replicates in mESCs using UMAP. e, UMAP showing cellular heterogeneity of individual cells using CoTECH-H3K4me3 profiles colored by different replicates of mESCs and NIH 3T3. f-g, Cell-normalized topic scores of specific topics for NIH 3T3 (f) and mESCs (g) identified by cisTopic using CoTECH-H3K4me3 data. The colors from grey to red indicate the topic scores from low to high. h, GO terms enriched in the NIH 3T3- and mESC-specific topic corresponding to (f) and (g). Top 5 terms of biological process were shown. P value was calculated by two-sided binomial test. Gene Ontology (GO) term enrichment analysis of each module was performed by GREAT. i, Heatmap showing the expression of feature genes in mESCs. Color bars indicate identified clusters (top row) as in Fig. 3b. The columns represent single cells ordered by pseudotime. j, Analytic strategy for examining H3K4me3-RNA concordance in a varying number of clusters of mESCs along pseudotime. k, Fraction of cells with concordance of RNA-H3K4me3 in a varying number of clusters of mESCs along pseudotime. l-m, Pseudotime of 6,993 single cells by monocle 2. Cells are colored by clusters (l) as in Fig. 3b and pseudotime (m). n, Heatmaps showing the normalized H3K4me3 signals (left) and gene expression (right) of genes upon changes in H3K4me3 signal across 3,907 mESCs. The columns represent single cells ordered by pseudotime. o, Scatter plot showing the pseudotime defined by H3K4me3 and RNA profiles for the same cells of mESCs. The scatters were fitted by locally weighted regression to a red curve.

Source data

Extended Data Fig. 5 Co-occupancy analysis of H3K4me3 and H3K27me3 in mESCs.

a, Track view displaying H3K4me3 and H3K27me3 signals at three different representative loci in mESC. b, Bivalency score at the promoter regions (TSS ± 3 kb) of genes. H3K4me3 and H3K27me3 signals were normalized using RPKM. 2,706 reported bivalent genes were colored with red. Bulk H3K4me3, H3K27me3 ChIP-seq and bulk RNA-seq data were downloaded from GSM1000124, GSM1000089 and GSM723776, respectively. c-d, Dimensionality reduction for visualizing both 6,993 single cells and 350 pseudosingle cells of mESCs colored by clusters (c) and pseudotime (d) ordered by RNA profiles. e, The pseudotemporal dynamics of H3K4me3, H3K27me3, RNA, and bivalency scores of representative pluripotency genes. f, UMAP plot showing bivalency scores (up) and gene expression (down) in 350 pseudosingle cells of mESCs. Notably, bivalent genes are typically silenced or expressed at a very low level in mESCs.

Source data

Extended Data Fig. 6 Multi-omics factor analysis reveals coordinated epigenetic and transcriptomic variation during endothelial-to-hematopoietic transition between YS and AGM.

a, The variance explained by DNA and RNA profiles of CoTECH experiments for all AGM cells. b, Scatter plot of MOFA factor 1 (x axis) and MOFA factor 2 (y axis) for all AGM cells. Cells are colored as in Fig. 6. c, UMAP visualization of expression of marker genes for AGM cells. d, The variance explained by DNA and RNA profiles of CoTECH experiments for all YS cells. e, Scatter plot of MOFA factor 1 (x axis) and MOFA factor 2 (y axis) for all YS cells. Cells are colored as in Fig. 6. f, UMAP visualization of expression of marker genes for YS cells. g, Heatmaps showing RNA (left) and H3K27ac (right) signals at gene-correlated peaks in AGM cells along the pseudotime during EHT. The rows represent 2,219 non-differential genes. h, Line plots showing average RNA and H3K27ac signals of three gene clusters ranked by gene expression levels as in (g). i, Heatmaps showing RNA (left) and H3K27ac (right) signals at gene-correlated peaks in YS cells along the pseudotime during EHT. The rows represent 2,008 non-differential genes. j, Line plots showing average RNA, H3K27ac signals of three gene clusters ranked by gene expression levels as in (i). k-l, Heatmaps showing the TF motif scores calculated using chromVAR with default parameters (g) and gene expression (j) during AGM-EHT. i-j, Heatmaps showing the TF motif scores calculated using chromVAR with default parameters (i) and gene expression (j) during YS-EHT. k, Inferred enhancer-to-target links for Runx1 in YS-EC, YS-HEC, AGM-earlyEC, and AGM-HEC. The enhancer-to-gene relationships were computed using enhancerToGene function in ScoMAP, and the links were plotted by Cicero.

Source data

Supplementary information

Supplementary Information

Supplementary Note

Reporting Summary

Supplementary Table 1

The sequences of custom primers used in this work.

Supplementary Table 2

Data quality of all CoTECH experiments in this study.

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 Fig. 6

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

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Xiong, H., Luo, Y., Wang, Q. et al. Single-cell joint detection of chromatin occupancy and transcriptome enables higher-dimensional epigenomic reconstructions. Nat Methods 18, 652–660 (2021). https://doi.org/10.1038/s41592-021-01129-z

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41592-021-01129-z

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