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.

  • Perspective
  • Published:

Single-cell and single-molecule epigenomics to uncover genome regulation at unprecedented resolution

Abstract

Recent advances in single-cell and single-molecule epigenomic technologies now enable the study of genome regulation and dynamics at unprecedented resolution. In this Perspective, we highlight some of these transformative technologies and discuss how they have been used to identify new modes of gene regulation. We also contrast these assays with recent advances in single-cell transcriptomics and argue for the essential role of epigenomic technologies in both understanding cellular diversity and discovering gene regulatory mechanisms. In addition, we provide our view on the next generation of biological tools that we expect will open new avenues for elucidating the fundamental principles of gene regulation. Overall, this Perspective motivates the use of these high-resolution epigenomic technologies for mapping cell states and understanding regulatory diversity at single-molecule resolution within single cells.

This is a preview of subscription content, access via your institution

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: Dynamic regulatory changes in development.
Fig. 2: Cis and trans modes of regulatory heterogeneity.
Fig. 3: Future approaches for uncovering single-cell regulatory principles.

Similar content being viewed by others

References

  1. ENCODE Project Consortium. An integrated encyclopedia of DNA elements in the human genome. Nature 489, 57–74 (2012).

    Article  CAS  Google Scholar 

  2. Roadmap Epigenomics Consortium. et al. Integrative analysis of 111 reference human epigenomes. Nature 518, 317–330 (2015).

    Article  CAS  PubMed Central  Google Scholar 

  3. Stunnenberg, H. G. & Hirst, M. The International Human Epigenome Consortium: a blueprint for scientific collaboration and discovery. Cell 167, 1145–1149 (2016).

    Article  CAS  PubMed  Google Scholar 

  4. Thurman, R. E. et al. The accessible chromatin landscape of the human genome. Nature 489, 75–82 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. 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 

  6. Tanay, A. & Regev, A. Scaling single-cell genomics from phenomenology to mechanism. Nature 541, 331–338 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. 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 

  8. Shema, E. et al. Single-molecule decoding of combinatorially modified nucleosomes. Science 352, 717–721 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Schwartzman, O. & Tanay, A. Single-cell epigenomics: techniques and emerging applications. Nat. Rev. Genet. 16, 716–726 (2015).

    Article  CAS  PubMed  Google Scholar 

  10. 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 

  11. 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 

  12. Mulqueen, R. M. et al. Highly scalable generation of DNA methylation profiles in single cells. Nat. Biotechnol. 36, 428–431 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Cusanovich, D. A. et al. The cis-regulatory dynamics of embryonic development at single-cell resolution. Nature 555, 538–542 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. 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 

  15. 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 

  16. Prakadan, S. M., Shalek, A. K. & Weitz, D. A. Scaling by shrinking: empowering single-cell ‘omics’ with microfluidic devices. Nat. Rev. Genet. 18, 345–361 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Crosetto, N., Bienko, M. & van Oudenaarden, A. Spatially resolved transcriptomics and beyond. Nat. Rev. Genet. 16, 57–66 (2015).

    Article  CAS  PubMed  Google Scholar 

  18. Pliner, H. et al. Cicero predicts cis-regulatory DNA interactions from single-cell chromatin accessibility data.Mol. Cell 71, 858–871.e8 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Buenrostro, J. D. et al. Integrated single-cell analysis maps the continuous regulatory landscape of human hematopoietic differentiation. Cell 173, 1535–1548.e16 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. 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 

  21. 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 

  22. 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 

  23. Long, H. K., Prescott, S. L. & Wysocka, J. Ever-changing landscapes: transcriptional enhancers in development and evolution. Cell 167, 1170–1187 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Graf, T. & Enver, T. Forcing cells to change lineages. Nature 462, 587–594 (2009).

    Article  CAS  PubMed  Google Scholar 

  25. Takahashi, K. & Yamanaka, S. Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell 126, 663–676 (2006).

    Article  CAS  PubMed  Google Scholar 

  26. Shapiro, M. D. et al. Genetic and developmental basis of evolutionary pelvic reduction in threespine sticklebacks. Nature 428, 717–723 (2004).

    Article  CAS  PubMed  Google Scholar 

  27. Gompel, N., Prud’homme, B., Wittkopp, P. J., Kassner, V. A. & Carroll, S. B. Chance caught on the wing: cis-regulatory evolution and the origin of pigment patterns in Drosophila. Nature 433, 481–487 (2005).

    Article  CAS  PubMed  Google Scholar 

  28. Maurano, M. T. et al. Systematic localization of common disease-associated variation in regulatory DNA. Science 337, 1190–1195 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Farh, K. K.-H. et al. Genetic and epigenetic fine mapping of causal autoimmune disease variants. Nature 518, 337–343 (2015).

    Article  CAS  PubMed  Google Scholar 

  30. Javierre, B. M. et al. Lineage-specific genome architecture links enhancers and non-coding disease variants to target gene promoters. Cell 167, 1369–1384.e19 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Lareau, C. A. et al. Interrogation of human hematopoiesis at single-cell and single-variant resolution. Preprint at https://doi.org/10.1101/255224 (2018).

  32. Yu, V. W. C. et al. Epigenetic memory underlies cell-autonomous heterogeneous behavior of hematopoietic stem cells. Cell 167, 1310–1322.e17 (2016).

    Article  CAS  PubMed  Google Scholar 

  33. John, S. et al. Chromatin accessibility pre-determines glucocorticoid receptor binding patterns. Nat. Genet. 43, 264–268 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Rabani, M. et al. Metabolic labeling of RNA uncovers principles of RNA production and degradation dynamics in mammalian cells. Nat. Biotechnol. 29, 436–442 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Angermueller, C. et al. Parallel single-cell sequencing links transcriptional and epigenetic heterogeneity. Nat. Methods 13, 229–232 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. 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  CAS  PubMed  PubMed Central  Google Scholar 

  37. Trapnell, C. et al. The dynamics and regulators of cell fate decisions are revealed by pseudotemporal ordering of single cells. Nat. Biotechnol. 32, 381–386 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Bar-Joseph, Z., Gitter, A. & Simon, I. Studying and modelling dynamic biological processes using time-series gene expression data. Nat. Rev. Genet. 13, 552–564 (2012).

    Article  CAS  PubMed  Google Scholar 

  39. Soufi, A. et al. Pioneer transcription factors target partial DNA motifs on nucleosomes to initiate reprogramming. Cell 161, 555–568 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Boyer, L. A. et al. Core transcriptional regulatory circuitry in human embryonic stem cells. Cell 122, 947–956 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. 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 

  42. Imayoshi, I. et al. Oscillatory control of factors determining multipotency and fate in mouse neural progenitors. Science 342, 1203–1208 (2013).

    Article  CAS  PubMed  Google Scholar 

  43. Levine, J. H., Lin, Y. & Elowitz, M. B. Functional roles of pulsing in genetic circuits. Science 342, 1193–1200 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Zhou, X. & O’Shea, E. K. Integrated approaches reveal determinants of genome-wide binding and function of the transcription factor Pho4. Mol. Cell 42, 826–836 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Levo, M. et al. Systematic investigation of transcription factor activity in the context of chromatin using massively parallel binding and expression assays. Mol. Cell 65, 604–617.e6 (2017).

    Article  CAS  PubMed  Google Scholar 

  46. Raj, A. & van Oudenaarden, A. Nature, nurture, or chance: stochastic gene expression and its consequences. Cell 135, 216–226 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Giorgetti, L. et al. Predictive polymer modeling reveals coupled fluctuations in chromosome conformation and transcription. Cell 157, 950–963 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Bartman, C. R., Hsu, S. C., Hsiung, C. C.-S., Raj, A. & Blobel, G. A. Enhancer regulation of transcriptional bursting parameters revealed by forced chromatin looping. Mol. Cell 62, 237–247 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Fukaya, T., Lim, B. & Levine, M. Enhancer control of transcriptional bursting. Cell 166, 358–368 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Frankel, N. et al. Phenotypic robustness conferred by apparently redundant transcriptional enhancers. Nature 466, 490–493 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Chen, K. H., Boettiger, A. N., Moffitt, J. R., Wang, S. & Zhuang, X. RNA imaging. Spatially resolved, highly multiplexed RNA profiling in single cells. Science 348, aaa6090 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Wang, S. et al. Spatial organization of chromatin domains and compartments in single chromosomes. Science 353, 598–602 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Ernst, J. & Kellis, M. Large-scale imputation of epigenomic datasets for systematic annotation of diverse human tissues. Nat. Biotechnol. 33, 364–376 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Woodworth, M. B., Girskis, K. M. & Walsh, C. A. Building a lineage from single cells: genetic techniques for cell lineage tracking. Nat. Rev. Genet. 18, 230–244 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Chen, B. et al. Dynamic imaging of genomic loci in living human cells by an optimized CRISPR/Cas system. Cell 155, 1479–1491 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Gu, B. et al. Transcription-coupled changes in nuclear mobility of mammalian cis-regulatory elements. Science 359, 1050–1055 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Smallwood, S. A. et al. Single-cell genome-wide bisulfite sequencing for assessing epigenetic heterogeneity. Nat. Methods 11, 817–820 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. 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 

  59. 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 

  60. 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 

  61. 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 

  62. Ramani, V. et al. Massively multiplex single-cell Hi-C. Nat. Methods 14, 263–266 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Flyamer, I. M. et al. Single-nucleus Hi-C reveals unique chromatin reorganization at oocyte-to-zygote transition. Nature 544, 110–114 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Kind, J. et al. Genome-wide maps of nuclear lamina interactions in single human cells. Cell 163, 134–147 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Blosser, T. R., Yang, J. G., Stone, M. D., Narlikar, G. J. & Zhuang, X. Dynamics of nucleosome remodelling by individual ACF complexes. Nature 462, 1022–1027 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Liu, Z., Lavis, L. D. & Betzig, E. Imaging live-cell dynamics and structure at the single-molecule level. Mol. Cell 58, 644–659 (2015).

    Article  CAS  PubMed  Google Scholar 

  67. Huang, B., Babcock, H. & Zhuang, X. Breaking the diffraction barrier: super-resolution imaging of cells. Cell 143, 1047–1058 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Boettiger, A. N. et al. Super-resolution imaging reveals distinct chromatin folding for different epigenetic states. Nature 529, 418–422 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Spitz, F. & Furlong, E. E. M. Transcription factors: from enhancer binding to developmental control. Nat. Rev. Genet. 13, 613–626 (2012).

    Article  CAS  PubMed  Google Scholar 

  70. Levo, M. & Segal, E. In pursuit of design principles of regulatory sequences. Nat. Rev. Genet. 15, 453–468 (2014).

    Article  CAS  PubMed  Google Scholar 

  71. Schübeler, D. Function and information content of DNA methylation. Nature 517, 321–326 (2015).

    Article  CAS  PubMed  Google Scholar 

  72. Strahl, B. D. & Allis, C. D. The language of covalent histone modifications. Nature 403, 41–45 (2000).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank L. Gaskell for insightful comments throughout the composition of this manuscript. E.S. is supported by the Jane Coffin Childs Memorial Fund for Medical Research. B.E.B. is an American Cancer Society Research Professor and is supported by funds from the National Human Genome Research Institute, the National Cancer Institute and the NIH Common Fund. J.D.B. acknowledges support from the Harvard Society of Fellows and Broad Institute Fellowship. J.D.B also acknowledges the Allen Institute and the NIH R21HG009749 for funding.

Author information

Authors and Affiliations

Authors

Contributions

E.S., B.E.B. and J.D.B conceived and wrote the manuscript.

Corresponding author

Correspondence to Jason D. Buenrostro.

Ethics declarations

Competing interests

E.S., B.E.B. and J.D.B have filed patents covering single-cell or single-molecule technologies. B.E.B. owns equity in Fulcrum Therapeutics, 1CellBio Inc, Nohla Therapeutics and HiFiBio Inc., and is an advisor for Fulcrum Therapeutics, HiFiBio Inc and Cell Signaling Technologies.

Additional information

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

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

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). https://doi.org/10.1038/s41588-018-0290-x

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41588-018-0290-x

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