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.

  • Protocol
  • Published:

Transcription factor chromatin profiling genome-wide using uliCUT&RUN in single cells and individual blastocysts

Abstract

Determining chromatin-associated protein localization across the genome has provided insight into the functions of DNA-binding proteins and their connections to disease. However, established protocols requiring large quantities of cell or tissue samples currently limit applications for clinical and biomedical research in this field. Furthermore, most technologies have been optimized to assess abundant histone protein localization, prohibiting the investigation of nonhistone protein localization in low cell numbers. We recently described a protocol to profile chromatin-associated protein localization in as low as one cell: ultra-low-input cleavage under targets and release using nuclease (uliCUT&RUN). Optimized from chromatin immunocleavage and CUT&RUN, uliCUT&RUN is a tethered enzyme-based protocol that utilizes a combination of recombinant protein, antibody recognition and stringent purification to selectively target proteins of interest and isolate the associated DNA. Performed in native conditions, uliCUT&RUN profiles protein localization to chromatin with low input and high precision. Compared with other profiling technologies, uliCUT&RUN can determine nonhistone protein chromatin occupancies in low cell numbers, permitting the investigation into the molecular functions of a range of DNA-binding proteins within rare samples. From sample preparation to sequencing library submission, the uliCUT&RUN protocol takes <2 d to perform, with the accompanying data analysis timeline dependent on experience level.

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 sample preparation for uliCUT&RUN. uliCUT&RUN is amenable to many sources of low-input material.
Fig. 2: Overview of the uliCUT&RUN approach.
Fig. 3: Overview of the uliCUT&RUN bioinformatic analysis pipeline.
Fig. 4: Example quality controls for uliCUT&RUN libraries.
Fig. 5: Expected results for 50 ES cell uliCUT&RUN for TFs.
Fig. 6: Expected results for CTCF blastocyst or single-cell uliCUT&RUN.

Similar content being viewed by others

Data availability

The GEO accession number for the sequencing data reported in this paper is GSE111121, originally generated for ref. 13.

Code availability

Code for sequencing data analysis is available at https://github.com/bjp86/Patty_uliCUTandRUNAnalysis.

References

  1. 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 

  2. Villar, D., Flicek, P. & Odom, D. T. Evolution of transcription factor binding in metazoans—mechanisms and functional implications. Nat. Rev. Genet. 15, 221–233 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Mitsis, T. et al. Transcription factors and evolution: an integral part of gene expression (Review). World Acad. Sci. J. 3–8 (2020)

  4. Andersson, R. & Sandelin, A. Determinants of enhancer and promoter activities of regulatory elements. Nat. Rev. Genet. 21, 71–87 (2020).

    Article  CAS  PubMed  Google Scholar 

  5. Whyte, W. A. et al. Master transcription factors and mediator establish super-enhancers at key cell identity genes. Cell 153, 307–319 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Lambert, S. A. et al. The human transcription factors. Cell 172, 650–665 (2018).

    Article  CAS  PubMed  Google Scholar 

  7. Zaret, K. S. & Mango, S. E. Pioneer transcription factors, chromatin dynamics, and cell fate control. Curr. Opin. Genet. Dev. 37, 76–81 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Iwafuchi-Doi, M. & Zaret, K. S. Cell fate control by pioneer transcription factors. Dev 143, 1833–1837 (2016).

    Article  CAS  Google Scholar 

  9. Hu, P., Zhang, W., Xin, H. & Deng, G. Single cell isolation and analysis. Front. Cell Dev. Biol. 4, 1–12 (2016).

    Article  Google Scholar 

  10. Pijuan-Sala, B. et al. A single-cell molecular map of mouse gastrulation and early organogenesis. Nature 566, 490–495 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Cheng, S. et al. Single-cell RNA-seq reveals cellular heterogeneity of pluripotency transition and X chromosome dynamics during early mouse development. Cell Rep 26, 2593–2607.e3 (2019).

    Article  CAS  PubMed  Google Scholar 

  12. Petropoulos, S. et al. Single-cell RNA-seq reveals lineage and X chromosome dynamics in human preimplantation embryos. Cell 165, 1012–1026 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Hainer, S. J., Bošković, A., McCannell, K. N., Rando, O. J. & Fazzio, T. G. Profiling of pluripotency factors in single cells and early embryos. Cell 177, 1319–1329.e11 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Hainer, S. J. & Fazzio, T. G. High-resolution chromatin profiling using CUT&RUN. Curr. Protoc. Mol. Biol. 126, 1–22 (2019).

    Article  Google Scholar 

  15. Skene, P. J. & Henikoff, S. A simple method for generating high resolution maps of genome-wide protein binding. eLife 4, 1–9 (2015).

    Article  Google Scholar 

  16. Xu, J. et al. Super-resolution imaging reveals the evolution of higher-order chromatin folding in early carcinogenesis. Nat. Commun. 11, 1–17 (2020).

    Google Scholar 

  17. Schmid, M., Durussel, T. & Laemmli, U. K. ChIC and ChEC: genomic mapping of chromatin proteins. Mol. Cell 16, 147–157 (2004).

    CAS  PubMed  Google Scholar 

  18. Solomon, M. J. & Varshavsky, A. Formaldehyde-mediated DNA-protein crosslinking: a probe for in vivo chromatin structures. Proc. Natl Acad. Sci. USA 82, 6470–6474 (1985).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Gilmour, D. S. & Lis, J. T. In vivo interactions of RNA polymerase II with genes of Drosophila melanogaster. Mol. Cell. Biol. 5, 2009–2018 (1985).

    CAS  PubMed  PubMed Central  Google Scholar 

  20. Gilmour, D. S. & Lis, J. T. Detecting protein-DNA interactions in vivo: distribution of RNA polymerase on specific bacterial genes. Proc. Natl Acad. Sci. USA 81, 4275–4279 (1984).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Irvine, R. A., Lin, I. G. & Hsieh, C.-L. DNA methylation has a local effect on transcription and histone acetylation. Mol. Cell. Biol. 22, 6689–6696 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Albert, I. et al. Translational and rotational settings of H2A.Z nucleosomes across the Saccharomyces cerevisiae genome. Nature 446, 572–576 (2007).

    Article  CAS  PubMed  Google Scholar 

  23. Dingwall, C., Lomonossoff, G. P. & Laskey, R. A. High sequence specificity of micrococcal nuclease. Nucleic Acids Res 9, 2659–2674 (1981).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Hörz, W. & Altenburger, W. Sequence specific cleavage of DNA by micrococcal nuclease. Nucleic Acids Res 9, 2643–2658 (1981).

    Article  PubMed  PubMed Central  Google Scholar 

  25. Klein, D. C. & Hainer, S. J. Genomic methods in profiling DNA accessibility and factor localization. Chromosom. Res. 28, 69–85 (2020).

    Article  CAS  Google Scholar 

  26. Fosslie, M. et al. Going low to reach high: small-scale ChIP-seq maps new terrain. Wiley Interdiscip. Rev. Syst. Biol. Med. 12, 1–24 (2020).

    Article  Google Scholar 

  27. 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 

  28. Buenrostro, J. D., Wu, B., Chang, H. Y. & Greenleaf, W. J. ATAC-seq: a method for assaying chromatin accessibility genome-wide. Curr. Protoc. Mol. Biol. 2015, 21.29.1–21.29.9 (2015).

    Google Scholar 

  29. 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 

  30. Schmidl, C., Rendeiro, A. F., Sheffield, N. C. & Bock, C. ChIPmentation: fast, robust, low-input ChIP-seq for histones and transcription factors. Nat. Methods 12, 963–965 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Cao, Z., Chen, C., He, B., Tan, K. & Lu, C. A microfluidic device for epigenomic profiling using 100 cells. Nat. Methods 12, 959–962 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Shankaranarayanan, P. et al. Single-tube linear DNA amplification (LinDA) for robust ChIP-seq. Nat. Methods 8, 565–568 (2011).

    Article  CAS  PubMed  Google Scholar 

  33. 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 

  34. 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 

  35. Rhee, H. S. & Pugh, B. F. Comprehensive genome-wide protein-DNA interactions detected at single-nucleotide resolution. Cell 147, 1408–1419 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. He, Q., Johnston, J. & Zeitlinger, J. ChIP-nexus enables improved detection of in vivo transcription factor binding footprints. Nat. Biotechnol. 33, 395–401 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Keller, C. A. et al. Effects of sheared chromatin length on ChIP-seq quality and sensitivity. G3 (Bethesda) https://doi.org/10.1093/g3journal/jkab101 (2021).

  38. Steensel, B. V. & Henikoff, S. Identification of in vivo DNA targets of chromatin proteins using tethered Dam methyltransferase. Nat. Biotechnol. 18, 424–428 (2000).

    Article  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  40. Zentner, G. E., Kasinathan, S., Xin, B., Rohs, R. & Henikoff, S. ChEC-seq kinetics discriminates transcription factor binding sites by DNA sequence and shape in vivo. Nat. Commun. 6, (2015).

  41. Kaya-Okur, H. S. et al. CUT&Tag for efficient epigenomic profiling of small samples and single cells. Nat. Commun. 10, 1–10 (2019).

    Article  CAS  Google Scholar 

  42. Carter, B. et al. Mapping histone modifications in low cell number and single cells using antibody-guided chromatin tagmentation (ACT-seq). Nat. Commun. 10, 1–5 (2019).

    Article  CAS  Google Scholar 

  43. Liu, B. et al. The landscape of RNA Pol II binding reveals a stepwise transition during ZGA. Nature 587, 139–144 (2020).

    Article  CAS  PubMed  Google Scholar 

  44. 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 

  45. Skene, P. J. & Henikoff, S. An efficient targeted nuclease strategy for high-resolution mapping of DNA binding sites. eLife 6, 1–35 (2017).

    Article  Google Scholar 

  46. Skene, P. J., Henikoff, J. G. & Henikoff, S. Targeted in situ genome-wide profiling with high efficiency for low cell numbers. Nat. Protoc. 13, 1006–1019 (2018).

    Article  CAS  PubMed  Google Scholar 

  47. Kaya-Okur, H. S., Janssens, D. H., Henikoff, J. G., Ahmad, K. & Henikoff, S. Efficient low-cost chromatin profiling with CUT&Tag. Nat. Protoc. 15, 3264–3283 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Skene, P. J. & Henikoff, S. A simple method for generating high resolution maps of genome-wide protein binding. eLife 4, 1–9 (2017).

    Google Scholar 

  49. Meers, M. P., Bryson, T. D., Henikoff, J. G. & Henikoff, S. Improved cut&run chromatin profiling tools. eLife 8, 1–16 (2019).

    Article  Google Scholar 

  50. 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 

  51. Shah, R. N. et al. Examining the roles of H3K4 methylation states with systematically characterized antibodies. Mol. Cell 72, 162–177.e7 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Zheng, X. Y. & Gehring, M. Low-input chromatin profiling in Arabidopsis endosperm using CUT&RUN. Plant Reprod 32, 63–75 (2019).

    Article  CAS  PubMed  Google Scholar 

  53. Chereji, R. V., Bryson, T. D. & Henikoff, S. Quantitative MNase-seq accurately maps nucleosome occupancy levels. Genome Biol. 20, 198 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  54. Oomen, M. E., Hansen, A. S., Liu, Y., Darzacq, X. & Dekker, J. CTCF sites display cell cycle-dependent dynamics in factor binding and nucleosome positioning. Genome Res. 29, 236–249 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Janssens, D. H. et al. Automated in situ chromatin profiling efficiently resolves cell types and gene regulatory programs. Epigenetics Chromatin 11, 1–14 (2018).

    Article  Google Scholar 

  56. Meers, M. P., Tenenbaum, D. & Henikoff, S. Peak calling by Sparse Enrichment Analysis for CUT&RUN chromatin profiling. Epigenetics Chromatin 12, 1–11 (2019).

    Article  CAS  Google Scholar 

  57. Zhu, Q., Liu, N., Orkin, S. H. & Yuan, G. C. CUT and RUNTools: a flexible pipeline for CUT and RUN processing and footprint analysis. Genome Biol. 20, 1–12 (2019).

    Article  Google Scholar 

  58. Baek, S. & Lee, I. Single-cell ATAC sequencing analysis: from data preprocessing to hypothesis generation. Comput. Struct. Biotechnol. J. 18, 1429–1439 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Urrutia, E., Chen, L., Zhou, H. & Jiang, Y. Destin: toolkit for single-cell analysis of chromatin accessibility. Bioinformatics 35, 3818–3820 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Cai, S., Georgakilas, G. K., Johnson, J. L. & Vahedi, G. A cosine similarity-based method to infer variability of chromatin accessibility at the single-cell level. Front. Genet. 9, 1–10 (2018).

    Article  Google Scholar 

  61. Zamanighomi, M. et al. Unsupervised clustering and epigenetic classification of single cells. Nat. Commun. 9, 1–8 (2018).

    Article  CAS  Google Scholar 

  62. Baker, S. M., Rogerson, C., Hayes, A., Sharrocks, A. D. & Rattray, M. Classifying cells with Scasat, a single-cell ATAC-seq analysis tool. Nucleic Acids Res. 47, (2019).

  63. Li, H. et al. The sequence alignment/map format and SAMtools. Bioinformatics 25, 2078–2079 (2009).

    Article  PubMed  PubMed Central  Google Scholar 

  64. 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 

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

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Luo, C. et al. Superovulation strategies for 6 commonly used mouse strains. J. Am. Assoc. Lab. Anim. Sci. 50, 471–478 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  68. Adli, M. & Bernstein, B. E. Whole-genome chromatin profiling from limited numbers of cells using nano-ChIP-seq. Nat. Protoc. 6, 1656–1668 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Zwart, W. et al. A carrier-assisted ChIP-seq method for estrogen receptor-chromatin interactions from breast cancer core needle biopsy samples. BMC Genomics 14, (2013).

  70. Valensisi, C., Liao, J. L., Andrus, C., Battle, S. L. & Hawkins, R. D. cChIP-seq: a robust small-scale method for investigation of histone modifications. BMC Genomics 16, 1–11 (2015).

    Article  Google Scholar 

  71. Ng, J. H. et al. In vivo epigenomic profiling of germ cells reveals germ cell molecular signatures. Dev. Cell 24, 324–333 (2013).

    Article  CAS  PubMed  Google Scholar 

  72. Liu, X. et al. Distinct features of H3K4me3 and H3K27me3 chromatin domains in pre-implantation embryos. Nature 537, 558–562 (2016).

    Article  CAS  PubMed  Google Scholar 

  73. Lara-Astiaso, D. et al. Immunogenetics. Chromatin state dynamics during blood formation. Science 345, 943–949 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. van Galen, P. et al. a multiplexed system for quantitative comparisons of chromatin landscapes. Mol. Cell 61, 170–180 (2016).

    Article  PubMed  Google Scholar 

  75. Zheng, X. et al. Low-cell-number epigenome profiling aids the study of lens aging and hematopoiesis. Cell Rep. 13, 1505–1518 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  76. Dahl, J. A. et al. Broad histone H3K4me3 domains in mouse oocytes modulate maternal-to-zygotic transition. Nature 537, 548–552 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. Zhang, B. et al. Allelic reprogramming of the histone modification H3K4me3 in early mammalian development. Nature 537, 553–557 (2016).

    Article  CAS  PubMed  Google Scholar 

  78. Zarnegar, M. A., Reinitz, F., Newman, A. M. & Clarke, M. F. Targeted chromatin ligation, a robust epigenetic profiling technique for small cell numbers. Nucleic Acids Res. 45, 1–9 (2017).

    Article  Google Scholar 

  79. 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 

  80. Akalin, A., Franke, V., Vlahoviček, K., Mason, C. E. & Schübeler, D. Genomation: a toolkit to summarize, annotate and visualize genomic intervals. Bioinformatics 31, 1127–1129 (2015).

    Article  CAS  PubMed  Google Scholar 

  81. Stempor, P. & Ahringer, J. SeqPlots—interactive software for exploratory data analyses, pattern discovery and visualization in genomics. Wellcome Open Res. 1, 14 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

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

  83. Bailey, T. L. et al. MEME Suite: tools for motif discovery and searching. Nucleic Acids Res. 37, 202–208 (2009).

    Article  Google Scholar 

  84. Chen, X. et al. Integration of external signaling pathways with the core transcriptional network in embryonic stem cells. Cell 133, 1106–1117 (2008).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank the Henikoff group for the original development of CUT&RUN and discussion regarding application. We thank A. Boskovic and T. Fazzio for assistance in development and application of uliCUT&RUN. We thank members of the Hainer lab for helpful comments on the manuscript. This work was supported by the National Institutes of Health grant R35GM133732 to S.J.H.

Author information

Authors and Affiliations

Authors

Contributions

S.J.H. optimized the protocol and performed experiments. B.J.P. analyzed the data with assistance from S.J.H., B.J.P. and S.J.H wrote the manuscript.

Corresponding author

Correspondence to Sarah J. Hainer.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Peer review information Nature Protocols thanks Sebastian Pott and the other, anonymous reviewer(s) for their contribution to the peer review of this work.

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

Related links

Key references using this protocol

Hainer, S. et al. Cell 177, 1319–1329.e11 (2019): https://doi.org/10.1016/j.cell.2019.03.014

Xu, J. et al. Nat. Commun. 11, 1899 (2020): https://doi.org/10.1038/s41467-020-15718-7

Supplementary information

Supplementary Information

Supplementary Methods and Supplementary Figs. 1 and 2.

Reporting Summary

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Patty, B.J., Hainer, S.J. Transcription factor chromatin profiling genome-wide using uliCUT&RUN in single cells and individual blastocysts. Nat Protoc 16, 2633–2666 (2021). https://doi.org/10.1038/s41596-021-00516-2

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41596-021-00516-2

This article is cited by

Comments

By submitting a comment you agree to abide by our Terms and Community Guidelines. If you find something abusive or that does not comply with our terms or guidelines please flag it as inappropriate.

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing