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:

Profiling the genetic determinants of chromatin accessibility with scalable single-cell CRISPR screens

Abstract

CRISPR screens have been used to connect genetic perturbations with changes in gene expression and phenotypes. Here we describe a CRISPR-based, single-cell combinatorial indexing assay for transposase-accessible chromatin (CRISPR–sciATAC) to link genetic perturbations to genome-wide chromatin accessibility in a large number of cells. In human myelogenous leukemia cells, we apply CRISPR–sciATAC to target 105 chromatin-related genes, generating chromatin accessibility data for ~30,000 single cells. We correlate the loss of specific chromatin remodelers with changes in accessibility globally and at the binding sites of individual transcription factors (TFs). For example, we show that loss of the H3K27 methyltransferase EZH2 increases accessibility at heterochromatic regions involved in embryonic development and triggers expression of genes in the HOXA and HOXD clusters. At a subset of regulatory sites, we also analyze changes in nucleosome spacing following the loss of chromatin remodelers. CRISPR–sciATAC is a high-throughput, single-cell method for studying the effect of genetic perturbations on chromatin in normal and disease states.

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: CRISPR screens with CRISPR–sciATAC enable the joint capture of chromatin accessibility profiles and CRISPR perturbations.
Fig. 2: CRISPR–sciATAC reveals changes in accessibility at HOX genes following loss of EZH2.
Fig. 3: CRISPR–sciATAC screen targeting 17 chromatin remodeling complexes uncovers widespread disruptions in accessibility following SWI/SNF disruption.
Fig. 4: Nucleosome dynamics around TFBS following CRISPR targeting of chromatin remodelers.

Similar content being viewed by others

Data availability

Processed and raw data can be downloaded from NCBI GEO (PRJNA674902, GSE161002).

Code availability

The scripts and pipeline for the analysis can be found at https://gitlab.com/sanjanalab/crispr-sciatac.

References

  1. Flavahan, W. A., Gaskell, E. & Bernstein, B. E. Epigenetic plasticity and the hallmarks of cancer. Science 357, eaal2380 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  2. Datlinger, P. et al. Pooled CRISPR screening with single-cell transcriptome readout. Nat. Methods 14, 297–301 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Adamson, B. et al. A multiplexed single-cell CRISPR screening platform enables systematic dissection of the unfolded protein response. Cell 167, 1867–1882 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Dixit, A. et al. Perturb-seq: dissecting molecular circuits with scalable single-cell RNA profiling of pooled genetic screens. Cell 167, 1853–1866 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Jaitin, D. A. et al. Dissecting immune circuits by linking CRISPR-pooled screens with single-cell RNA-seq. Cell 167, 1883–1896 (2016).

    Article  CAS  PubMed  Google Scholar 

  6. Rubin, A. J. et al. Coupled single-cell CRISPR screening and epigenomic profiling reveals causal gene regulatory networks. Cell 176, 361–376 (2019).

    Article  CAS  PubMed  Google Scholar 

  7. 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 

  8. Forbes, S. A. et al. COSMIC: somatic cancer genetics at high-resolution. Nucleic Acids Res. 45, D777–D783 (2017).

    Article  CAS  PubMed  Google Scholar 

  9. Kolde, R., Laur, S., Adler, P. & Vilo, J. Robust rank aggregation for gene list integration and meta-analysis. Bioinformatics 28, 573–580 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Wang, T. et al. Identification and characterization of essential genes in the human genome. Science 350, 1096–1101 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Margueron, R. & Reinberg, D. The Polycomb complex PRC2 and its mark in life. Nature 469, 343–349 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Margueron, R. et al. Ezh1 and Ezh2 maintain repressive chromatin through different mechanisms. Mol. Cell 32, 503–518 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Mathelier, A. et al. JASPAR 2016: a major expansion and update of the open-access database of transcription factor binding profiles. Nucleic Acids Res. 44, D110–D115 (2016).

    Article  CAS  PubMed  Google Scholar 

  14. 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 

  15. Medvedeva, Y. A. et al. EpiFactors: a comprehensive database of human epigenetic factors and complexes. Database (Oxford) 2015, bav067 (2015).

  16. Lejeune, E. et al. The chromatin-remodeling factor FACT contributes to centromeric heterochromatin independently of RNAi. Curr. Biol. 17, 1219–1224 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Mathur, R. et al. ARID1A loss impairs enhancer-mediated gene regulation and drives colon cancer in mice. Nat. Genet. 49, 296–302 (2017).

    Article  CAS  PubMed  Google Scholar 

  18. Nord, A. S. et al. Rapid and pervasive changes in genome-wide enhancer usage during mammalian development. Cell 155, 1521–1531 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Vierbuchen, T. et al. AP-1 transcription factors and the BAF complex mediate signal-dependent enhancer selection. Mol. Cell 68, 1067–1082 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Hakimi, M. A. et al. A chromatin remodelling complex that loads cohesin onto human chromosomes. Nature 123, 3175–3184 (2002).

    Google Scholar 

  21. Wen, Z., Huang, Z. T., Zhang, R. & Peng, C. ZNF143 is a regulator of chromatin loop. Cell Biol. Toxicol. 34, 471–478 (2018).

    Article  CAS  PubMed  Google Scholar 

  22. Swiers, G., Patient, R. & Loose, M. Genetic regulatory networks programming hematopoietic stem cells and erythroid lineage specification. Dev. Biol. 294, 525–540 (2006).

    Article  CAS  PubMed  Google Scholar 

  23. Li, M. et al. Dynamic regulation of transcription factors by nucleosome remodeling. Elife 4, e06249 (2015).

    Article  PubMed Central  CAS  Google Scholar 

  24. Kundaje, A. et al. Ubiquitous heterogeneity and asymmetry of the chromatin environment at regulatory elements. Genome Res. 22, 1735–1747 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Kelso, T. W. R. et al. Chromatin accessibility underlies synthetic lethality of SWI/SNF subunits in ARID1A-mutant cancers. Elife 6, e30506 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  26. Guo, X., Chitale, P. & Sanjana, N. E. Target discovery for precision medicine using high-throughput genome engineering. Adv. Exp. Med. Biol. 1016, 123–145 (2017).

    Article  CAS  PubMed  Google Scholar 

  27. Papalexi, E. et al. Characterizing the molecular regulation of inhibitory immune checkpoints with multimodal single-cell screens. Nat. Genet. 53, 322–331 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Shalem, O. et al. Genome-scale CRISPR-Cas9 knockout screening in human cells. Science 343, 84–87 (2014).

    Article  CAS  PubMed  Google Scholar 

  29. Shi, J. et al. Discovery of cancer drug targets by CRISPR-Cas9 screening of protein domains. Nat. Biotechnol. 33, 661–667 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Meier, J. A., Zhang, F. & Sanjana, N. E. GUIDES: sgRNA design for loss-of-function screens. Nat. Methods 14, 831–832 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. 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 

  32. Thompson, J. D., Higgins, D. G. & Gibson, T. J. CLUSTAL W: improving the sensitivity of progressive multiple sequence alignment through sequence weighting, position-specific gap penalties and weight matrix choice. Nucleic Acids Res. 22, 4673–4680 (1994).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Goryshin, I. Y. & Reznikoff, W. S. Tn5 in vitro transposition. J. Biol. Chem. 273, 7367–7374 (1998).

    Article  CAS  PubMed  Google Scholar 

  34. Nørholm, M. A mutant Pfu DNA polymerase designed for advanced uracil-excision DNA engineering. BMC Biotechnol. 10, 21 (2010).

  35. 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 

  36. Richter, K. N. et al. Glyoxal as an alternative fixative to formaldehyde in immunostaining and super‐resolution microscopy. EMBO J. 37, 139–159 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  37. Adey, A. et al. In vitro, long-range sequence information for de novo genome assembly via transposase contiguity. Genome Res. 24, 2041–2049 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Amini, S. et al. Haplotype-resolved whole-genome sequencing by contiguity-preserving transposition and combinatorial indexing. Nat. Genet. 46, 1343–1349 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Liu, X. S. et al. MAGeCK enables robust identification of essential genes from genome-scale CRISPR/Cas9 knockout screens. Genome Biol. 15, 554 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  40. 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 

  41. 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).

  42. 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 

  43. Eden, E., Navon, R., Steinfeld, I., Lipson, D. & Yakhini, Z. GOrilla: a tool for discovery and visualization of enriched GO terms in ranked gene lists. BMC Bioinformatics 10, 48 (2009).

    Article  PubMed  PubMed Central  Google Scholar 

  44. Võsa, U. et al. Unraveling the polygenic architecture of complex traits using blood eQTL meta-analysis. Preprint at bioRxiv https://doi.org/10.1101/447367 (2018).

  45. The GTEx Consortium. The GTEx Consortium atlas of genetic regulatory effects across human tissues. Science 369, 1318–1330 (2020).

  46. Quinlan, A. R. & Hall, I. M. BEDTools: a flexible suite of utilities for comparing genomic features. Bioinformatics 26, 841–842 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Duttke, S. et al. Identification and dynamic quantification of regulatory elements using total RNA. Genome Res. 29, 1836–1846 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Benjamini, Y. & Hochberg, Y. Controlling the false discovery rate: a practical and powerful approach to multiple testing. J. R. Stat. Soc. Ser. B Stat. Methodol. 57, 289–300 (1995).

    Google Scholar 

Download references

Acknowledgements

We thank the entire Sanjana laboratory for support and advice. We thank J. Morris for help with eQTL resources, M. Zaran and R. Satija for computational resources and the NYGC Sequencing Platform and NYU Biology Genomics Core for sequencing resources. BL21(DE3) cells transformed with pET-PfuX7 were kindly provided by J. Gregory. N.L.-B. is supported by a postdoctoral fellowship from the Human Frontier Science Program Organization (no. LT000672/2019-L), an EMBO long-term fellowship (no. ALTF 826-2018) and the Weizmann Institute of Science National Postdoctoral Award Program for Advancing Women in Science. N.E.S. is supported by NYU and NYGC startup funds, NIH/NHGRI (nos. R00HG008171 and DP2HG010099), NIH/NCI (no. R01CA218668), DARPA (no. D18AP00053), the Sidney Kimmel Foundation, the Melanoma Research Alliance and the Brain and Behavior Foundation.

Author information

Authors and Affiliations

Authors

Contributions

N.E.S. conceived and supervised the project. N.E.S., A.M. and N.L.-B. designed the experiments. A.M., N.L.-B., J.D., A.M.-M., C.-Y.K. and A.S. performed the experiments. N.L.-B., A.M., J.D., N.E.S., H.-H.W. and N.G.M. analyzed the data. P.S. isolated TnY. S.J. purified PhuX7. A.M., J.D., C.-Y.K., A.S., P.S. and S.J. purified TnY. N.L.-B., A.M. and N.E.S. wrote the manuscript with input from all authors.

Corresponding author

Correspondence to Neville E. Sanjana.

Ethics declarations

Competing interests

The New York Genome Center and New York University have applied for patents relating to the work in this article. N.E.S. is an adviser to Vertex.

Additional information

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

Supplementary information

Supplementary Information

Supplementary Figs. 1–14 and Sequences.

Reporting Summary

Supplementary Table 1

Number of cells in each step of the CRISPR–sciATAC protocol.

Supplementary Table 2

Sequences of oligonucleotides for CRISPR–sciATAC, CRISPR libraries and RT–qPCR.

Supplementary Table 3

Gene and gRNA enrichment from essentiality screen.

Supplementary Table 4

ENCODE ChIP data sources.

Supplementary Table 5

Histone mark differential accessibility.

Supplementary Table 6

GO enrichment results for differential accessibility in EZH2-targeted cells.

Supplementary Table 7

Transcription factor binding site differential accessibility.

Supplementary Table 8

Cost comparison between CRISPR–sciATAC and Perturb–ATAC protocols.

Supplementary Table 9

Time comparison between CRISPR–sciATAC and Perturb–ATAC protocols.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Liscovitch-Brauer, N., Montalbano, A., Deng, J. et al. Profiling the genetic determinants of chromatin accessibility with scalable single-cell CRISPR screens. Nat Biotechnol 39, 1270–1277 (2021). https://doi.org/10.1038/s41587-021-00902-x

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41587-021-00902-x

This article is cited by

Search

Quick links

Nature Briefing: Cancer

Sign up for the Nature Briefing: Cancer newsletter — what matters in cancer research, free to your inbox weekly.

Get what matters in cancer research, free to your inbox weekly. Sign up for Nature Briefing: Cancer