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.

Highly specific epigenome editing by CRISPR-Cas9 repressors for silencing of distal regulatory elements

Abstract

Epigenome editing with the CRISPR (clustered, regularly interspaced, short palindromic repeats)-Cas9 platform is a promising technology for modulating gene expression to direct cell phenotype and to dissect the causal epigenetic mechanisms of gene regulation. Fusions of nuclease-inactive dCas9 to the Krüppel-associated box (KRAB) repressor (dCas9-KRAB) can silence target gene expression, but the genome-wide specificity and the extent of heterochromatin formation catalyzed by dCas9-KRAB are not known. We targeted dCas9-KRAB to the HS2 enhancer, a distal regulatory element that orchestrates the expression of multiple globin genes, and observed highly specific induction of H3K9 trimethylation (H3K9me3) at the enhancer and decreased chromatin accessibility of both the enhancer and its promoter targets. Targeted epigenetic modification of HS2 silenced the expression of multiple globin genes, with minimal off-target changes in global gene expression. These results demonstrate that repression mediated by dCas9-KRAB is sufficiently specific to disrupt the activity of individual enhancers via local modification of the epigenome.

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

Figure 1: Silencing of downstream globin genes by dCas9-KRAB transcription factors targeted to the distal HS2 enhancer.
Figure 2: Specificity of gene regulation by dCas9-KRAB repressors targeted to the HS2 enhancer.
Figure 3: Genome-wide binding activity of dCas9 repressors targeted to the HS2 enhancer.
Figure 4: Genome-wide H3K9me3 signal in K562 cells treated with dCas9-KRAB targeted to the HS2 enhancer.
Figure 5: Changes in global chromatin landscape with dCas9-KRAB localized to the HS2 distal enhancer.

Similar content being viewed by others

Accession codes

Primary accessions

Gene Expression Omnibus

References

  1. Beerli, R.R., Dreier, B. & Barbas, C.F. III Positive and negative regulation of endogenous genes by designed transcription factors. Proc. Natl. Acad. Sci. USA 97, 1495–1500 (2000).

    CAS  PubMed  PubMed Central  Google Scholar 

  2. Snowden, A.W., Gregory, P.D., Case, C.C. & Pabo, C.O. Gene-specific targeting of H3K9 methylation is sufficient for initiating repression in vivo. Curr. Biol. 12, 2159–2166 (2002).

    CAS  PubMed  Google Scholar 

  3. Cong, L., Zhou, R., Kuo, Y.C., Cunniff, M. & Zhang, F. Comprehensive interrogation of natural TALE DNA-binding modules and transcriptional repressor domains. Nat. Commun. 3, 968 (2012).

    PubMed  Google Scholar 

  4. Konermann, S. et al. Optical control of mammalian endogenous transcription and epigenetic states. Nature 500, 472–476 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  5. Mendenhall, E.M. et al. Locus-specific editing of histone modifications at endogenous enhancers. Nat. Biotechnol. 31, 1133–1136 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  6. Gilbert, L.A. et al. CRISPR-mediated modular RNA-guided regulation of transcription in eukaryotes. Cell 154, 442–451 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  7. Perez-Pinera, P. et al. RNA-guided gene activation by CRISPR-Cas9–based transcription factors. Nat. Methods 10, 973–976 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  8. Maeder, M.L. et al. CRISPR RNA-guided activation of endogenous human genes. Nat. Methods 10, 977–979 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  9. Kearns, N.A. et al. Functional annotation of native enhancers with a Cas9-histone demethylase fusion. Nat. Methods 12, 401–403 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  10. Hilton, I.B. et al. Epigenome editing by a CRISPR-Cas9–based acetyltransferase activates genes from promoters and enhancers. Nat. Biotechnol. 33, 510–517 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  11. Maeder, M.L. et al. Targeted DNA demethylation and activation of endogenous genes using programmable TALE-TET1 fusion proteins. Nat. Biotechnol. 31, 1137–1142 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  12. Rivenbark, A.G. et al. Epigenetic reprogramming of cancer cells via targeted DNA methylation. Epigenetics 7, 350–360 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  13. Jinek, M. et al. A programmable dual-RNA-guided DNA endonuclease in adaptive bacterial immunity. Science 337, 816–821 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  14. Cong, L. et al. Multiplex genome engineering using CRISPR/Cas systems. Science 339, 819–823 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  15. Mali, P. et al. RNA-guided human genome engineering via Cas9. Science 339, 823–826 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  16. Gilbert, L.A. et al. Genome-Scale CRISPR-mediated control of gene repression and activation. Cell 159, 647–661 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  17. Konermann, S. et al. Genome-scale transcriptional activation by an engineered CRISPR-Cas9 complex. Nature 517, 583–588 (2015).

    CAS  PubMed  Google Scholar 

  18. Gao, X. et al. Comparison of TALE designer transcription factors and the CRISPR/dCas9 in regulation of gene expression by targeting enhancers. Nucleic Acids Res. 42, e155 (2014).

    PubMed  PubMed Central  Google Scholar 

  19. Chakraborty, S. et al. A CRISPR/Cas9-based system for reprogramming cell lineage specification. Stem Cell Reports 3, 940–947 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  20. Chavez, A. et al. Highly efficient Cas9-mediated transcriptional programming. Nat. Methods 12, 326–328 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  21. Nissim, L., Perli, S.D., Fridkin, A., Perez-Pinera, P. & Lu, T.K. Multiplexed and programmable regulation of gene networks with an integrated RNA and CRISPR/Cas toolkit in human cells. Mol. Cell 54, 698–710 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  23. Boyle, A.P. et al. High-resolution mapping and characterization of open chromatin across the genome. Cell 132, 311–322 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

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

  25. Sripathy, S.P., Stevens, J. & Schultz, D.C. The KAP1 corepressor functions to coordinate the assembly of de novo HP1-demarcated microenvironments of heterochromatin required for KRAB zinc finger protein-mediated transcriptional repression. Mol. Cell. Biol. 26, 8623–8638 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

  26. Groner, A.C. et al. KRAB-zinc finger proteins and KAP1 can mediate long-range transcriptional repression through heterochromatin spreading. PLoS Genet. 6, e1000869 (2010).

    PubMed  PubMed Central  Google Scholar 

  27. Schultz, D.C., Ayyanathan, K., Negorev, D., Maul, G.G. & Rauscher, F.J. 3rd SETDB1: a novel KAP-1-associated histone H3, lysine 9-specific methyltransferase that contributes to HP1-mediated silencing of euchromatic genes by KRAB zinc-finger proteins. Genes Dev. 16, 919–932 (2002).

    CAS  PubMed  PubMed Central  Google Scholar 

  28. Reynolds, N. et al. NuRD-mediated deacetylation of H3K27 facilitates recruitment of Polycomb Repressive Complex 2 to direct gene repression. EMBO J. 31, 593–605 (2012).

    CAS  PubMed  Google Scholar 

  29. Wu, X. et al. Genome-wide binding of the CRISPR endonuclease Cas9 in mammalian cells. Nat. Biotechnol. 32, 670–676 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  30. Kuscu, C., Arslan, S., Singh, R., Thorpe, J. & Adli, M. Genome-wide analysis reveals characteristics of off-target sites bound by the Cas9 endonuclease. Nat. Biotechnol. 32, 677–683 (2014).

    CAS  PubMed  Google Scholar 

  31. Grimmer, M.R. et al. Analysis of an artificial zinc finger epigenetic modulator: widespread binding but limited regulation. Nucleic Acids Res. 42, 10856–10868 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  32. Ayyanathan, K. et al. Regulated recruitment of HP1 to a euchromatic gene induces mitotically heritable, epigenetic gene silencing: a mammalian cell culture model of gene variegation. Genes Dev. 17, 1855–1869 (2003).

    CAS  PubMed  PubMed Central  Google Scholar 

  33. Hathaway, N.A. et al. Dynamics and memory of heterochromatin in living cells. Cell 149, 1447–1460 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  34. Hardison, R. et al. Locus control regions of mammalian beta-globin gene clusters: combining phylogenetic analyses and experimental results to gain functional insights. Gene 205, 73–94 (1997).

    CAS  PubMed  Google Scholar 

  35. Tuan, D., Kong, S. & Hu, K. Transcription of the hypersensitive site HS2 enhancer in erythroid cells. Proc. Natl. Acad. Sci. USA 89, 11219–11223 (1992).

    CAS  PubMed  PubMed Central  Google Scholar 

  36. McDowell, J.C. & Dean, A. Structural and functional cross-talk between a distant enhancer and the epsilon-globin gene promoter shows interdependence of the two elements in chromatin. Mol. Cell. Biol. 19, 7600–7609 (1999).

    CAS  PubMed  PubMed Central  Google Scholar 

  37. Vakoc, C.R. et al. Proximity among distant regulatory elements at the beta-globin locus requires GATA-1 and FOG-1. Mol. Cell 17, 453–462 (2005).

    CAS  PubMed  Google Scholar 

  38. Dostie, J. et al. Chromosome Conformation Capture Carbon Copy (5C): a massively parallel solution for mapping interactions between genomic elements. Genome Res. 16, 1299–1309 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

  39. Dean, A., Ley, T.J., Humphries, R.K., Fordis, M. & Schechter, A.N. Inducible transcription of five globin genes in K562 human leukemia cells. Proc. Natl. Acad. Sci. USA 80, 5515–5519 (1983).

    CAS  PubMed  PubMed Central  Google Scholar 

  40. Polstein, L.R. et al. Genome-wide specificity of DNA-binding, gene regulation, and chromatin remodeling by TALE- and CRISPR/Cas9-based transcriptional activators. Genome Res. 25, 1158–1169 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  41. O'Geen, H., Henry, I.M., Bhakta, M.S., Meckler, J.F. & Segal, D.J. A genome-wide analysis of Cas9 binding specificity using ChIP-seq and targeted sequence capture. Nucleic Acids Res. 43, 3389–3404 (2015).

    PubMed  PubMed Central  Google Scholar 

  42. Ikonomi, P. et al. Levels of GATA-1/GATA-2 transcription factors modulate expression of embryonic and fetal hemoglobins. Gene 261, 277–287 (2000).

    CAS  PubMed  Google Scholar 

  43. Deng, W. et al. Reactivation of developmentally silenced globin genes by forced chromatin looping. Cell 158, 849–860 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  44. Kabadi, A.M., Ousterout, D.G., Hilton, I.B. & Gersbach, C.A. Multiplex CRISPR/Cas9-based genome engineering from a single lentiviral vector. Nucleic Acids Res. 42, e147 (2014).

    PubMed  PubMed Central  Google Scholar 

  45. Salmon, P. & Trono, D. Production and titration of lentiviral vectors. Curr. Protoc. Neurosci. Chapter 4, Unit 4.21 (2006).

  46. Gertz, J. et al. Transposase mediated construction of RNA-seq libraries. Genome Res. 22, 134–141 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  48. Anders, S. & Huber, W. Differential expression analysis for sequence count data. Genome Biol. 11, R106 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  49. Li, H. et al. The Sequence Alignment/Map format and SAMtools. Bioinformatics 25, 2078–2079 (2009).

    PubMed  PubMed Central  Google Scholar 

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

    PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  52. Love, M.I., Huber, W. & Anders, S. Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol. 15, 550 (2014).

    PubMed  PubMed Central  Google Scholar 

  53. Song, L. & Crawford, G.E. DNase-seq: a high-resolution technique for mapping active gene regulatory elements across the genome from mammalian cells. Cold Spring Harb. Protoc. 2010 10.1101/pdb.prot5384 (February 2010).

  54. 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).

    PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank the Duke Genome Sequencing & Analysis Core for sequencing the RNA-seq, ChIP-seq and DNase-seq libraries. This work was supported by US National Institutes of Health (NIH) grants R01DA036865, U01HG007900, R21AR065956 and P30AR066527; an NIH Director's New Innovator Award (DP2OD008586); a US National Science Foundation (NSF) Faculty Early Career Development (CAREER) Award (CBET-1151035); and an American Heart Association Scientist Development grant (10SDG3060033) to C.A.G. P.I.T. was supported by an NSF Graduate Research Fellowship and an American Heart Association Mid-Atlantic Affiliate Predoctoral Fellowship.

Author information

Authors and Affiliations

Authors

Contributions

P.I.T., G.E.C., T.E.R. and C.A.G. designed experiments. P.I.T., A.M.D., A.S., N.K.S. and A.M.K. performed the experiments. P.I.T., A.M.D., L.S., A.M.K., G.E.C., T.E.R. and C.A.G. analyzed the data. P.I.T., G.E.C., T.E.R. and C.A.G. wrote the manuscript. All authors contributed to editing of the manuscript.

Corresponding authors

Correspondence to Timothy E Reddy, Gregory E Crawford or Charles A Gersbach.

Ethics declarations

Competing interests

C.A.G., G.E.C., T.E.R., P.I.T. and A.M.K. are inventors on patent applications related to genome engineering with the CRISPR-Cas9 system. C.A.G. is a scientific advisor to Editas Medicine, a company engaged in therapeutic development of genome engineering technologies.

Integrated supplementary information

Supplementary Figure 1 Screening single gRNAs targeted to the HS2 enhancer.

(a) K562 cells that were not transduced, transduced with dCas9 lentivirus, and transduced with dCas9-KRAB were transfected with a panel of 21 HS2 sgRNAs and assayed by qRT-PCR at 3 days post-transfection. (b-d) Gene expression of single gRNAs targeted to the HS2 enhancer for silencing of (d) HBE1, (c) HBG1 and HBG2 (HBG1/2), and (d) HBB by (mean ± s.e.m, n = 3 – 4 independent experiments).

Source data

Supplementary Figure 2 HBG1/2 expression after transient delivery of sgRNAs.

(a,b) Gene expression of HBG1 and HBG2 (HBG1/2) from (a) 3 to (b) 6 days after transient electroporation of sgRNA plasmids in K562 cells expressing dCas9-KRAB (mean ± s.e.m, n = 2 independent experiments).

Source data

Supplementary Figure 3 Protein silencing of HBG1 by dCas9-KRAB transcription factors targeted to the distal HS2 enhancer.

Western blot for γ-globin and GAPDH demonstrate globin silencing in K562 cells treated with dCas9-KRAB (dCK) and sgRNAs compared to non-transduced controls (No LV), no sgRNA controls, IL1RN-targeted sgRNA controls, and dCas9 (dC) with sgRNA (cropped from representative images from n = 3 biological replicates). Western blot for the FLAG epitope show dCas9 and dCas9-KRAB expression in transduced K562 cells.

Supplementary Figure 4 Specificity of gene regulation by dCas9-KRAB repressors targeted to the HS2 enhancer.

(a-e) Differential analysis was performed to evaluate the genome-wide effects of lentiviral transduction of dCas9-KRAB guided by (a) Cr4 and (b) Cr10 compared to dCas9 with the same gRNA, dCas9 guided by (c) Cr4 and (d) Cr10 compared to non-treated K562s (No LV CTL), and (e) dCas9-KRAB without gRNA compared to No LV CTL K562s.. Red data points indicate FDR < 0.01 by differential expression analysis compared to dCas9-KRAB only controls. Points labeled in blue indicate other globin genes.

Source data

Supplementary Figure 5 Genome-wide binding activity of dCas9-KRAB targeted to the HS2 enhancer.

(a,b) Differential analyses of global binding activity include comparisons of dCas9-KRAB versus dCas9 targeted by (a) Cr4 and (b) Cr10. Points labeled in red indicate FDR < 0.05 by differential DESeq analysis (n = 3 biological replicates).

Source data

Supplementary Figure 6 Effect of dCas9-KRAB localization on binding of endogenous transcription factors GATA2 and FOSL1 at HS2.

(a) The HS2 regulatory element contains a GATA2 binding site and two adjacent FOSL1 binding sites proximal to Cr4 and Cr10 target sites. (b,c) ChIP-qPCR demonstrates reduced (b) GATA2 and (c) FOSL1 binding when dCas9-KRAB was targeted to the HS2 enhancer (mean ± s.e.m). * indicates p<0.05 by Student’s t-test compared to dCas9-KRAB only control (n = 3 independent experiments).

Source data

Supplementary Figure 7 Genome-wide H3K9me3 signal in K562 cells treated with dCas9-KRAB targeted to the HS2 enhancer.

Global analysis of H3K9me3 patterns was assessed by ChIP-seq. (a,b) Volcano plots demonstrate significance (p-value) versus fold-change for dCas9-KRAB with (a) Cr4 or (b) Cr10 compared to dCas9-KRAB without sgRNA. (c-f) H3K9me3 ChIP-seq differential analysis was also performed for dCas9-KRAB versus dCas9 guided by (c,e) Cr4 or (d,f) Cr10. Points labeled red indicate FDR < 0.05 by differential expression analysis compared to dCas9-KRAB without sgRNA or dCas9 + Cr4/10.

Source data

Source data

Source data

Supplementary Figure 8 ChIP-qPCR of H3K9 trimethylation at the HS2 enhancer.

(a) ChIP-seq tracks show increased H3K9me3 signal at the HS2 enhancer (shaded area, magnified inset). An ENCODE K562 DNase I hypersensitivity DNase-seq track is included to highlight the globin LCR49. Two primer sets were designed for the HS2 enhancer for ChIP-qPCR of H3K9me3. (b) ChIP-qPCR demonstrates increased H3K9me3 when dCas9-KRAB was targeted to the HS2 enhancer (mean ± s.e.m., n = 3 independent experiments).

Source data

Supplementary Figure 9 Changes in chromatin accessibility at the globin gene locus with dCas9-KRAB localized to the HS2 distal enhancer.

(a-h) Normalized DNase-seq cut counts within an 800 bp window surrounding the (a) HS1 enhancer, (b) HS3 enhancer, (c) HS4 enhancer, (d) HS5 enhancer, (e) HBE1 promoter, (f) HBG1 promoter, (g) HBD promoter, and (h) HBB promoter are shown (mean ± s.e.m, n = 3 independent experiments). * indicates p <0.05 compared to the dCas9-KRAB only sample (Student’s t-test).

Source data

Supplementary Figure 10 Changes in global chromatin accessibility with dCas9-KRAB localized to the HS2 distal enhancer.

(a,b) Differential genome-wide analysis of changes in chromatin accessibility induced by dCas9-KRAB versus dCas9 guided by (a) Cr4 and (b) Cr10. (c,d) Volcano plots of significance (p-value) versus fold change for differential expression analysis of dCas9-KRAB compared to dCas9 guided by (c) Cr4 or (d) Cr10. Points labeled red indicate FDR < 0.05 by DESeq analysis. Points labeled in blue indicate other regions in the globin promoters or globin LCR.

Source data

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–10 and Supplementary Tables 1–16

Source data

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Thakore, P., D'Ippolito, A., Song, L. et al. Highly specific epigenome editing by CRISPR-Cas9 repressors for silencing of distal regulatory elements. Nat Methods 12, 1143–1149 (2015). https://doi.org/10.1038/nmeth.3630

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nmeth.3630

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