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.

Epigenome editing by a CRISPR-Cas9-based acetyltransferase activates genes from promoters and enhancers

Abstract

Technologies that enable targeted manipulation of epigenetic marks could be used to precisely control cell phenotype or interrogate the relationship between the epigenome and transcriptional control. Here we describe a programmable, CRISPR-Cas9-based acetyltransferase consisting of the nuclease-null dCas9 protein fused to the catalytic core of the human acetyltransferase p300. The fusion protein catalyzes acetylation of histone H3 lysine 27 at its target sites, leading to robust transcriptional activation of target genes from promoters and both proximal and distal enhancers. Gene activation by the targeted acetyltransferase was highly specific across the genome. In contrast to previous dCas9-based activators, the acetyltransferase activates genes from enhancer regions and with an individual guide RNA. We also show that the core p300 domain can be fused to other programmable DNA-binding proteins. These results support targeted acetylation as a causal mechanism of transactivation and provide a robust tool for manipulating gene regulation.

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: The dCas9p300 Core fusion protein activates transcription of endogenous genes from proximal promoter regions.
Figure 2: The dCas9p300 Core fusion protein activates transcription of endogenous genes from distal enhancer regions.
Figure 3: dCas9p300 Core-targeted transcriptional activation is specific and robust.
Figure 4: The dCas9p300 Core fusion protein acetylates chromatin at a targeted enhancer and corresponding downstream genes.
Figure 5: The dCas9p300 Core fusion protein activates transcription of endogenous genes from regulatory regions with a single gRNA.
Figure 6: The p300 Core can be targeted to genomic loci by diverse programmable DNA-binding proteins.

Similar content being viewed by others

Accession codes

Primary accessions

Gene Expression Omnibus

References

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

  2. Bernstein, B.E. et al. The NIH Roadmap Epigenomics Mapping Consortium. Nat. Biotechnol. 28, 1045–1048 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. 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).

    Article  CAS  PubMed  Google Scholar 

  4. 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).

    Article  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).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Keung, A.J., Bashor, C.J., Kiriakov, S., Collins, J.J. & Khalil, A.S. Using targeted chromatin regulators to engineer combinatorial and spatial transcriptional regulation. Cell 158, 110–120 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Hsu, P.D., Lander, E.S. & Zhang, F. Development and applications of CRISPR-Cas9 for genome engineering. Cell 157, 1262–1278 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Doudna, J.A. & Charpentier, E. Genome editing. The new frontier of genome engineering with CRISPR-Cas9. Science 346, 1258096 (2014).

    PubMed  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  15. Mali, P. et al. CAS9 transcriptional activators for target specificity screening and paired nickases for cooperative genome engineering. Nat. Biotechnol. 31, 833–838 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Qi, L.S. et al. Repurposing CRISPR as an RNA-guided platform for sequence-specific control of gene expression. Cell 152, 1173–1183 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Cheng, A.W. et al. Multiplexed activation of endogenous genes by CRISPR-on, an RNA-guided transcriptional activator system. Cell Res. 23, 1163–1171 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Farzadfard, F., Perli, S.D. & Lu, T.K. Tunable and multifunctional eukaryotic transcription factors based on CRISPR/Cas. ACS Synth. Biol. 2, 604–613 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Tanenbaum, M.E., Gilbert, L.A., Qi, L.S., Weissman, J.S. & Vale, R.D. A protein-tagging system for signal amplification in gene expression and fluorescence imaging. Cell 159, 635–646 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Chavez, A. et al. Highly efficient Cas9-mediated transcriptional programming. Nat. Methods doi:10.1038/nmeth.3312 (2 March 2015).

  24. Perez-Pinera, P. et al. Synergistic and tunable human gene activation by combinations of synthetic transcription factors. Nat. Methods 10, 239–242 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Maeder, M.L. et al. Robust, synergistic regulation of human gene expression using TALE activators. Nat. Methods 10, 243–245 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. 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).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Choy, B. & Green, M.R. Eukaryotic activators function during multiple steps of preinitiation complex assembly. Nature 366, 531–536 (1993).

    Article  CAS  PubMed  Google Scholar 

  28. Memedula, S. & Belmont, A.S. Sequential recruitment of HAT and SWI/SNF components to condensed chromatin by VP16. Curr. Biol. 13, 241–246 (2003).

    Article  CAS  PubMed  Google Scholar 

  29. Delvecchio, M., Gaucher, J., Aguilar-Gurrieri, C., Ortega, E. & Panne, D. Structure of the p300 catalytic core and implications for chromatin targeting and HAT regulation. Nat. Struct. Mol. Biol. 20, 1040–1046 (2013).

    Article  CAS  PubMed  Google Scholar 

  30. Ogryzko, V.V., Schiltz, R.L., Russanova, V., Howard, B.H. & Nakatani, Y. The transcriptional coactivators p300 and CBP are histone acetyltransferases. Cell 87, 953–959 (1996).

    Article  CAS  PubMed  Google Scholar 

  31. Chen, J. & Li, Q. Life and death of transcriptional co-activator p300. Epigenetics 6, 957–961 (2011).

    Article  CAS  PubMed  Google Scholar 

  32. 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).

    Article  PubMed  PubMed Central  Google Scholar 

  33. Gao, X. et al. Reprogramming to pluripotency using designer TALE transcription factors targeting enhancers. Stem Cell Reports 1, 183–197 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Ji, Q. et al. Engineered zinc-finger transcription factors activate OCT4 (POU5F1), SOX2, KLF4, c-MYC (MYC) and miR302/367. Nucleic Acids Res. 42, 6158–6167 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Rada-Iglesias, A. et al. A unique chromatin signature uncovers early developmental enhancers in humans. Nature 470, 279–283 (2011).

    Article  CAS  PubMed  Google Scholar 

  37. Visel, A. et al. ChIP-seq accurately predicts tissue-specific activity of enhancers. Nature 457, 854–858 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Chen, J.C., Love, C.M. & Goldhamer, D.J. Two upstream enhancers collaborate to regulate the spatial patterning and timing of MyoD transcription during mouse development. Dev. Dyn. 221, 274–288 (2001).

    Article  CAS  PubMed  Google Scholar 

  39. Nordhoff, V. et al. Comparative analysis of human, bovine, and murine Oct-4 upstream promoter sequences. Mamm. Genome 12, 309–317 (2001).

    Article  CAS  PubMed  Google Scholar 

  40. Carter, D., Chakalova, L., Osborne, C.S., Dai, Y.F. & Fraser, P. Long-range chromatin regulatory interactions in vivo. Nat. Genet. 32, 623–626 (2002).

    Article  CAS  PubMed  Google Scholar 

  41. Kim, Y.W. & Kim, A. Histone acetylation contributes to chromatin looping between the locus control region and globin gene by influencing hypersensitive site formation. Biochim. Biophys. Acta 1829, 963–969 (2013).

    Article  CAS  PubMed  Google Scholar 

  42. 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).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Esvelt, K.M. et al. Orthogonal Cas9 proteins for RNA-guided gene regulation and editing. Nat. Methods 10, 1116–1121 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Su, M.Y. et al. Identification of biologically relevant enhancers in human erythroid cells. J. Biol. Chem. 288, 8433–8444 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Doench, J.G. et al. Rational design of highly active sgRNAs for CRISPR-Cas9-mediated gene inactivation. Nat. Biotechnol. 32, 1262–1267 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Polstein, L.R. & Gersbach, C.A. A light-inducible CRISPR-Cas9 system for control of endogenous gene activation. Nat. Chem. Biol. 11, 198–200 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Zetsche, B., Volz, S.E. & Zhang, F. A split-Cas9 architecture for inducible genome editing and transcription modulation. Nat. Biotechnol. 33, 139–142 (2015).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Gibson, D.G. et al. Enzymatic assembly of DNA molecules up to several hundred kilobases. Nat. Methods 6, 343–345 (2009).

    Article  CAS  PubMed  Google Scholar 

  52. Chen, L.F., Mu, Y. & Greene, W.C. Acetylation of RelA at discrete sites regulates distinct nuclear functions of NF-kappaB. EMBO J. 21, 6539–6548 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Hu, J. et al. Direct activation of human and mouse Oct4 genes using engineered TALE and Cas9 transcription factors. Nucleic Acids Res. 42, 4375–4390 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Magnenat, L., Blancafort, P. & Barbas, C.F. III. In vivo selection of combinatorial libraries and designed affinity maturation of polydactyl zinc finger transcription factors for ICAM-1 provides new insights into gene regulation. J. Mol. Biol. 341, 635–649 (2004).

    Article  CAS  PubMed  Google Scholar 

  55. Hotta, A. et al. Isolation of human iPS cells using EOS lentiviral vectors to select for pluripotency. Nat. Methods 6, 370–376 (2009).

    Article  CAS  PubMed  Google Scholar 

  56. Schmittgen, T.D. & Livak, K.J. Analyzing real-time PCR data by the comparative C(T) method. Nat. Protoc. 3, 1101–1108 (2008).

    Article  CAS  PubMed  Google Scholar 

  57. 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 

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

    Article  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

P. Perez-Pinera, D. Kocak, D. Ousterout and D. Lim provided assistance with gRNA design, plasmid cloning, PCR primer validation, and/or RNA isolations. The gene encoding the ICAM1-targeted zinc finger protein was provided by C. Barbas, III. This work was supported by a US National Institutes of Health (NIH) grants to G.E.C., C.A.G. and T.E.R. (R01DA036865 and U01HG007900), a NIH Director's New Innovator Award (DP2OD008586) and National Science Foundation (NSF) Faculty Early Career Development (CAREER) Award (CBET-1151035) to C.A.G. and NIH grant P30AR066527.

Author information

Authors and Affiliations

Authors

Contributions

I.B.H., A.M.D., C.M.V., P.I.T., G.E.C., T.E.R. and C.A.G. designed experiments. I.B.H., A.M.D. and C.M.V. performed the experiments. I.B.H., A.M.D., C.M.V., P.I.T., G.E.C., T.E.R. and C.A.G. analyzed the data. I.B.H. and C.A.G. wrote the manuscript with contributions by all authors.

Corresponding authors

Correspondence to Timothy E Reddy or Charles A Gersbach.

Ethics declarations

Competing interests

C.A.G., I.B.H. and P.I.T. have filed patent applications related to genome engineering technologies. 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 dCas9p300 Core mutant fusion protein activities.

(a) Schematic depiction of the WT dCas9p300 Core fusion protein and p300 Core mutant derivatives. Relative locations of mutated amino acids are displayed as yellow bars within the p300 Core effector domain. (b) dCas9p300 Core variants were transiently co-transfected with four IL1RN promoter gRNAs and were screened for hyperactivity1 (amino acid 1645/1646 RR/EE and C1204R mutations) or hypoactivity1, 2 (denoted by ‡) via mRNA production from the IL1RN locus (top panel, n=2 independent experiments, error bars: s.e.m.). Experiments were performed in duplicate with one well used for RNA isolation and the other for western blotting to validate expression (bottom panels). The nitrocellulose membrane was cut and incubated with α-FLAG primary antibody (top, Sigma-Aldrich cat.# F7425) or α-GAPDH (bottom, Cell Signaling Technology cat.# 14C10) then α-Rabbit HRP secondary antibody (Sigma-Aldrich cat.# A6154). (c) Full membranes from western blot shown in main text (Figure 1b). The nitrocellulose membrane was cut and incubated with α-FLAG primary antibody (top, Sigma-Aldrich cat.# F7425) or α-GAPDH (bottom, Cell Signaling Technology cat.# 14C10) then α-Rabbit HRP secondary antibody (Sigma-Aldrich cat.# A6154). Membrane was imaged for the indicated durations after careful re-alignment of trimmed pieces.

Supplementary Figure 2 Target gene activation is unaffected by overexpression of synthetic dCas9 fusion proteins.

dCas9 fusion proteins were transiently co-transfected with an empty gRNA vector backbone and mRNA expression of IL1RN, MYOD, and OCT4 was assayed as in the main text. Red dashed line indicates background expression level from No DNA-transfected cells. n=2 independent experiments, error bars: s.e.m., no significant activation was observed for any target gene assayed.

Supplementary Figure 3 Comparison of Sp. dCas9 and Nm. dCas9 gene induction from the HS2 enhancer with individual and pooled gRNAs.

(a) Schematic display of the human β-globin locus including Streptococcus pyogenes dCas9 (Sp. dCas9) and Neisseria meningitidis dCas9 (Nm. dCas9) gRNA locations at the HS2 enhancer. Layered transcription profiles scaled to a vertical viewing range of 8 from nine ENCODE cell lines (GM12878, H1-hESC, HeLa-S3, HepG2, HSMM, HUVEC, K562, NHEK, and NHLF) is shown in addition to ENCODE p300 binding peaks in K562, A549 (EtOH.02), HeLA-S3, and SKN_SH_RA cell lines. An ENCODE HEK293T DNase hypersensitive site (HEK293T DHS) is shown in the HS2 Enhancer inset. (b–e) Relative transcriptional induction of HBE, HBG, HBD, and HBD transcripts from single and pooled Sp. dCas9 gRNAs (A–D) or single and pooled Nm. dCas9 gRNAs (A–E) in response to co-transfection with Sp. dCas9p300 Core or Nm. dCas9p300 Core respectively. gRNAs are tiled for each dCas9 ortholog corresponding to their location in GRCh37/hg19. Gray dashed line indicates background expression level in transiently co-transfected HEK293T cells. Note shared logarithmic scale among panels b–e. Numbers above bars in b–e indicate mean expression (n = at least 3 independent experiments, error bars: s.e.m.).

Supplementary Figure 4 dCas9VP64 and dCas9p300 Core induce H3K27ac enrichment at IL1RN gRNA-targeted chromatin.

The IL1RN locus on GRCh37/hg19 is shown along with IL1RN gRNA target sites. In addition layered ENCODE H3K27ac enrichment from seven cell lines (GM12878, H1-hESC, HSMM, HUVEC, K562, NHEK, and NHLF) is indicated with the vertical range setting set to 50. Tiled IL1RN ChIP qPCR amplicons (1–13) are also shown in corresponding locations on GRCh37/hg19. H3K27ac enrichment for dCas9VP64 and dCas9p300 Core co-transfected with four IL1RN-targeted gRNAs and normalized to dCas9 co-transfected with four IL1RN gRNAs is indicated for each ChIP qPCR locus assayed. 5ng of ChIP-prepared DNA was used for each reacton (n = 3 independent experiments, error bars: s.e.m.)

Supplementary Figure 5 Direct comparison of VP64 and p300 Core effector domains between TALE and dCas9 programmable DNA binding proteins.

(a) The GRCh37/hg19 region encompassing the IL1RN transcription start site is shown schematically along with IL1RN TALE binding sites and dCas9 IL1RN gRNA target sites. (b) Direct comparison of IL1RN activation in HEK293T cells when transfected with individual or pooled (A–D) IL1RN TALEVP64 fusion proteins or when co-transfected with dCas9VP64 and individual or pooled (A–D) IL1RN-targeting gRNAs. (c) Direct comparison of IL1RN activation in HEK293T cells when transfected with individual or pooled (A–D) IL1RN TALEp300 Core fusion proteins or when co-transfected with dCas9p300 Core and individual or pooled (A–D) IL1RN-targeting gRNAs. Note shared logarithmic scale between panels b and c. Numbers above bars in panels b and c indicate mean values. Tukey test, *P-value <0.05, n = at least 3 independent experiments, error bars: s.e.m.

Supplementary Figure 6 TALE and ZF fusion protein expression.

(a) Western blotting was carried out on cells transiently transfected with individual or pooled IL1RN TALE proteins. Nitrocellulose membranes were cut and probed with α-HA primary antibody (1:1000 dilution in TBST + 5% Milk, top, Covance cat.# MMS-101P) or α-GAPDH (bottom, Cell Signaling Technology cat.# 14C10) then α-Mouse HRP (Santa Cruz, sc-2005) or α-Rabbit HRP (Sigma-Aldrich cat.# A6154) secondary antibody, respectively. (b) Western blotting was carried out on cells transiently transfected with ICAM1 ZF-effector proteins and nitrocellulose membranes were cut and probed with α-FLAG primary antibody (top, Sigma-Aldrich cat.# F7425) or α-GAPDH (bottom, Cell Signaling Technology cat.# 14C10) then α-Rabbit HRP secondary antibody (Sigma-Aldrich cat.# A6154). Red asterisk indicates non-specific band.

Supplementary Figure 7 dCas9p300 Core and dCas9VP64 do not display synergy in transactivation.

(a) dCas9p300 Core was co-transfected at a 1:1 mass ratio to PL-SIN-EF1α-EGFP3 (GFP), dCas9, or dCas9VP64 with four IL1RN promoter gRNAs as indicated (n = 2 independent experiments, error bars: s.e.m.). (b) dCas9p300 Core was co-transfected at a 1:1 mass ratio to GFP, dCas9, or dCas9VP64 with four MYOD promoter gRNAs as indicated (n = 2 independent experiments, error bars: s.e.m.). No significant differences were observed using Tukey’s test (n.s).

Supplementary Figure 8 Underlying chromatin context of dCas9p300 Core target loci.

(a–d) Indicated loci are shown along with associated Streptococcus pyogenes gRNAs used in this study at corresponding genomic locations in GRCh37/hg19. ENCODE HEK293T DNase hypersensitivity enrichment is shown (note changes in scale) along with regions of significant DNase hypersensitivity in HEK293T cells (“DHS”). In addition ENCODE master DNase clusters across 125 cell types are shown. Layered ENCODE H3K27ac and H3K4me3 enrichment across seven cell lines (GM12878, H1-hESC, HSMM, HUVEC, K562, NHEK, and NHLF) is also displayed and scaled to a vertical viewing range of 50 and 150 respectively. Endogenous p300 binding profiles are also indicated for each locus and respective cell line. (e) An overview of the information provided in a–d.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–8, Supplementary Tables 1–5 and Supplementary Notes 1 and 2 (PDF 1261 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Hilton, I., D'Ippolito, A., Vockley, C. et al. Epigenome editing by a CRISPR-Cas9-based acetyltransferase activates genes from promoters and enhancers. Nat Biotechnol 33, 510–517 (2015). https://doi.org/10.1038/nbt.3199

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nbt.3199

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