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:

FiTAc-seq: fixed-tissue ChIP-seq for H3K27ac profiling and super-enhancer analysis of FFPE tissues

Abstract

Fixed-tissue ChIP-seq for H3K27 acetylation (H3K27ac) profiling (FiTAc-seq) is an epigenetic method for profiling active enhancers and promoters in formalin-fixed, paraffin-embedded (FFPE) tissues. We previously developed a modified ChIP-seq protocol (FiT-seq) for chromatin profiling in FFPE. FiT-seq produces high-quality chromatin profiles particularly for methylated histone marks but is not optimized for H3K27ac profiling. FiTAc-seq is a modified protocol that replaces the proteinase K digestion applied in FiT-seq with extended heating at 65 °C in a higher concentration of detergent and a minimized sonication step, to produce robust genome-wide H3K27ac maps from clinical samples. FiTAc-seq generates high-quality enhancer landscapes and super-enhancer (SE) annotation in numerous archived FFPE samples from distinct tumor types. This approach will be of great interest for both basic and clinical researchers. The entire protocol from FFPE blocks to sequence-ready library can be accomplished within 4 d.

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: Schematic overview of the FiTAc-seq H3K27ac profiling protocol in FFPE samples.
Fig. 2: Optimization of the FiTAc-seq protocol and application for enhancer and SE annotation.
Fig. 3: FiTAc-seq performance on clinical FFPE specimens in multiple tumor types.

Similar content being viewed by others

Data availability

Figures 2 and 3, and Extended Data Figs. 1, 3 and 4 all have associated raw data. The ChIP-seq data have been uploaded to the Gene Expression Omnibus (GSE140808). In Extended Data Fig. 3, we compare our data to previously published data available on the Gene Expression Omnibus (GSE128202).

References

  1. Cejas, P. & Long, H. W. Principles and methods of integrative chromatin analysis in primary tissues and tumors. Biochem. Biophys. Acta Rev. Cancer 1873, 188333 (2020).

    Article  Google Scholar 

  2. Lin, C. Y. et al. Active medulloblastoma enhancers reveal subgroup-specific cellular origins. Nature 530, 57–62 (2016).

    Article  CAS  Google Scholar 

  3. Consortium, T. E. P., The ENCODE Project Consortium. The ENCODE (ENCyclopedia Of DNA Elements) Project. Science 306, 636–640 (2004).

    Article  Google Scholar 

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

    Article  CAS  Google Scholar 

  5. Cejas, P. et al. Chromatin immunoprecipitation from fixed clinical tissues reveals tumor-specific enhancer profiles. Nat. Med 22, 685–691 (2016).

    Article  Google Scholar 

  6. Creyghton, M. P. et al. Histone H3K27ac separates active from poised enhancers and predicts developmental state. Proc. Natl Acad. Sci. USA 107, 21931–21936 (2010).

    Article  CAS  Google Scholar 

  7. Calo, E. & Wysocka, J. Modification of enhancer chromatin: what, how, and why? Mol. Cell 49, 825–837 (2013).

    Article  CAS  Google Scholar 

  8. Zentner, G. E. & Scacheri, P. C. The chromatin fingerprint of gene enhancer elements. J. Biol. Chem. 287, 30888–30896 (2012).

    Article  CAS  Google Scholar 

  9. 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  Google Scholar 

  10. Hnisz, D. et al. Super-enhancers in the control of cell identity and disease. Cell 155, 934–947 (2013).

    Article  CAS  Google Scholar 

  11. Lovén, J. et al. Selective inhibition of tumor oncogenes by disruption of super-enhancers. Cell 153, 320–334 (2013).

    Article  Google Scholar 

  12. Chipumuro, E. et al. CDK7 inhibition suppresses super-enhancer-linked oncogenic transcription in MYCN-driven cancer. Cell 159, 1126–1139 (2014).

    Article  CAS  Google Scholar 

  13. Mack, S. C. et al. Therapeutic targeting of ependymoma as informed by oncogenic enhancer profiling. Nature 553, 101–105 (2018).

    Article  CAS  Google Scholar 

  14. Cejas, P. et al. Enhancer signatures stratify and predict outcomes of non-functional pancreatic neuroendocrine tumors. Nat. Med 25, 1260–1265 (2019).

    Article  CAS  Google Scholar 

  15. Zhong, J. et al. Enhanced and controlled chromatin extraction from FFPE tissues and the application to ChIP-seq. BMC Genomics 20, 249 (2019).

    Article  Google Scholar 

  16. Amatori, S. et al. Epigenomic profiling of archived FFPE tissues by enhanced PAT-ChIP (EPAT-ChIP) technology. Clin. Epigenetics 10, 143 (2018).

    Article  CAS  Google Scholar 

  17. Shi, S. R., Imam, S. A., Young, L., Cote, R. J. & Taylor, C. R. Antigen retrieval immunohistochemistry under the influence of pH using monoclonal antibodies. J. Histochem. Cytochem. 43, 193–201 (1995).

    Article  CAS  Google Scholar 

  18. Qin, Q. et al. ChiLin: a comprehensive ChIP-seq and DNase-seq quality control and analysis pipeline. BMC Bioinforma. 17, 404 (2016).

    Article  Google Scholar 

  19. Wang, S. et al. Modeling cis-regulation with a compendium of genome-wide histone H3K27ac profiles. Genome Res. 26, 1417–1429 (2016).

    Article  CAS  Google Scholar 

  20. 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, 232 (2013).

    Article  CAS  Google Scholar 

  21. He, H. H. et al. Nucleosome dynamics define transcriptional enhancers. Nat. Genet. 42, 343–347 (2010).

    Article  CAS  Google Scholar 

  22. Corces, M. R. et al. An improved ATAC-seq protocol reduces background and enables interrogation of frozen tissues. Nat. Methods 14, 959–962 (2017).

    Article  CAS  Google Scholar 

  23. Hunt, J. L. & Finkelstein, S. D. Microdissection techniques for molecular testing in surgical pathology. Arch. Pathol. Lab. Med. 128, 1372–1378 (2004).

    PubMed  Google Scholar 

  24. Mägel, L., Bartels, S. & Lehmann, U. Next- generation sequencing analysis of laser-microdissected formalin-fixed and paraffin-embedded (FFPE) tissue specimens. Methods Mol. Biol. 1723, 111–118 (2018).

    Article  Google Scholar 

  25. Mes, S. W. et al. Development and validation of a novel and rapid molecular detection method for high-risk human papillomavirus in formalin-fixed, paraffin-embedded tumor tissue. J. Mol. Diagn. 22, 262–271 (2020).

    Article  CAS  Google Scholar 

  26. Kuilman, T. et al. CopywriteR: DNA copy number detection from off-target sequence data. Genome Biol. 16, 49 (2015).

    Article  Google Scholar 

  27. Li, H. & Durbin, R. Fast and accurate short read alignment with Burrows–Wheeler transform. Bioinformatics 25, 1754–1760 (2009).

    Article  CAS  Google Scholar 

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

    Article  Google Scholar 

  29. Thorvaldsdóttir, H., Robinson, J. T. & Mesirov, J. P. Integrative Genomics Viewer (IGV): high-performance genomics data visualization and exploration. Brief. Bioinform. 14, 178–192 (2013).

    Article  Google Scholar 

  30. Ramírez, F. et al. deepTools2: a next generation web server for deep-sequencing data analysis. Nucleic Acids Res 44, W160–W165 (2016).

    Article  Google Scholar 

  31. Kinkley, S. et al. reChIP-seq reveals widespread bivalency of H3K4me3 and H3K27me3 in CD4+ memory T cells. Nat. Commun. 7, 12514 (2016).

    Article  CAS  Google Scholar 

  32. Shin, H., Liu, T., Duan, X., Zhang, Y. & Shirley Liu, X. Computational methodology for ChIP-seq analysis. Quant. Biol. 1, 54–70 (2013).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

A.F.-T. has been supported by Associació per a la recerca oncològica (APRO) for the present study. P.C. acknowledges funding from the Ministry of Economy and Competitiveness, Instituto de Salud Carlos III (Institute of Health Carlos III)—PI18-01604. E.M.V.A acknowledges funding from US NIH grant R37CA222574. M.B. acknowledges funding from NIH grant 2PO1CA163227. We thank R. Vadhi, M. A. Berkeley and Z. Herbert from the Molecular Biology Core Facility (MBCF) at the Dana-Farber Cancer Institute for preparing and sequencing Illumina libraries. We acknowledge X. Qiu, K. Lim and S. Syamala for valuable technical support.

Author information

Authors and Affiliations

Authors

Contributions

A.F.-T., P.C. and H.W.L. designed the study. A.F.-T. developed the protocol and performed the experiments. N.K., Y.X., L.T. and A.F.-T. performed the computational and statistical analyses. A.L., F.S.H., D.V., L.S.Y., A.A.H., E.M.V.A., C.J.S., E.G. and J.B. supplied the clinical tissue and histological analysis. A.F.-T., P.C. and H.W.L. wrote the paper. A.F.-T., M.B., P.C. and H.W.L. supervised the study and revised the paper.

Corresponding authors

Correspondence to Paloma Cejas or Henry W. Long.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

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

Cejas, P. et al. Nat. Med. 22, 685–691 (2016): https://doi.org/10.1038/nm.4085

Cejas, P. et al. Nat. Med. 25, 1260–1265 (2019): https://doi.org/10.1038/s41591-019-0493-4

Extended data

Extended Data Fig. 1 FiT-seq performance on FFPE mouse liver tissue.

(a) Bar plot of total and 10-fold enriched peaks called from H3K4Me2 and H3K27Ac data generated by the optimized original FiT-seq protocol. (b) Average H3K4Me2 and H3K27Ac signal distribution for normal mouse liver tissue obtained by the optimized original FiT-Seq protocol centered at H3K4Me2 called peaks. (c) IGV traces at GAPDH locus. Y axis indicates the number of reads per million per base pair (rbm). Appropriate institutional regulatory board permission was obtained for animal experiments.

Extended Data Fig. 2 FiTAc-seq performance on clinical FFPE specimens.

Unsupervised hierarchical clustering of the sample-sample correlation of the H3K27Ac signal at the union of all enhancers annotated for the five tumor types (total enhancers = 117397): bladder cancers (BlCa), breast cancer brain metastasis (BrCaBM), melanomas (MEL), pancreatic neuroendocrine tumors (PNETs) and seminomas (SEM). Each row and column represents a different sample. Scale represents Pearson’s correlation.

Extended Data Fig. 3 Comparison of FiTAc-seq and ChromEX-PE performance for H3K27Ac on FFPE mouse liver tissue.

ChromEX-PE15 publicly available data was analyzed at the same sequencing depth and with the exact same analysis parameters as the FiTAc-seq data. (a) Average distribution of H3K27Ac signal at enhancers (TSS excluded) in normal mouse liver FFPE tissue obtained from FiTAc-seq and ChromEX-PE protocols in duplicate as indicated in the legend. The union of enhancer peaks from all the replicates was used to center the H3K27Ac signal to perform the aggregation plot. (b) Integrative genomics viewer (IGV) tracks at the Hnf4a locus comparing replicates of the FiTAc-seq (tracks 2 and 3) and ChromEX-PE (tracks 5 and 6) datasets. FF data from ChromEX-PE publication (track 4) and from our mouse model system (track 1) are used as reference. Y axis represents the number of reads per million per base pair (rbm). Appropriate institutional regulatory board permission was obtained for animal experiments.

Extended Data Fig. 4 Representative traces of chromatin fragmentation.

(a) Fragment size distribution achieved with 5 min sonication (tissue homogenization at 65 °C, two replicates) and with 40 min sonication (tissue homogenization at 50 °C, 65 °C or 80 °C) for mouse liver tissue sample. (b) Fragment size distribution for several clinical FFPE archived samples.

Extended Data Fig. 5 Peak calling performance on FiTAc-seq signal.

(a) Bar plot of total, 10-fold and 20-fold enriched peaks for two replicates of mouse FFPE normal liver tissue (FiTAc-seq 65 °C_5min) called with and without input. (b) Corresponding IGV tracks at representative Hnf4a locus with annotation of peaks (black boxes below signal). Y axis indicates the number of reads per million per base pair (rbm). Appropriate institutional regulatory board permission was obtained for animal experiments.

Extended Data Fig. 6 H3K27Ac signal conservation at peaks.

(a) Plots representing evolutionary conservation across mammalian genomes for FF and FFPE 65 °C_5min and 65 °C_40min mouse liver samples. Appropriate institutional regulatory board permission was obtained for animal experiments. (b) Plots representing evolutionary conservation across mammalian genomes for all FiTAc-seq FFPE clinical samples. Y axis represents PhastCons score, X axis represents distance from center in kb.

Supplementary information

Reporting Summary

Supplementary Table 1

Sequencing and peak calling statistics results from normal mouse liver FFPE FiTAc-seq and FF counterpart.

Supplementary Table 2

List of SE annotated by ROSE algorithm for H3K27Ac signal from FF and FiTAc-seq 65°C_5min FFPE mouse liver samples.

Supplementary Table 3

RP scores from marge-potential analysis on H3K27Ac signal for FF and FiTAc-seq 65°C_5min FFPE mouse liver samples.

Supplementary Table 4

Sequencing and peak calling statistics of FiTAc-seq in clinical tissue samples, including (a) mean values in each tissue-type and (b) specific results for each of the samples.

Supplementary Table 5

List of SE identified by ROSE algorithm for FiTAc-seq FFPE clinical tumor samples, including bladder cancers (BlCa), breast cancer brain metastasis (BrCaBM), melanoma (MEL), pancreatic neuroendocrine tumors (PNETs) and seminomas (SEM).

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Font-Tello, A., Kesten, N., Xie, Y. et al. FiTAc-seq: fixed-tissue ChIP-seq for H3K27ac profiling and super-enhancer analysis of FFPE tissues. Nat Protoc 15, 2503–2518 (2020). https://doi.org/10.1038/s41596-020-0340-6

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41596-020-0340-6

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