DOI QR코드

DOI QR Code

ChIP-seq Analysis of Histone H3K27ac and H3K27me3 Showing Different Distribution Patterns in Chromatin

  • Kang, Jin (Department of Molecular Biology, College of Natural Sciences, Pusan National University) ;
  • Kim, AeRi (Department of Molecular Biology, College of Natural Sciences, Pusan National University)
  • 투고 : 2022.06.03
  • 심사 : 2022.06.22
  • 발행 : 2022.06.30

초록

Histone proteins can be modified by the addition of acetyl group or methyl group to specific amino acids. The modifications have different distribution patterns in chromatin. Recently, histone modifications are studied based on ChIP-seq data, which requires reasonable analysis of sequencing data depending on their distribution patterns. Here we have analyzed histone H3K27ac and H3K27me3 ChIP-seq data and it showed that the H3K27ac is enriched at narrow regions while H3K27me3 distributes broadly. To properly analyze the ChIP-seq data, we called peaks for H3K27ac and H3K27me3 using MACS2 (narrow option and broad option) and SICER methods, and compared propriety of the peaks using signal-to-background ratio. As results, H3K27ac-enriched regions were well identified by both methods while H3K27me3 peaks were properly identified by SICER, which indicates that peak calling method is more critical for histone modifications distributed broadly. When ChIP-seq data were compared in different sequencing depth (15, 30, 60, 120 M), high sequencing depth caused high false-positive rate in H3K27ac peak calling, but it reflected more properly the broad distribution pattern of H3K27me3. These results suggest that sequencing depth affects peak calling from ChIP-seq data and high sequencing depth is required for H3K27me3. Taken together, peak calling tool and sequencing depth should be chosen depending on the distribution pattern of histone modification in ChIP-seq analysis.

키워드

과제정보

This study was supported by the 『BK21 Four Program』 of Pusan National University.

참고문헌

  1. Bannister AJ, Kouzarides T. Regulation of chromatin by histone modifications. Cell Res. 2011. 21: 381-395. https://doi.org/10.1038/cr.2011.22
  2. Bolger AM, Lohse M, Usadel B. Trimmomatic: a flexible trimmer for Illumina sequence data. Bioinformatics. 2014. 30: 2114-2120. https://doi.org/10.1093/bioinformatics/btu170
  3. Bowman SK, Deaton AM, Domingues H, et al. H3K27 modifications define segmental regulatory domains in the Drosophila bithorax complex. eLife. 2014. 3: e02833. https://doi.org/10.7554/elife.02833
  4. Cao R, Wang L, Wang H, et al. Role of histone H3 lysine 27 methylation in Polycomb-group silencing. Science. 2002. 298: 1039-1043. https://doi.org/10.1126/science.1076997
  5. Chi P, Allis CD, Wang GG. Covalent histone modifications-miswritten, misinterpreted and mis-erased in human cancers. Nat Rev Cancer. 2010. 10: 457-469. https://doi.org/10.1038/nrc2876
  6. Chrysanthou S, Tang Q, Lee J, et al. The DNA dioxygenase Tet1 regulates H3K27 modification and embryonic stem cell biology independent of its catalytic activity. Nucleic Acids Res. 2022. 50: 3169-3189. https://doi.org/10.1093/nar/gkac089
  7. Cutter AR, Hayes JJ. A brief review of nucleosome structure. FEBS Lett. 2015. 589: 2914-2922. https://doi.org/10.1016/j.febslet.2015.05.016
  8. Fernandes MT, Almeida-Lousada H, Castelo-Branco P. Histone modifications in diseases. 2020. Vol 20, pp. 1-15. Academic Press. USA.
  9. Fiskus W, Wang Y, Sreekumar A, et al. Combined epigenetic therapy with the histone methyltransferase EZH2 inhibitor 3-deazaneplanocin A and the histone deacetylase inhibitor panobinostat against human AML cells. Blood. 2009. 114: 2733-2743.
  10. Fujii S, Ochiai A. Enhancer of zeste homolog 2 downregulates E-cadherin by mediating histone H3 methylation in gastric cancer cells. Cancer Sci. 2008. 99: 738-746. https://doi.org/10.1111/j.1349-7006.2008.00743.x
  11. Jeon H, Lee H, Kang B, Jang I, Roh TY. Comparative analysis of commonly used peak calling programs for ChIP-Seq analysis. Genomics Inform. 2020. 18: e42. https://doi.org/10.5808/GI.2020.18.4.e42
  12. Kang J, Kim YW, Park S, Kang Y, Kim A. Multiple CTCF sites cooperate with each other to maintain a TAD for enhancer-promoter interaction in the β-globin locus. FASEB J. 2021a. 35: e21768.
  13. Kang Y, Kang J, Kim A. Histone H3K4me1 strongly activates the DNase I hypersensitive sites in super-enhancers than those in typical enhancers. Biosci Rep. 2021b. 41: BSR20210691. https://doi.org/10.1042/BSR20210691
  14. Kang Y, Kim YW, Kang J, Kim A. Histone H3K4me1 and H3K27ac play roles in nucleosome eviction and eRNA transcription, respectively, at enhancers. FASEB J. 2021c. 35: e21781.
  15. Katoh N, Kuroda K, Tomikawa J, et al. Reciprocal changes of H3K27ac and H3K27me3 at the promoter regions of the critical genes for endometrial decidualization. Epigenomics. 2018. 10: 1243-1257. https://doi.org/10.2217/epi-2018-0006
  16. Khan A, Mathelier A. Intervene: a tool for intersection and visualization of multiple gene or genomic region sets. BMC Bioinformatics. 2017. 18: 287. https://doi.org/10.1186/s12859-017-1708-7
  17. Kim J, Kang J, Kim YW, Kim A. The human β-globin enhancer LCR HS2 plays a role in forming a TAD by activating chromatin structure at neighboring CTCF sites. FASEB J. 2021. 35: e21669.
  18. Kim YW, Kang Y, Kang J, Kim A. GATA-1-dependent histone H3K27 acetylation mediates erythroid cell-specific chromatin interaction between CTCF sites. FASEB J. 2020. 34: 14736-14749. https://doi.org/10.1096/fj.202001526R
  19. Kim YW, Kim A. Characterization of histone H3K27 modifications in the β-globin locus. Biochem Biophys Res Commun. 2011. 405: 210-215. https://doi.org/10.1016/j.bbrc.2011.01.010
  20. Kuo MH, Allis CD. In vivo cross-linking and immunoprecipitation for studying dynamic Protein:DNA associations in a chromatin environment. Methods. 1999. 19: 425-433. https://doi.org/10.1006/meth.1999.0879
  21. Landt SG, Marinov GK, Kundaje A, et al. ChIP-seq guidelines and practices of the ENCODE and modENCODE consortia. Genome Res. 2012. 22: 1813-1831. https://doi.org/10.1101/gr.136184.111
  22. Langmead B, Salzberg SL. Fast gapped-read alignment with Bowtie 2. Nat Methods. 2012. 9: 357-359. https://doi.org/10.1038/nmeth.1923
  23. Lauberth SM, Nakayama T, Wu X, et al. H3K4me3 interactions with TAF3 regulate preinitiation complex assembly and selective gene activation. Cell. 2013. 152: 1021-1036. https://doi.org/10.1016/j.cell.2013.01.052
  24. Lee HA, Cho HM, Lee DY, Kim KC, Han HS, Kim IK. Tissue-specific upregulation of angiotensin-converting enzyme 1 in spontaneously hypertensive rats through histone code modifications. Hypertension. 2012. 59: 621-626. https://doi.org/10.1161/HYPERTENSIONAHA.111.182428
  25. Marzi SJ, Leung SK, Ribarska T, et al. A histone acetylome-wide association study of Alzheimer's disease identifies disease-associated H3K27ac differences in the entorhinal cortex. Nat Neurosci. 2018. 21: 1618-1627. https://doi.org/10.1038/s41593-018-0253-7
  26. McErlean P, Kelly A, Dhariwal J, et al. Profiling of H3K27Ac reveals the influence of asthma on the epigenome of the airway epithelium. Front Genet. 2020. 11: 585746. https://doi.org/10.3389/fgene.2020.585746
  27. Min J, Zhang Y, Xu RM. Structural basis for specific binding of Polycomb chromodomain to histone H3 methylated at Lys 27. Genes Dev. 2003. 17: 1823-1828. https://doi.org/10.1101/gad.269603
  28. Ougolkov AV, Bilim VN, Billadeau DD. Regulation of pancreatic tumor cell proliferation and chemoresistance by the histone methyltransferase enhancer of zeste homologue 2. Clin Cancer Res. 2008. 14: 6790-6796. https://doi.org/10.1158/1078-0432.ccr-08-1013
  29. Ramirez F, Ryan DP, Gruning B, et al. deepTools2: a next generation web server for deep-sequencing data analysis. Nucleic Acids Res. 2016. 44: W160-W165. https://doi.org/10.1093/nar/gkw257
  30. Thorvaldsdottir H, Robinson JT, Mesirov JP. Integrative Genomics Viewer (IGV): high-performance genomics data visualization and exploration. Brief Bioinform. 2013. 14: 178-192. https://doi.org/10.1093/bib/bbs017
  31. Vermeulen M, Mulder KW, Denissov S, et al. Selective anchoring of TFIID to nucleosomes by trimethylation of histone H3 lysine 4. Cell. 2007. 131: 58-69. https://doi.org/10.1016/j.cell.2007.08.016
  32. Wang Y, Hou C, Wisler J, et al. Elevated histone H3 acetylation is associated with genes involved in T lymphocyte activation and glutamate decarboxylase antibody production in patients with type 1 diabetes. J Diabetes Investig. 2019. 10: 51-61. https://doi.org/10.1111/jdi.12867
  33. Wang Z, Zang C, Rosenfeld JA, et al. Combinatorial patterns of histone acetylations and methylations in the human genome. Nat Genet. 2008. 40: 897-903. https://doi.org/10.1038/ng.154
  34. Wolffe AP, Guschin D. Review: chromatin structural features and targets that regulate transcription. J Struct Biol. 2000. 129: 102-122. https://doi.org/10.1006/jsbi.2000.4217
  35. Yu J, Cao Q, Mehra R, et al. Integrative genomics analysis reveals silencing of β-adrenergic signaling by polycomb in prostate cancer. Cancer Cell. 2007. 12: 419-431. https://doi.org/10.1016/j.ccr.2007.10.016
  36. Zang C, Schones DE, Zeng C, Cui K, Zhao K, Peng W. A clustering approach for identification of enriched domains from histone modification ChIP-Seq data. Bioinformatics. 2009. 25: 1952-1958. https://doi.org/10.1093/bioinformatics/btp340
  37. Zhang L, Xiong D, Liu Q, et al. Genome-wide histone H3K27 acetylation profiling identified genes correlated with prognosis in papillary thyroid carcinoma. Front Cell Dev Biol. 2021. 9: 682561. https://doi.org/10.3389/fcell.2021.682561
  38. Zhang Y, Liu T, Meyer CA, et al. Model-based analysis of ChIPSeq (MACS). Genome Biol. 2008. 9: R137. https://doi.org/10.1186/gb-2008-9-9-r137