Browse > Article
http://dx.doi.org/10.5483/BMBRep.2020.53.5.045

Analysis of opposing histone modifications H3K4me3 and H3K27me3 reveals candidate diagnostic biomarkers for TNBC and gene set prediction combination  

Park, Hyoung-Min (Department of Biochemistry, BK21 Plus and Research Institute for Veterinary Science, School of Veterinary Medicine, Seoul National University)
Kim, HuiSu (Department of Biochemistry, BK21 Plus and Research Institute for Veterinary Science, School of Veterinary Medicine, Seoul National University)
Lee, Kang-Hoon (Department of Biochemistry, BK21 Plus and Research Institute for Veterinary Science, School of Veterinary Medicine, Seoul National University)
Cho, Je-Yoel (Department of Biochemistry, BK21 Plus and Research Institute for Veterinary Science, School of Veterinary Medicine, Seoul National University)
Publication Information
BMB Reports / v.53, no.5, 2020 , pp. 266-271 More about this Journal
Abstract
Breast cancer encompasses a major portion of human cancers and must be carefully monitored for appropriate diagnoses and treatments. Among the many types of breast cancers, triple negative breast cancer (TNBC) has the worst prognosis and the least cases reported. To gain a better understanding and a more decisive precursor for TNBC, two major histone modifications, an activating modification H3K4me3 and a repressive modification H3K27me3, were analyzed using data from normal breast cell lines against TNBC cell lines. The combination of these two histone markers on the gene promoter regions showed a great correlation with gene expression. A list of signature genes was defined as active (highly enriched H3K4me3), including NOVA1, NAT8L, and MMP16, and repressive genes (highly enriched H3K27me3), IRX2 and ADRB2, according to the distribution of these histone modifications on the promoter regions. To further enhance the investigation, potential candidates were also compared with other types of breast cancer to identify signs specific to TNBC. RNA-seq data was implemented to confirm and verify gene regulation governed by the histone modifications. Combinations of the biomarkers based on H3K4me3 and H3K27me3 showed the diagnostic value AUC 93.28% with P-value of 1.16e-226. The results of this study suggest that histone modification analysis of opposing histone modifications may be valuable toward developing biomarkers and targets for TNBC.
Keywords
ChIP-seq; Histone modification; Human; RNA-seq; Triple negative breast cancer;
Citations & Related Records
연도 인용수 순위
  • Reference
1 Zand B, Previs RA, Zacharias NM et al (2016) Role of Increased n-acetylaspartate Levels in Cancer. J Natl Cancer Inst 108, djv426   DOI
2 Sun C, Ban Y, Wang K, Sun Y and Zhao Z (2019) SOX5 promotes breast cancer proliferation and invasion by transactivation of EZH2. Oncol Lett 17, 2754-2762
3 Wolf J, Muller-Decker K, Flechtenmacher C et al (2014) An in vivo RNAi screen identifies SALL1 as a tumor suppressor in human breast cancer with a role in CDH1 regulation. Oncogene 33, 4273-4278   DOI
4 Ma C, Wang F, Han B et al (2018) SALL1 functions as a tumor suppressor in breast cancer by regulating cancer cell senescence and metastasis through the NuRD complex. Mol Cancer 17, 78   DOI
5 Rahman M, Jackson LK, Johnson WE, Li DY, Bild AH and Piccolo SR (2015) Alternative preprocessing of RNASequencing data in The Cancer Genome Atlas leads to improved analysis results. Bioinformatics 31, 3666-3672   DOI
6 Jemal A, Bray F, Center MM, Ferlay J, Ward E and Forman D (2011) Global cancer statistics. CA Cancer J Clin 61, 69-90   DOI
7 O'Brien KM, Cole SR, Tse CK et al (2010) Intrinsic breast tumor subtypes, race, and long-term survival in the Carolina Breast Cancer Study. Clin Cancer Res 16, 6100-6110   DOI
8 Carey LA, Perou CM, Livasy CA et al (2006) Race, breast cancer subtypes, and survival in the Carolina Breast Cancer Study. JAMA 295, 2492-2502   DOI
9 Jones PA and Baylin SB (2007) The epigenomics of cancer. Cell 128, 683-692   DOI
10 Chen X, Hu H, He L et al (2016) A novel subtype classification and risk of breast cancer by histone modification profiling. Breast Cancer Research and Treatment 157, 267-279   DOI
11 Koch CM, Andrews RM, Flicek P et al (2007) The landscape of histone modifications across 1% of the human genome in five human cell lines. Genome Res 17, 691-707   DOI
12 Barski A, Cuddapah S, Cui K et al (2007) High-resolution profiling of histone methylations in the human genome. Cell 129, 823-837   DOI
13 Tang Z, Li C, Kang B, Gao G, Li C and Zhang Z (2017) GEPIA: a web server for cancer and normal gene expression profiling and interactive analyses. Nucleic Acids Res 45, W98-W102   DOI
14 Xi Y, Shi J, Li W et al (2018) Histone modification profiling in breast cancer cell lines highlights commonalities and differences among subtypes. BMC Genomics 19, 150   DOI
15 Gomez-Cabrero D, Abugessaisa I, Maier D et al (2014) Data integration in the era of omics: current and future challenges. BMC Syst Biol 8 Suppl 2, I1   DOI
16 Chaligne R, Popova T, Mendoza-Parra MA et al (2015) The inactive X chromosome is epigenetically unstable and transcriptionally labile in breast cancer. Genome Res 25, 488-503   DOI
17 Perou CM (2011) Molecular stratification of triple-negative breast cancers. Oncologist 16 Suppl 1, 61-70   DOI
18 Nebbioso A, Tambaro FP, Dell'Aversana C and Altucci L (2018) Cancer epigenetics: Moving forward. PLoS Genet 14, e1007362   DOI