Please wait a minute...
Frontiers in Biology

ISSN 1674-7984

ISSN 1674-7992(Online)

CN 11-5892/Q

Front. Biol.    2016, Vol. 11 Issue (4) : 311-322    https://doi.org/10.1007/s11515-016-1411-5
RESEARCH ARTICLE
Locus- and cell type-specific epigenetic switching during cellular differentiation in mammals
Ying-Tao Zhao,Maria Fasolino,Zhaolan Zhou()
Department of Genetics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
 Download: PDF(2323 KB)   HTML
 Export: BibTeX | EndNote | Reference Manager | ProCite | RefWorks
Abstract

BACKGROUND: Epigenomic reconfiguration, including changes in DNA methylation and histone modifications, is crucial for the differentiation of embryonic stem cells (ESCs) into somatic cells. However, the extent to which the epigenome is reconfigured and the interplay between components of the epigenome during cellular differentiation remain poorly defined.

METHODS: We systematically analyzed and compared DNA methylation, various histone modification, and transcriptome profiles in ESCs with those of two distinct types of somatic cells from human and mouse.

RESULTS: We found that global DNA methylation levels are lower in somatic cells compared to ESCs in both species. We also found that 80% of regions with histone modification occupancy differ between human ESCs and the two human somatic cell types. Approximately 70% of the reconfigurations in DNA methylation and histone modifications are locus- and cell type-specific. Intriguingly, the loss of DNA methylation is accompanied by the gain of different histone modifications in a locus- and cell type-specific manner. Further examination of transcriptional changes associated with epigenetic reconfiguration at promoter regions revealed an epigenetic switching for gene regulation—a transition from stable gene silencing mediated by DNA methylation in ESCs to flexible gene repression facilitated by repressive histone modifications in somatic cells.

CONCLUSIONS: Our findings demonstrate that the epigenome is reconfigured in a locus- and cell type-specific manner and epigenetic switching is common during cellular differentiation in both human and mouse.

Keywords DNA methylation      histone modifications      epigenome      epigenetic switch      cellular differentiation     
Corresponding Author(s): Zhaolan Zhou   
Just Accepted Date: 23 June 2016   Online First Date: 18 July 2016    Issue Date: 30 August 2016
 Cite this article:   
Ying-Tao Zhao,Maria Fasolino,Zhaolan Zhou. Locus- and cell type-specific epigenetic switching during cellular differentiation in mammals[J]. Front. Biol., 2016, 11(4): 311-322.
 URL:  
https://academic.hep.com.cn/fib/EN/10.1007/s11515-016-1411-5
https://academic.hep.com.cn/fib/EN/Y2016/V11/I4/311
Fig.1  Global DNA hypomethylation in somatic cells in both human and mouse. (A) Methylation levels (ML) of differentially methylated CpGs (diff-mCpGs) between ESCs and two somatic cell types in human and mouse. Each circle represents a CpG. The deeper the shade of blue, the higher the point density (number of CpGs). (B) The percentage of diff-mCpGs (FDR<0.05, two-tailed Fisher’s exact test, B-H correction) that are hypomethylated or hypermethylated during cellular differentiation. Confident CpGs are those with 10 or more read coverage in both ESCs and somatic cells.
Fig.2  DNA hypomethylation during cellular differentiation is locus- and cell type-specific. (A) A schematic of the enrichment-based statistical approach used to identify differentially methylated genomic regions (e.g. promoters). Confident CpG, CpGs with 10 or more read coverage in both data sets; diff-mCpGs, differentially methylated CpGs. (B) Heat maps of differentially methylated genomic regions between hESC and the two human somatic cell types after clustering analysis. (C) Browser representation of methylation profiles of the genes with changes in methylation levels at their promoter regions during cellular differentiation. Each green vertical bar represents a CpG, and the height of the bar represents its methylation level (from 0 to 100%). Grey arrows indicate the transcriptional orientation of each gene. Regions marked by orange bars indicate the locus- and cell type-specific DNA methylation changes.
Fig.3  Changes in histone modifications during cellular differentiation are locus- and cell type-specific. (A) The percentage of enriched sites for each histone modification that were gained (gain), lost (lose), and remained (remain) during differentiation of hESC to hFB and hEP. (B) Heat maps of changes of enriched sites for 11 histone modifications during cellular differentiation in human after clustering analysis.(C) The width of H3K27me3 and H3K9me3 enriched sites in hESC and hFB. ★★★, p-value<2.2 × 10-16, Student’s t-test. (D) Browser representation of two regions with H3K27me3 and H3K9me3 expansion during cellular differentiation in human.
Fig.4  A switching from DNA methylation to histone modifications during cellular differentiation. (A) DNA methylation changes in the highly dynamic regions that gain histone modifications during the differentiation of hESC to hFB. Green regions, histone modifications that are associated with a decrease in DNA methylation; Gray regions, histone modifications that are not associated with a change in DNA methylation; Red regions, histone modifications that are associated with an increase in DNA methylation. (B) DNA methylation changes in the highly dynamic gain regions during the differentiation of hESC to hEP. (C) DNA methylation changes in the highly dynamic regions that gain or lose H3K4me3 and H3K27ac during the differentiation of mESC to mFC. Enrichment score for each region was calculated by dividing the number of normalized reads aligned in a particular region in mFC by that in mESC. (D) Browser representation of three regions that exhibit switching from DNA methylation to histone modifications during cellular differentiation in human. Regions marked by gray bars indicate the cell type-specific switching. The track scale for all histone modifications is from 0 to 50 normalized reads.
Fig.5  Switching from DNA methylation to repressive histone modifications at promoter regions uncovers a change of gene regulation from ESCs to somatic cells. (A) Scatter plots of the number of genomic CpGs, the ratio of mCpG, and the ratio of diif-mCpG in 10 bp bins within the region 5 kb upstream and 1 kb downstream of known transcription start site (TSS). (B) Heat map-pie charts of the expression changes of the genes with hypomethylated promoters. FC, fold change, gene expression levels in somatic cell types divided by that in ESCs. (C) Pie charts of percentages of genes, which are hypomethylated at the promoter regions but with no expression changes during cellular differentiation, marked by H3K27me3 or H3K9me3 or both in somatic cells. (D) Browser representation of four genes with epigenomic reconfiguration at their promoter regions. These genes were silent in both hESC and somatic cell types. The track scale for all mRNA-seq is from 0 to 45 normalized reads. The track scale for all histone modifications is from 0 to 50 normalized reads. (E) A model depicting gene regulation changes during cellular differentiation.
ESCs:embryonic stem cells;
mCpGs:methylated CpGs;
hESC:human H1 ESCs;
hFB:IMR90 fetal lung fibroblasts;
hEP:mammary epithelial cells;
mESC:mouse ESCs;
mFB:mouse primary dermal fibroblasts;
mFC:mouse frontal cortex;
diff-mCpGs:differentially methylated CpG sites;
B-H:Benjamini and Hochberg;
TSSs:transcriptional start sites;
GO:gene ontology;
IGV:Integrative Genomics Viewer;
WGBS:whole-genome bisulfite sequencing;
FDR:False Discovery Rate;
CPM:count per million.
Tab.1  
1 Azuara V, Perry P, Sauer S, Spivakov M, Jørgensen H F, John R M, Gouti M, Casanova M, Warnes G, Merkenschlager M, Fisher A G (2006). Chromatin signatures of pluripotent cell lines. Nat Cell Biol, 8(5): 532–538
https://doi.org/10.1038/ncb1403 pmid: 16570078
2 Ball M P, Li J B, Gao Y, Lee J H, LeProust E M, Park I H, Xie B, Daley G Q, Church G M (2009). Targeted and genome-scale strategies reveal gene-body methylation signatures in human cells. Nat Biotechnol, 27(4): 361–368
https://doi.org/10.1038/nbt.1533 pmid: 19329998
3 Ben-Shushan E, Pikarsky E, Klar A, Bergman Y (1993). Extinction of Oct-3/4 gene expression in embryonal carcinoma x fibroblast somatic cell hybrids is accompanied by changes in the methylation status, chromatin structure, and transcriptional activity of the Oct-3/4 upstream region. Mol Cell Biol, 13(2): 891–901
https://doi.org/10.1128/MCB.13.2.891 pmid: 7678695
4 Benjamini Y, Hochberg Y (1995). Controlling the false discovery rate- a practical and powerful approach to multiple testing. J Roy Stat Soc B Met, 57: 289–300
5 Berger S L (2007). The complex language of chromatin regulation during transcription. Nature, 447(7143): 407–412
https://doi.org/10.1038/nature05915 pmid: 17522673
6 Bernstein B E, Mikkelsen T S, Xie X, Kamal M, Huebert D J, Cuff J, Fry B, Meissner A, Wernig M, Plath K, Jaenisch R, Wagschal A, Feil R, Schreiber S L, Lander E S (2006). A bivalent chromatin structure marks key developmental genes in embryonic stem cells. Cell, 125(2): 315–326
https://doi.org/10.1016/j.cell.2006.02.041 pmid: 16630819
7 Bird A (2002). DNA methylation patterns and epigenetic memory. Genes Dev, 16(1): 6–21
https://doi.org/10.1101/gad.947102 pmid: 11782440
8 Deb-Rinker P, Ly D, Jezierski A, Sikorska M, Walker P R (2005). Sequential DNA methylation of the Nanog and Oct-4 upstream regions in human NT2 cells during neuronal differentiation. J Biol Chem, 280(8): 6257–6260
https://doi.org/10.1074/jbc.C400479200 pmid: 15615706
9 Dobin A, Davis C A, Schlesinger F, Drenkow J, Zaleski C, Jha S, Batut P, Chaisson M, Gingeras T R (2013). STAR: ultrafast universal RNA-seq aligner. Bioinformatics, 29(1): 15–21
https://doi.org/10.1093/bioinformatics/bts635 pmid: 23104886
10 Gifford C A, Ziller M J, Gu H, Trapnell C, Donaghey J, Tsankov A, Shalek A K, Kelley D R, Shishkin A A, Issner R, Zhang X, Coyne M, Fostel J L, Holmes L, Meldrim J, Guttman M, Epstein C, Park H, Kohlbacher O, Rinn J, Gnirke A, Lander E S, Bernstein B E, Meissner A (2013). Transcriptional and epigenetic dynamics during specification of human embryonic stem cells. Cell, 153(5): 1149–1163
https://doi.org/10.1016/j.cell.2013.04.037 pmid: 23664763
11 Hawkins R D, Hon G C, Lee L K, Ngo Q, Lister R, Pelizzola M, Edsall L E, Kuan S, Luu Y, Klugman S, Antosiewicz-Bourget J, Ye Z, Espinoza C, Agarwahl S, Shen L, Ruotti V, Wang W, Stewart R, Thomson J A, Ecker J R, Ren B (2010). Distinct epigenomic landscapes of pluripotent and lineage-committed human cells. Cell Stem Cell, 6(5): 479–491
https://doi.org/10.1016/j.stem.2010.03.018 pmid: 20452322
12 Heinz S, Benner C, Spann N, Bertolino E, Lin Y C, Laslo P, Cheng J X, Murre C, Singh H, Glass C K (2010). Simple combinations of lineage-determining transcription factors prime cis-regulatory elements required for macrophage and B cell identities. Mol Cell, 38(4): 576–589
https://doi.org/10.1016/j.molcel.2010.05.004 pmid: 20513432
13 Hon G C, Hawkins R D, Caballero O L, Lo C, Lister R, Pelizzola M, Valsesia A, Ye Z, Kuan S, Edsall L E, Camargo A A, Stevenson B J, Ecker J R, Bafna V, Strausberg R L, Simpson A J, Ren B (2012). Global DNA hypomethylation coupled to repressive chromatin domain formation and gene silencing in breast cancer. Genome Res, 22(2): 246–258
https://doi.org/10.1101/gr.125872.111 pmid: 22156296
14 Hon G C, Rajagopal N, Shen Y, McCleary D F, Yue F, Dang M D, Ren B (2013). Epigenetic memory at embryonic enhancers identified in DNA methylation maps from adult mouse tissues. Nat Genet, 45(10): 1198–1206
https://doi.org/10.1038/ng.2746 pmid: 23995138
15 Huang W, Sherman B T, Lempicki R A (2009a). Bioinformatics enrichment tools: paths toward the comprehensive functional analysis of large gene lists. Nucleic Acids Res, 37(1): 1–13
https://doi.org/10.1093/nar/gkn923 pmid: 19033363
16 Huang W, Sherman B T, Lempicki R A (2009b). Systematic and integrative analysis of large gene lists using DAVID bioinformatics resources. Nat Protoc, 4(1): 44–57
https://doi.org/10.1038/nprot.2008.211 pmid: 19131956
17 Jackson M, Krassowska A, Gilbert N, Chevassut T, Forrester L, Ansell J, Ramsahoye B (2004). Severe global DNA hypomethylation blocks differentiation and induces histone hyperacetylation in embryonic stem cells. Mol Cell Biol, 24(20): 8862–8871
https://doi.org/10.1128/MCB.24.20.8862-8871.2004 pmid: 15456861
18 Jaenisch R, Bird A (2003). Epigenetic regulation of gene expression: how the genome integrates intrinsic and environmental signals. Nat Genet, 33(3s Suppl): 245–254
https://doi.org/10.1038/ng1089 pmid: 12610534
19 Jenuwein T, Allis C D (2001). Translating the histone code. Science, 293(5532): 1074–1080
https://doi.org/10.1126/science.1063127 pmid: 11498575
20 Jones P A (2012). Functions of DNA methylation: islands, start sites, gene bodies and beyond. Nat Rev Genet, 13(7): 484–492
https://doi.org/10.1038/nrg3230 pmid: 22641018
21 Koh K P, Yabuuchi A, Rao S, Huang Y, Cunniff K, Nardone J, Laiho A, Tahiliani M, Sommer C A, Mostoslavsky G, Lahesmaa R, Orkin S H, Rodig S J, Daley G Q, Rao A (2011). Tet1 and Tet2 regulate 5-hydroxymethylcytosine production and cell lineage specification in mouse embryonic stem cells. Cell Stem Cell, 8(2): 200–213
https://doi.org/10.1016/j.stem.2011.01.008 pmid: 21295276
22 Krueger F, Andrews S R (2011). Bismark: a flexible aligner and methylation caller for Bisulfite-Seq applications. Bioinformatics, 27(11): 1571–1572
https://doi.org/10.1093/bioinformatics/btr167 pmid: 21493656
23 Langmead B, Trapnell C, Pop M, Salzberg S L (2009). Ultrafast and memory-efficient alignment of short DNA sequences to the human genome. Genome Biol, 10(3): R25
https://doi.org/10.1186/gb-2009-10-3-r25 pmid: 19261174
24 Li H, Handsaker B, Wysoker A, Fennell T, Ruan J, Homer N, Marth G, Abecasis G, Durbin R, the 1000 Genome Project Data Processing Subgroup (2009). The sequence alignment/map format and SAMtools. Bioinformatics, 25(16): 2078–2079
https://doi.org/10.1093/bioinformatics/btp352 pmid: 19505943
25 Lister R, Ecker J R (2009). Finding the fifth base: genome-wide sequencing of cytosine methylation. Genome Res, 19(6): 959–966
https://doi.org/10.1101/gr.083451.108 pmid: 19273618
26 Lister R, Mukamel E A, Nery J R, Urich M, Puddifoot C A, Johnson N D, Lucero J, Huang Y, Dwork A J, Schultz M D, Yu M, Tonti-Filippini J, Heyn H, Hu S, Wu J C, Rao A, Esteller M, He C, Haghighi F G, Sejnowski T J, Behrens M M, Ecker J R (2013). Global epigenomic reconfiguration during mammalian brain development. Science, 341(6146): 1237905
https://doi.org/10.1126/science.1237905 pmid: 23828890
27 Lister R, Pelizzola M, Dowen R H, Hawkins R D, Hon G, Tonti-Filippini J, Nery J R, Lee L, Ye Z, Ngo Q M, Edsall L, Antosiewicz-Bourget J, Stewart R, Ruotti V, Millar A H, Thomson J A, Ren B, Ecker J R (2009). Human DNA methylomes at base resolution show widespread epigenomic differences. Nature, 462(7271): 315–322
https://doi.org/10.1038/nature08514 pmid: 19829295
28 Maniatis T, Reed R (2002). An extensive network of coupling among gene expression machines. Nature, 416(6880): 499–506
https://doi.org/10.1038/416499a pmid: 11932736
29 Mann I K, Chatterjee R, Zhao J, He X, Weirauch M T, Hughes T R, Vinson C (2013). CG methylated microarrays identify a novel methylated sequence bound by the CEBPB|ATF4 heterodimer that is active in vivo. Genome Res, 23(6): 988–997
https://doi.org/10.1101/gr.146654.112 pmid: 23590861
30 Margueron R, Reinberg D (2011). The Polycomb complex PRC2 and its mark in life. Nature, 469(7330): 343–349
https://doi.org/10.1038/nature09784 pmid: 21248841
31 Métivier R, Penot G, Hübner M R, Reid G, Brand H, Kos M, Gannon F (2003). Estrogen receptor-alpha directs ordered, cyclical, and combinatorial recruitment of cofactors on a natural target promoter. Cell, 115(6): 751–763
https://doi.org/10.1016/S0092-8674(03)00934-6 pmid: 14675539
32 Pan G, Tian S, Nie J, Yang C, Ruotti V, Wei H, Jonsdottir G A, Stewart R, Thomson J A (2007). Whole-genome analysis of histone H3 lysine 4 and lysine 27 methylation in human embryonic stem cells. Cell Stem Cell, 1(3): 299–312
https://doi.org/10.1016/j.stem.2007.08.003 pmid: 18371364
33 Reik W (2007). Stability and flexibility of epigenetic gene regulation in mammalian development. Nature, 447(7143): 425–432
https://doi.org/10.1038/nature05918 pmid: 17522676
34 Rivera C M, Ren B (2013). Mapping human epigenomes. Cell, 155(1): 39–55
https://doi.org/10.1016/j.cell.2013.09.011 pmid: 24074860
35 Robinson J T, Thorvaldsdóttir H, Winckler W, Guttman M, Lander E S, Getz G, Mesirov J P (2011). Integrative genomics viewer. Nat Biotechnol, 29(1): 24–26
https://doi.org/10.1038/nbt.1754 pmid: 21221095
36 Robinson M D, McCarthy D J, Smyth G K (2010). edgeR: a Bioconductor package for differential expression analysis of digital gene expression data. Bioinformatics, 26(1): 139–140
https://doi.org/10.1093/bioinformatics/btp616 pmid: 19910308
37 Smith Z D, Meissner A (2013). DNA methylation: roles in mammalian development. Nat Rev Genet, 14(3): 204–220
https://doi.org/10.1038/nrg3354 pmid: 23400093
38 Stadler M B, Murr R, Burger L, Ivanek R, Lienert F, Schöler A, van Nimwegen E, Wirbelauer C, Oakeley E J, Gaidatzis D, Tiwari V K, Schübeler D (2011). DNA-binding factors shape the mouse methylome at distal regulatory regions. Nature, 480(7378): 490–495
pmid: 22170606
39 Tahiliani M, Koh K P, Shen Y, Pastor W A, Bandukwala H, Brudno Y, Agarwal S, Iyer L M, Liu D R, Aravind L, Rao A (2009). Conversion of 5-methylcytosine to 5-hydroxymethylcytosine in mammalian DNA by MLL partner TET1. Science, 324(5929): 930–935
https://doi.org/10.1126/science.1170116 pmid: 19372391
40 Thorvaldsdóttir H, Robinson J T, Mesirov J P (2013). Integrative genomics viewer (IGV): high-performance genomics data visualization and exploration. Brief Bioinform, 14(2): 178–192
https://doi.org/10.1093/bib/bbs017 pmid: 22517427
41 Trapnell C, Pachter L, Salzberg S L (2009). TopHat: discovering splice junctions with RNA-Seq. Bioinformatics, 25(9): 1105–1111
https://doi.org/10.1093/bioinformatics/btp120 pmid: 19289445
42 Tsumura A, Hayakawa T, Kumaki Y, Takebayashi S, Sakaue M, Matsuoka C, Shimotohno K, Ishikawa F, Li E, Ueda H R, Nakayama J, Okano M (2006). Maintenance of self-renewal ability of mouse embryonic stem cells in the absence of DNA methyltransferases Dnmt1, Dnmt3a and Dnmt3b. Genes Cells, 11(7): 805–814
https://doi.org/10.1111/j.1365-2443.2006.00984.x pmid: 16824199
43 Turner B M (2007). Defining an epigenetic code. Nat Cell Biol, 9(1): 2–6
https://doi.org/10.1038/ncb0107-2 pmid: 17199124
44 Xie W, Barr C L, Kim A, Yue F, Lee A Y, Eubanks J, Dempster E L, Ren B (2012). Base-resolution analyses of sequence and parent-of-origin dependent DNA methylation in the mouse genome. Cell, 148(4): 816–831
https://doi.org/10.1016/j.cell.2011.12.035 pmid: 22341451
45 Xie W, Schultz M D, Lister R, Hou Z, Rajagopal N, Ray P, Whitaker J W, Tian S, Hawkins R D, Leung D, Yang H, Wang T, Lee A Y, Swanson S A, Zhang J, Zhu Y, Kim A, Nery J R, Urich M A, Kuan S, Yen C A, Klugman S, Yu P, Suknuntha K, Propson N E, Chen H, Edsall L E, Wagner U, Li Y, Ye Z, Kulkarni A, Xuan Z, Chung W Y, Chi N C, Antosiewicz-Bourget J E, Slukvin I, Stewart R, Zhang M Q, Wang W, Thomson J A, Ecker J R, Ren B (2013). Epigenomic analysis of multilineage differentiation of human embryonic stem cells. Cell, 153(5): 1134–1148
https://doi.org/10.1016/j.cell.2013.04.022 pmid: 23664764
46 Ziller M J, Gu H, Müller F, Donaghey J, Tsai L T, Kohlbacher O, De Jager P L, Rosen E D, Bennett D A, Bernstein B E, Gnirke A, Meissner A (2013). Charting a dynamic DNA methylation landscape of the human genome. Nature, 500(7463): 477–481
https://doi.org/10.1038/nature12433 pmid: 23925113
[1] FIB-10411-of-ZLZ_suppl_1 Download
[2] FIB-10411-of-ZLZ_suppl_2 Download
[1] Andrew Brandmaier, Sheng-Qi Hou, Sandra Demaria, Silvia C. Formenti, Wen H. Shen. PTEN at the interface of immune tolerance and tumor suppression[J]. Front. Biol., 2017, 12(3): 163-174.
[2] Anita E. Autry,Megumi Adachi,Lisa M. Monteggia. Dynamic methylation driven by neuronal activity in hippocampal neurons impacts complex behavior[J]. Front. Biol., 2015, 10(5): 439-447.
[3] Joshua D. TOMPKINS,Arthur D. RIGGS. An epigenetic perspective on the failing heart and pluripotent-derived-cardiomyocytes for cell replacement therapy[J]. Front. Biol., 2015, 10(1): 11-27.
[4] Qichao WANG, Xianmin ZHU, Yun FENG, Zhigang XUE, Guoping FAN. Single-cell genomics: An overview[J]. Front Biol, 2013, 8(6): 569-576.
[5] Feng C. ZHOU. DNA methylation program during development[J]. Front Biol, 2012, 7(6): 485-494.
[6] Xiangpeng DAI, Xiaoyang ZHAO, Hai TANG, Jie HAO, Jean-Paul RENARD, Qi ZHOU, Alice JOUNEAU, Liu WANG. Cloning efficiency following ES cell nuclear transfer is influenced by the methylation state of the donor nucleus altered by mutation of DNA methyltransferase 3a and 3b[J]. Front Biol, 2010, 5(5): 439-444.
[7] Wei YANG, Qi ZHOU, Xiu-Jie WANG, . Regulation beyond genome sequences: DNA and histone methylation in embryonic stem cells[J]. Front. Biol., 2010, 5(1): 41-47.
Viewed
Full text


Abstract

Cited

  Shared   
  Discussed