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.

  • Article
  • Published:

Senescent cells harbour features of the cancer epigenome

Abstract

Altered DNA methylation and associated destabilization of genome integrity and function is a hallmark of cancer. Replicative senescence is a tumour suppressor process that imposes a limit on the proliferative potential of normal cells that all cancer cells must bypass. Here we show by whole-genome single-nucleotide bisulfite sequencing that replicative senescent human cells exhibit widespread DNA hypomethylation and focal hypermethylation. Hypomethylation occurs preferentially at gene-poor, late-replicating, lamin-associated domains and is linked to mislocalization of the maintenance DNA methyltransferase (DNMT1) in cells approaching senescence. Low-level gains of methylation are enriched in CpG islands, including at genes whose methylation and silencing is thought to promote cancer. Gains and losses of methylation in replicative senescence are thus qualitatively similar to those in cancer, and this ‘reprogrammed’ methylation landscape is largely retained when cells bypass senescence. Consequently, the DNA methylome of senescent cells might promote malignancy, if these cells escape the proliferative barrier.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Figure 1: Senescent cells exhibit overall hypomethylation and focal hypermethylation.
Figure 2: Promoter proximal methylation of transcriptionally repressed cell cycle genes.
Figure 3: DNA hypomethylation occurs before cell cycle exit and is associated with mislocalization of DNMT1.
Figure 4: Hypomethylation and expression of late-replicating satellite sequences in senescent cells.
Figure 5: Hypomethylation occurs at gene-poor, late-replicating and lamin-associated domains whereas hypermethylation occurs at gene-rich, early-replicating regions and CpG islands.
Figure 6: Methylation changes in senescence resemble those in cancer.
Figure 7: Altered methylation is retained in cells that bypass senescence.
Figure 8: The altered epigenome of senescent cells might promote age-associated increase in incidence of human cancers.

Similar content being viewed by others

Accession codes

Primary accessions

Gene Expression Omnibus

Referenced accessions

Gene Expression Omnibus

References

  1. Ting, A. H., McGarvey, K. M. & Baylin, S. B. The cancer epigenome–components and functional correlates. Genes Dev. 20, 3215–3231 (2006).

    Article  CAS  PubMed  Google Scholar 

  2. Sproul, D. & Meehan, R. R. Genomic insights into cancer-associated aberrant CpG island hypermethylation. Brief Funct. Genomic. 12, 174–190 (2013).

    Article  CAS  Google Scholar 

  3. Hon, G. C. et al. Global DNA hypomethylation coupled to repressive chromatin domain formation and gene silencing in breast cancer. Genome. Res. 22, 246–258 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Kuilman, T., Michaloglou, C., Mooi, W. J. & Peeper, D. S. The essence of senescence. Genes Dev. 24, 2463–2479 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Michaloglou, C. et al. BRAFE600-associated senescence-like cell cycle arrest of human naevi. Nature 436, 720–724 (2005).

    Article  CAS  PubMed  Google Scholar 

  6. Chen, Z. et al. Crucial role of p53-dependent cellular senescence in suppression of Pten-deficient tumorigenesis. Nature 436, 725–730 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Braig, M. et al. Oncogene-induced senescence as an initial barrier in lymphoma development. Nature 436, 660–665 (2005).

    Article  CAS  PubMed  Google Scholar 

  8. Feldser, D. M. & Greider, C. W. Short telomeres limit tumour progression in vivo by inducing senescence. Cancer Cell 11, 461–469 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Cosme-Blanco, W. et al. Telomere dysfunction suppresses spontaneous tumorigenesis in vivo by initiating p53-dependent cellular senescence. EMBO Rep. 8, 497–503 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Choi, M. R. et al. Genome-scale DNA methylation pattern profiling of human bone marrow mesenchymal stem cells in long-term culture. Exp. Mol. Med. 44, 503–512 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Narita, M. et al. Rb-mediated heterochromatin formation and silencing of E2F target genes during cellular senescence. Cell 113, 703–716 (2003).

    Article  CAS  PubMed  Google Scholar 

  12. Zhang, R. et al. Formation of MacroH2A-containing senescence-associated heterochromatin foci and senescence driven by ASF1a and HIRA. Dev. Cell 8, 19–30 (2005).

    Article  CAS  PubMed  Google Scholar 

  13. O’Sullivan, R. J., Kubicek, S., Schreiber, S. L. & Karlseder, J. Reduced histone biosynthesis and chromatin changes arising from a damage signal at telomeres. Nat. Struct. Mol. Biol. 17, 1218–1225 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

  14. Wilson, V. L. & Jones, P. A. DNA methylation decreases in ageing but not in immortal cells. Science 220, 1055–1057 (1983).

    Article  CAS  PubMed  Google Scholar 

  15. Schellenberg, A. et al. Replicative senescence of mesenchymal stem cells causes DNA-methylation changes which correlate with repressive histone marks. Aging 3, 873–888 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Chandra, T. et al. Independence of repressive histone marks and chromatin compaction during senescent heterochromatic layer formation. Mol. Cell 47, 203–214 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Sadaie, M. et al. Redistribution of the Lamin B1 genomic binding profile affects rearrangement of heterochromatic domains and SAHF formation during senescence. Genes Dev. 27, 1800–1808 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Shah, P. P. et al. Lamin B1 depletion in senescent cells triggers large-scale changes in gene expression and the chromatin landscape. Genes Dev. 27, 1787–1799 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Forsyth, N. R., Evans, A. P., Shay, J. W. & Wright, W. E. Developmental differences in the immortalization of lung fibroblasts by telomerase. Aging Cell 2, 235–243 (2003).

    Article  CAS  PubMed  Google Scholar 

  20. Coppe, J. P., Desprez, P. Y., Krtolica, A. & Campisi, J. The senescence-associated secretory phenotype: the dark side of tumour suppression. Annu. Rev. Pathol. 5, 99–118 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Young, J. I., Sedivy, J. M. & Smith, J. R. Telomerase expression in normal human fibroblasts stabilizes DNA 5-methylcytosine transferase I. J. Biol. Chem. 278, 19904–19908 (2003).

    Article  CAS  PubMed  Google Scholar 

  22. Lister, R. et al. Human DNA methylomes at base resolution show widespread epigenomic differences. Nature 462, 315–322 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Vanderkraats, N. D., Hiken, J. F., Decker, K. F. & Edwards, J. R. Discovering high-resolution patterns of differential DNA methylation that correlate with gene expression changes. Nucleic Acids Res. 41, 6816–6827 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Leonhardt, H., Page, A. W., Weier, H. U. & Bestor, T. H. A targeting sequence directs DNA methyltransferase to sites of DNA replication in mammalian nuclei. Cell 71, 865–873 (1992).

    Article  CAS  PubMed  Google Scholar 

  25. Lopatina, N. et al. Differential maintenance and de novo methylating activity by three DNA methyltransferases in ageing and immortalized fibroblasts. J. Cell Biochem. 84, 324–334 (2002).

    Article  PubMed  Google Scholar 

  26. Suzuki, T., Fujii, M. & Ayusawa, D. Demethylation of classical satellite 2 and 3 DNA with chromosomal instability in senescent human fibroblasts. Exp. Gerontol. 37, 1005–1014 (2002).

    Article  CAS  PubMed  Google Scholar 

  27. Enukashvily, N. I., Donev, R., Waisertreiger, I. S. & Podgornaya, O. I. Human chromosome 1 satellite 3 DNA is decondensed, demethylated and transcribed in senescent cells and in A431 epithelial carcinoma cells. Cytogenet. Genome. Res. 118, 42–54 (2007).

    Article  CAS  PubMed  Google Scholar 

  28. Yamakoshi, K. et al. Real-time in vivo imaging of p16Ink4a reveals cross talk with p53. J. Cell Biol. 186, 393–407 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Young, J. I. & Smith, J. R. DNA methyltransferase inhibition in normal human fibroblasts induces a p21-dependent cell cycle withdrawal. J. Biol. Chem. 276, 19610–19616 (2001).

    Article  CAS  PubMed  Google Scholar 

  30. Aran, D., Toperoff, G., Rosenberg, M. & Hellman, A. Replication timing-related and gene body-specific methylation of active human genes. Hum. Mol. Genet. 20, 670–680 (2011).

    Article  CAS  PubMed  Google Scholar 

  31. Hansen, R. S. et al. Sequencing newly replicated DNA reveals widespread plasticity in human replication timing. Proc. Natl Acad. Sci. USA 107, 139–144 (2010).

    Article  CAS  PubMed  Google Scholar 

  32. Moir, R. D., Montag-Lowy, M. & Goldman, R. D. Dynamic properties of nuclear lamins: lamin B is associated with sites of DNA replication. J. Cell Biol. 125, 1201–1212 (1994).

    Article  CAS  PubMed  Google Scholar 

  33. Peric-Hupkes, D. et al. Molecular maps of the reorganization of genome-nuclear lamina interactions during differentiation. Mol. Cell 38, 603–613 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Yaffe, E. et al. Comparative analysis of DNA replication timing reveals conserved large-scale chromosomal architecture. PLoS Genet. 6, e1001011 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

  35. Berman, B. P. et al. Regions of focal DNA hypermethylation and long-range hypomethylation in colorectal cancer coincide with nuclear lamina-associated domains. Nat. Genet. 44, 40–46 (2012).

    Article  CAS  Google Scholar 

  36. Hansen, K. D. et al. Increased methylation variation in epigenetic domains across cancer types. Nat. Genet. 43, 768–775 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Ting, D. T. et al. Aberrant overexpression of satellite repeats in pancreatic and other epithelial cancers. Science 331, 593–596 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Toyota, M. et al. CpG island methylator phenotype in colorectal cancer. Proc. Natl Acad. Sci. USA 96, 8681–8686 (1999).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Weisenberger, D. J. et al. CpG island methylator phenotype underlies sporadic microsatellite instability and is tightly associated with BRAF mutation in colorectal cancer. Nat. Genet. 38, 787–793 (2006).

    Article  CAS  PubMed  Google Scholar 

  40. Irizarry, R. A et al. The human colon cancer methylome shows similar hypo- and hypermethylation at conserved tissue-specific CpG island shores. Nat. Genet. 41, 178–186 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. De Carvalho, D. D. et al. DNA methylation screening identifies driver epigenetic events of cancer cell survival. Cancer Cell 21, 655–667 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Shimi, T. et al. The role of nuclear lamin B1 in cell proliferation and senescence. Genes Dev. 25, 2579–2593 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Unterberger, A., Andrews, S. D., Weaver, I. C. & Szyf, M. DNA methyltransferase 1 knockdown activates a replication stress checkpoint. Mol. Cell Biol. 26, 7575–7586 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Suzuki, H. et al. Epigenetic inactivation of SFRP genes allows constitutive WNT signalling in colorectal cancer. Nat. Genet. 36, 417–422 (2004).

    Article  CAS  PubMed  Google Scholar 

  45. Zhang, W. et al. Comparison of global DNA methylation profiles in replicative versus premature senescence. Life Sci. 83, 475–480 (2008).

    Article  CAS  PubMed  Google Scholar 

  46. Casillas, M. A. Jr., Lopatina, N., Andrews, L. G. & Tollefsbol, T. O. Transcriptional control of the DNA methyltransferases is altered in ageing and neoplastically-transformed human fibroblasts. Mol. Cell Biochem. 252, 33–43 (2003).

    Article  CAS  PubMed  Google Scholar 

  47. Eden, A., Gaudet, F., Waghmare, A. & Jaenisch, R. Chromosomal instability and tumours promoted by DNA hypomethylation. Science 300, 455 (2003).

    Article  CAS  PubMed  Google Scholar 

  48. Gaudet, F. et al. Induction of tumours in mice by genomic hypomethylation. Science 300, 489–492 (2003).

    Article  CAS  PubMed  Google Scholar 

  49. Song, J. Z., Stirzaker, C., Harrison, J., Melki, J. R. & Clark, S. J. Hypermethylation trigger of the glutathione-S-transferase gene (GSTP1) in prostate cancer cells. Oncogene 21, 1048–1061 (2002).

    Article  CAS  PubMed  Google Scholar 

  50. Landan, G. et al. Epigenetic polymorphism and the stochastic formation of differentially methylated regions in normal and cancerous tissues. Nat. Genet. 44, 1207–1214 (2012).

    Article  CAS  PubMed  Google Scholar 

  51. O’Hagan, H. M., Mohammad, H. P. & Baylin, S. B. Double strand breaks can initiate gene silencing and SIRT1-dependent onset of DNA methylation in an exogenous promoter CpG island. PLoS Genet. 4, e1000155 (2008).

    Article  PubMed  PubMed Central  Google Scholar 

  52. Ibrahim, A. E. et al. Sequential DNA methylation changes are associated with DNMT3B overexpression in colorectal neoplastic progression. Gut 60, 499–508 (2011).

    Article  CAS  PubMed  Google Scholar 

  53. Nosho, K. et al. DNMT3B expression might contribute to CpG island methylator phenotype in colorectal cancer. Clin. Cancer Res. 15, 3663–3671 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Carragher, L. A. et al. V600EBraf induces gastrointestinal crypt senescence and promotes tumour progression through enhanced CpG methylation of p16INK4a. EMBO Mol. Med. 2, 458–471 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Walsh, C. P., Chaillet, J. R. & Bestor, T. H. Transcription of IAP endogenous retroviruses is constrained by cytosine methylation. Nat. Genet. 20, 116–117 (1998).

    Article  CAS  PubMed  Google Scholar 

  56. O’Neill, R. J., O’Neill, M. J. & Graves, J. A. Undermethylation associated with retroelement activation and chromosome remodelling in an interspecific mammalian hybrid. Nature 393, 68–72 (1998).

    Article  PubMed  Google Scholar 

  57. Lee, E. et al. Landscape of somatic retrotransposition in human cancers. Science 337, 967–971 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. De Cecco, M. et al. Genomes of replicatively senescent cells undergo global epigenetic changes leading to gene silencing and activation of transposable elements. Aging Cell 12, 247–256 (2013).

    Article  CAS  PubMed  Google Scholar 

  59. Dimri, G. P. et al. A biomarker that identifies senescent human cells in culture and in ageing skin in vivo. Proc. Natl Acad. Sci. USA 92, 9363–9367 (1995).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Herbig, U., Ferreira, M., Condel, L., Carey, D. & Sedivy, J. M. Cellular senescence in ageing primates. Science 311, 1257 (2006).

    Article  CAS  PubMed  Google Scholar 

  61. Sedelnikova, O. A. et al. Senescing human cells and ageing mice accumulateDNA lesions with unrepairable double-strand breaks. Nat. Cell Biol. 6, 168–170 (2004).

    Article  CAS  PubMed  Google Scholar 

  62. Feinberg, A. P., Ohlsson, R. & Henikoff, S. The epigenetic progenitor origin of human cancer. Nat. Rev. Genet. 7, 21–33 (2006).

    Article  CAS  PubMed  Google Scholar 

  63. Vredeveld, L. C. et al. Abrogation of BRAFV600E-induced senescence by PI3K pathway activation contributes to melanomagenesis. Genes. Dev. 26, 1055–1069 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Kennedy, A. L. et al. Activation of the PIK3CA/AKT pathway suppresses senescence induced by an activated RAS oncogene to promote tumorigenesis. Mol. Cell 42, 36–49 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Pchelintsev, N. A. et al. the HIRA histone chaperone complex in the chromatin landscape. Cell Rep. 3, 1012–1019 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Debacq-Chainiaux, F., Erusalimsky, J. D., Campisi, J. & Toussaint, O. Protocols to detect senescence-associated beta-galactosidase (SA-betagal) activity, a biomarker of senescent cells in culture and in vivo. Nat. Protoc. 4, 1798–1806 (2009).

    Article  CAS  PubMed  Google Scholar 

  67. Harlow, E. & Lane, D. Antibodies: A Laboratory Manual (Cold Spring Harbor Laboratory Press, 1988).

    Google Scholar 

  68. Langmead, B., Trapnell, C., Pop, M. & Salzberg, S. L. Ultrafast and memory-efficient alignment of short DNA sequences to the human genome. Genome Biol. 10, R25 (2009).

    Article  PubMed  PubMed Central  Google Scholar 

  69. Krueger, F. & Andrews, S. R. Bismark: a flexible aligner and methylation caller for Bisulfite-Seq applications. Bioinformatics 27, 1571–1572 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Hansen, K. D., Langmead, B. & Irizarry, R. A. BSmooth: from whole genome bisulfite sequencing reads to differentially methylated regions. Genome Biol. 13, R83 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

Thanks to S. Pepper in the CRUK microarray facility and to S. Hansen for assistance with DNA replication timing data. Thanks to Beijing Genome Institute for bisulfite sequencing. Work in the laboratory of P.D.A. was funded by NIA Program Project P01 AG031862 and CRUK Program A10250. S.L.B.’s laboratory was funded by NIA Program Project P01 AG031862. R.R.M.’s laboratory was funded by the MRC and the BBSRC. P.D.A. thanks P. Cairns for critical formative discussions.

Author information

Authors and Affiliations

Authors

Contributions

H.A.C. carried out the bulk of the experiments. D.M.N., P.P.S., J.v.T., T.S.R., C.B., M.E.D. and D.S.D. carried out further experiments. T.M. carried out the bulk of the data analysis. N.D.V. and G.D. carried out further data analyses. H.A.C. and T.M. provided substantial and critical intellectual input. R.R.M., J.R.E. and S.L.B. provided further intellectual input. P.D.A., H.A.C. and T.M. conceived the project and wrote the manuscript.

Corresponding author

Correspondence to Peter D. Adams.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Integrated supplementary information

Supplementary Figure 1 Confirmation of senescence in IMR-90 cells.

(A) Growth curve of IMR-90 cells grown in 3% O2 shows proliferation ceased at population doubling (PD) 88. Scale bar = 30 μm. (B) Senescence-Associated β-galactosidase staining in proliferating (prolif), PD28 and senescent (sen), PD88. (C) Quantitation of senescence-associated β-galactosidase (SA β-gal) positive cells. (D) Immunofluorescence of proliferating and senescent cells with a marker of cell proliferation, cyclin A and 4’, 6-diamidino-2-phenylindole (DAPI). Scale bar = 5 μm. (E) Quantitation of cyclin A positive cells. (F) Staining of proliferating and senescent cells with DAPI shows senescent cells displaying senescence associated heterochromatin foci (SAHF). Scale bar = 5 μm. (G) Quantitation of SAHF positive cells. (H) Immunofluorescence of PML and HIRA in proliferating and senescent cells shows co-localization of these 2 proteins, a known marker of senescence in the senescent population. (I) Quantitation of cells with HIRA and PML co-localization. In panels (C), (E), (G) and (I), data was obtained from at least 100 cells scored from a single sample, representative of at least 10 independent samples.

Supplementary Figure 2 Altered gene expression in senescence.

(A) Heatmap showing hierarchical clustering of gene expression in proliferating and senescent cells. Significant changed probes of fold change > = 1.5 and BH-fdr(tt) < = 0.05. (B) Gene set enrichment analysis of downregulated genes with normalized enrichment score (NES) and family wise-error rate (FWER) p-value (C) Gene set enrichment plot of cell cycle process in senescence, top part shows the enrichment value for each gene in this class and the bottom part, the ranked list metric of these genes. (D) Heatmap showing hierarchical clustering of expression of genes in gene set “inflammatory response” (http://www.broadinstitute.org/gsea/msigdb/cards/INFLAMMATORY{_}RESPONSE.html) in proliferating and senescent cells. Significant changed probes of fold change > = 1.5 and BH-fdr(tt) < = 0.05.

Supplementary Figure 3 Concordance of replicates and methylation changes across all chromosomes.

(A) Overlayed percentage methylated basecall plots of proliferating (blue) and senescent (orange) for each replicate pair. Chromosome 1 (chr 1) is shown as a representative region. (B) Difference p plots of all chromosomes (chr).

Supplementary Figure 4 Methylation changes relative to gene expression.

(A)–(C) Relative level of gene expression in proliferating cells against the difference in methylation between proliferating and senescent cells (Sen-Prolif). Methylation was scored at promoters, gene bodies and promoters containing CpG islands defined in UCSC, as indicated. (D)–(F) Ln fold change of gene expression between proliferating and senescent cells (positive values, increased expression in senescence; negative values, decreased expression in senescence) against the difference in methylation between proliferating and senescent cells (Sen-Prolif). Methylation was scored at promoters, gene bodies and promoters containing CpG islands, as indicated. (G) Same analysis as in F, but only for genes expressed above the median level of expression in proliferating cells.

Supplementary Figure 5 Promoters of repressed cell cycle genes are methylated in senescence.

Plots of differential methylation versus position up and downstream of TSS (−5kb to +5kb) for selected genes. Y-axis is a differential methylation score that ranges from −1 to 1, denoting complete hypomethylation and hypermethylation, respectively. The full list of genes is in Supplementary Dataset 3. In each plot, the vertical line marks the TSS. Fold change (log2) gene expression of each gene is indicated in green. For each gene, data from all 3 replicates is shown.

Supplementary Figure 6 Knock down of DNMT1 triggers cell senescence, expression of satellite 2 RNA and senescence-associated chromatin changes; and overlap of hypomethylated DMRs in senescence and cancer.

(A) Proliferating IMR90 fibroblasts were infected with control lentivirus (EV) or lentivirus encoding independent shRNAs to DNMT1 (shDNMT1-a or shDNMT1-b), selected in puromycin and western blotted to detect DNMT1. (B) Cells from (A) were passaged until proliferation arrest. (CG) After proliferation arrest, cells from (A) were scored by immunofluorescence for expression of cyclin A (C), expression of SA β-gal. (Scale bar = 30 μM), (D), expression of satellite 2 RNA (E), Senescence Associated Heterochromatin Foci (F) and localization of histone chaperone HIRA to PML bodies (G). For (C), (F) and (G) n = 1, but results shown with 2 independent shRNAs and similar results previously reported by others (see main text). (H) Series of graphs assessing percent overlap in total bp over whole genome of indicated features (observed) compared to overlaps calculated for random. Asterisks indicate statistical significance and a p-value of 0.001. Hypomethylated cancer (hypo cDMR) and senescence DMRs (hypo sDMR) greater than or equal to 100Kb, greater than or equal to 250Kb, greater than or equal to 500Kb, greater than or equal to 1Mb, greater than or equal to 2Mb, as indicated.

Supplementary Figure 7 Increased methylation of CpG islands in senescence.

Methylation of indicated CpG islands in senescence. Plot of percent methylated basecalls in proliferating (orange) and senescent cells (blue), from whole genome bisulfite sequencing data of 3 replicates of proliferating cells and 3 replicates of senescent cells. The orange and blue lines show the smoothed average percent methylated basecalls at corresponding CpGs. Individual CpGs are indicated by black ticks along the x-axis. The UCSC genes (blue bar) and CpG islands (green bar) are also shown. The transcription start sites (TSS) are indicated by vertical black arrows. Gene, chromosome and bp of CpG island are indicated top left.

Supplementary Figure 8 SV40-infected “bypass” cells proliferate and uncropped versions of Figures.

(A) Bypass cells (SV40) exhibit a low frequency of SA β-gal (<1%). (B) A large proportion of bypass cells (SV40) incorporate a EdU (DNA synthesis) after a 24hr pulse. (C) Quantitation of results from (B), compared to uninfected PD 22 proliferating cells. Error bars indicate standard deviation. Source data for panel (C) can be found in Supplemental Table 22. (D) Uncropped Figure 3c. (E) Uncropped Supplementary Figure 8a. For (D) and (E), see main figures for loading controls. Scale bar in (A) and (B) = 10 μM.

Supplementary Table 1 Sequence yield.
Supplementary Table 2 CpG Methylation.
Supplementary Table 3 Methylated CpG sites.
Supplementary Table 4 Pearson Correlation Coefficients.
Supplementary Table 5 CHG Methylation.
Supplementary Table 6 CHH Methylation.
Supplementary Table 7 Error rates.
Supplementary Table 8 Individual CpG differences.
Supplementary Table 9 Genes whose promoter methylation increases and expression decreases in senescence.
Supplementary Table 10 Overlap of sDMRs, LADs, CpG islands, early and late replicating regions of the genome.
Supplementary Table 11 Overlap of sDMRs with cDMRs described in Ref. 36.
Supplementary Table 12 Overlap of sDMRs with cDMRs described in Ref. 35.
Supplementary Table 13 Overlap of sDMRs with cDMRs described in Ref. 3.
Supplementary Table 14 Number (no.) of CpGs call classified as methylated and unmethylated in indicated regions (CpG islands) in proliferating cells (P) and senescent cells (S).
Supplementary Table 15 Overlap of sDMRs with bypass DMRs.
Supplementary Table 16 Overlap of sDMR and bypass DMR intersect with cDMRs described in Ref. 36.
Supplementary Table 17 Overlap of sDMR and bypass DMR intersect with cDMRs described in Ref. 35.
Supplementary Table 18 Overlap of sDMR and bypass DMR intersect with cDMRs described in Ref. 3.
Supplementary Table 19 Statistics source data.
Supplementary Table 20 Antibodies and shRNAs used in this study.

Supplementary information

Supplementary Information

Supplementary Information (PDF 4428 kb)

Supplementary Table 1

Supplementary Information (XLSX 9 kb)

Supplementary Table 2

Supplementary Information (XLSX 8 kb)

Supplementary Table 3

Supplementary Information (XLSX 8 kb)

Supplementary Table 4

Supplementary Information (XLSX 8 kb)

Supplementary Table 5

Supplementary Information (XLSX 8 kb)

Supplementary Table 6

Supplementary Information (XLSX 8 kb)

Supplementary Table 7

Supplementary Information (XLSX 8 kb)

Supplementary Table 8

Supplementary Information (XLSX 7 kb)

Supplementary Table 9

Supplementary Information (XLSX 28 kb)

Supplementary Table 10

Supplementary Information (XLSX 125 kb)

Supplementary Table 11

Supplementary Information (XLSX 20 kb)

Supplementary Table 12

Supplementary Information (XLSX 107 kb)

Supplementary Table 13

Supplementary Information (XLSX 9 kb)

Supplementary Table 14

Supplementary Information (XLSX 9 kb)

Supplementary Table 15

Supplementary Information (XLSX 9 kb)

Supplementary Table 16

Supplementary Information (XLSX 9 kb)

Supplementary Table 17

Supplementary Information (XLSX 11 kb)

Supplementary Table 18

Supplementary Information (XLSX 9 kb)

Supplementary Table 19

Supplementary Information (XLSX 9 kb)

Supplementary Table 20

Supplementary Information (XLSX 9 kb)

Supplementary Table 21

Supplementary Information (XLSX 9 kb)

Supplementary Table 22

Supplementary Information (XLSX 56 kb)

Supplementary Table 23

Supplementary Information (XLSX 10 kb)

Rights and permissions

Reprints and permissions

About this article

Cite this article

Cruickshanks, H., McBryan, T., Nelson, D. et al. Senescent cells harbour features of the cancer epigenome. Nat Cell Biol 15, 1495–1506 (2013). https://doi.org/10.1038/ncb2879

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/ncb2879

This article is cited by

Search

Quick links

Nature Briefing: Cancer

Sign up for the Nature Briefing: Cancer newsletter — what matters in cancer research, free to your inbox weekly.

Get what matters in cancer research, free to your inbox weekly. Sign up for Nature Briefing: Cancer