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:

Incomplete DNA methylation underlies a transcriptional memory of somatic cells in human iPS cells

Abstract

Human induced pluripotent stem (iPS) cells are remarkably similar to embryonic stem (ES) cells, but recent reports indicate that there may be important differences between them. We carried out a systematic comparison of human iPS cells generated from hepatocytes (representative of endoderm), skin fibroblasts (mesoderm) and melanocytes (ectoderm). All low-passage iPS cells analysed retain a transcriptional memory of the original cells. The persistent expression of somatic genes can be partially explained by incomplete promoter DNA methylation. This epigenetic mechanism underlies a robust form of memory that can be found in iPS cells generated by multiple laboratories using different methods, including RNA transfection. Incompletely silenced genes tend to be isolated from other genes that are repressed during reprogramming, indicating that recruitment of the silencing machinery may be inefficient at isolated genes. Knockdown of the incompletely reprogrammed gene C9orf64 (chromosome 9 open reading frame 64) reduces the efficiency of human iPS cell generation, indicating that somatic memory genes may be functionally relevant during reprogramming.

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: Pluripotency validation for the derived Hep-iPS cells used for the microarray studies.
Figure 2: Multiple cell types undergo extensive transcriptional reprogramming to the human iPS cell state.
Figure 3: iPS cells retain a transcriptional memory of the original somatic cell.
Figure 4: DNA methylation can partially explain somatic gene expression in iPS cells.
Figure 5: Meta-analysis of DNA-methylation-associated transcriptional memory in independent data sets.
Figure 6: The somatic cell memory gene C9orf64 is required for efficient generation of iPS cells.
Figure 7: Proximity in the genome affects efficiency of gene silencing in iPS cells.
Figure 8: Model for the role of DNA methylation in reprogramming to the human iPS cell state.

Similar content being viewed by others

References

  1. Yamanaka, S. A fresh look at iPS cells. Cell 137, 13–17 (2009).

    Article  CAS  Google Scholar 

  2. Feng, Q. et al. Hemangioblastic derivatives from human induced pluripotent stem cells exhibit limited expansion and early senescence. Stem Cells 28, 704–712 (2010).

    Article  Google Scholar 

  3. Hu, B. Y. et al. Neural differentiation of human induced pluripotent stem cells follows developmental principles but with variable potency. Proc. Natl Acad. Sci. USA 107, 4335–4340 (2010).

    Article  CAS  Google Scholar 

  4. Chin, M. H. et al. Induced pluripotent stem cells and embryonic stem cells are distinguished by gene expression signatures. Cell Stem Cell 5, 111–123 (2009).

    Article  CAS  Google Scholar 

  5. Ghosh, Z. et al. Persistent donor cell gene expression among human induced pluripotent stem cells contributes to differences with human embryonic stem cells. PLoS One 5, e8975 (2010).

    Article  Google Scholar 

  6. Marchetto, M. C. et al. Transcriptional signature and memory retention of human-induced pluripotent stem cells. PLoS One 4, e7076 (2009).

    Article  Google Scholar 

  7. Guenther, M. G. et al. Chromatin structure and gene expression programs of human embryonic and induced pluripotent stem cells. Cell Stem Cell 7, 249–257 (2010).

    Article  CAS  Google Scholar 

  8. Newman, A. M. & Cooper, J.B. Lab-specific gene expression signatures in pluripotent stem cells. Cell Stem Cell 7, 258–262 (2010).

    Article  CAS  Google Scholar 

  9. Bock, C. et al. Reference maps of human ES and iPS cell variation enable high-throughput characterization of pluripotent cell lines. Cell 144, 439–452 (2011).

    Article  CAS  Google Scholar 

  10. Deng, J. et al. Targeted bisulfite sequencing reveals changes in DNA methylation associated with nuclear reprogramming. Nat. Biotechnol. 27, 353–360 (2009).

    Article  CAS  Google Scholar 

  11. Doi, A. et al. Differential methylation of tissue- and cancer-specific CpG island shores distinguishes human induced pluripotent stem cells, embryonic stem cells and fibroblasts. Nat. Genet. 41, 1350–1353 (2009).

    Article  CAS  Google Scholar 

  12. Kim, K. et al. Epigenetic memory in induced pluripotent stem cells. Nature 467, 285–290 (2010).

    Article  CAS  Google Scholar 

  13. Lister, R. et al. Hotspots of aberrant epigenomic reprogramming in human induced pluripotent stem cells. Nature 471, 68–73 (2011).

    Article  CAS  Google Scholar 

  14. Polo, J. M. et al. Cell type of origin influences the molecular and functional properties of mouse induced pluripotent stem cells. Nat. Biotechnol. 28, 848–855 (2010).

    Article  CAS  Google Scholar 

  15. Hockemeyer, D. et al. A drug-inducible system for direct reprogramming of human somatic cells to pluripotency. Cell Stem Cell 3, 346–353 (2008).

    Article  CAS  Google Scholar 

  16. Maherali, N. et al. A high-efficiency system for the generation and study of human induced pluripotent stem cells. Cell Stem Cell 3, 340–345 (2008).

    Article  CAS  Google Scholar 

  17. Utikal, J., Maherali, N., Kulalert, W. & Hochedlinger, K. Sox2 is dispensable for the reprogramming of melanocytes and melanoma cells into induced pluripotent stem cells. J. Cell Sci. 122, 3502–3510 (2009).

    Article  CAS  Google Scholar 

  18. Yang, Y. H., Xiao, Y. & Segal, M. R. Identifying differentially expressed genes from microarray experiments via statistic synthesis. Bioinformatics 21, 1084–1093 (2005).

    Article  CAS  Google Scholar 

  19. Hemesath, T. J. et al. Microphthalmia, a critical factor in melanocyte development, defines a discrete transcription factor family. Genes Dev. 8, 2770–2780 (1994).

    Article  CAS  Google Scholar 

  20. Odom, D. T. et al. Control of pancreas and liver gene expression by HNF transcription factors. Science 303, 1378–1381 (2004).

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  22. Irizarry, R. A. et al. Comprehensive high-throughput arrays for relative methylation (CHARM). Genome Res. 18, 780–790 (2008).

    Article  CAS  Google Scholar 

  23. Avery, K., Avery, S., Shepherd, J., Heath, P. R. & Moore, H. Sphingosine-1-phosphate mediates transcriptional regulation of key targets associated with survival, proliferation, and pluripotency in human embryonic stem cells. Stem. Cells Dev. 17, 1195–1205 (2008).

    Article  CAS  Google Scholar 

  24. Baker, D. E. et al. Adaptation to culture of human embryonic stem cells and oncogenesis in vivo. Nat. Biotechnol. 25, 207–215 (2007).

    Article  CAS  Google Scholar 

  25. Li, S. S. et al. Target identification of microRNAs expressed highly in human embryonic stem cells. J. Cell Biochem. 106, 1020–1030 (2009).

    Article  CAS  Google Scholar 

  26. Lowry, W. E. et al. Generation of human induced pluripotent stem cells from dermal fibroblasts. Proc. Natl Acad. Sci. USA 105, 2883–2888 (2008).

    Article  CAS  Google Scholar 

  27. Soldner, F. et al. Parkinson’s disease patient-derived induced pluripotent stem cells free of viral reprogramming factors. Cell 136, 964–977 (2009).

    Article  CAS  Google Scholar 

  28. Yu, J. et al. Human induced pluripotent stem cells free of vector and transgene sequences. Science 324, 797–801 (2009).

    Article  CAS  Google Scholar 

  29. Warren, L. et al. Highly efficient reprogramming to pluripotency and directed differentiation of human cells with synthetic modified mRNA. Cell Stem Cell 7, 618–630 (2010).

    Article  CAS  Google Scholar 

  30. Campain, A. & Yang, Y. H. Comparison study of microarray meta-analysis methods. BMC Bioinform. 11, 408–418 (2010).

    Article  Google Scholar 

  31. Mikkelsen, T. S. et al. Dissecting direct reprogramming through integrative genomic analysis. Nature 454, 49–55 (2008).

    Article  CAS  Google Scholar 

  32. Hanna, J. et al. Direct cell reprogramming is a stochastic process amenable to acceleration. Nature 462, 595–601 (2009).

    Article  CAS  Google Scholar 

  33. Meissner, A., Wernig, M. & Jaenisch, R. Direct reprogramming of genetically unmodified fibroblasts into pluripotent stem cells. Nat. Biotechnol. 25, 1177–1181 (2007).

    Article  CAS  Google Scholar 

  34. Chapman, E. J., Kelly, G. & Knowles, M. A. Genes involved in differentiation, stem cell renewal, and tumorigenesis are modulated in telomerase-immortalized human urothelial cells. Mol. Cancer Res. 6, 1154–1168 (2008).

    Article  CAS  Google Scholar 

  35. Jung, Y., Park, J., Bang, Y. J. & Kim, T. Y. Gene silencing of TSPYL5 mediated by aberrant promoter methylation in gastric cancers. Lab. Invest. 88, 153–160 (2008).

    Article  CAS  Google Scholar 

  36. Kim, T. Y., Zhong, S., Fields, C. R., Kim, J. H. & Robertson, K. D. Epigenomic profiling reveals novel and frequent targets of aberrant DNA methylation-mediated silencing in malignant glioma. Cancer Res. 66, 7490–7501 (2006).

    Article  CAS  Google Scholar 

  37. Sweetser, D. A. et al. Delineation of the minimal commonly deleted segment and identification of candidate tumour-suppressor genes in del(9q) acute myeloid leukemia. Genes Chromosomes Cancer 44, 279–291 (2005).

    Article  CAS  Google Scholar 

  38. Cai, L. Y. et al. Identification of PRTFDC1 silencing and aberrant promoter methylation of GPR150, ITGA8 and HOXD11 in ovarian cancers. Life Sci. 80, 1458–1465 (2007).

    Article  CAS  Google Scholar 

  39. Hirasawa, Y. et al. Methylation status of genes upregulated by demethylating agent 5-aza-2’-deoxycytidine in hepatocellular carcinoma. Oncology 71, 77–85 (2006).

    Article  CAS  Google Scholar 

  40. Kroon, E. et al. Pancreatic endoderm derived from human embryonic stem cells generates glucose-responsive insulin-secreting cells in vivo. Nat. Biotechnol. 26, 443–452 (2008).

    Article  CAS  Google Scholar 

  41. Dai, M. H. et al. Evolving gene/transcript definitions significantly alter the interpretation of GeneChip data. Nucleic Acids Res. 33, E175 (2005).

    Article  Google Scholar 

  42. Irizarry, R. A. et al. Exploration, normalization, and summaries of high density oligonucleotide array probe level data. Biostatistics 4, 249–264 (2003).

    Article  Google Scholar 

  43. Johnson, W. E., Li, C. & Rabinovic, A. Adjusting batch effects in microarray expression data using empirical Bayes methods. Biostatistics 8, 118–127 (2007).

    Article  Google Scholar 

  44. Grunau, C., Clark, S. J. & Rosenthal, A. Bisulfite genomic sequencing: systematic investigation of critical experimental parameters. Nucleic Acids Res. 29, E65 (2001).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

The authors wish to thank S. Fisher, O. Genbachev, A. Leavitt and B. Conklin for expert advice on culturing human ES cells, D. Subramanyam and R. Blelloch for the Adult Fibroblast-iPS 1 cell line, L. Ta, A. Williams and A. Holloway at the Gladstone Institutes, J. Bolen at the Mouse Pathology Core Facility for expert assistance, J. Utikal for technical advice, and J. Yang and A. Campain for sharing their meta-DEDS code. We thank members of the Santos laboratory, R. Blelloch, H. Willenbring, S. Fisher and M. Grskovic for helpful discussions and critical reading of the manuscript. Work in the Santos laboratory is supported by CIRM, JDRF, an NIH Director’s New Innovator Award and the Leona M. and Harry B. Helmsley Charitable Trust. Y.O. was partially supported by the UCSF Diabetes Center and a T32 grant from the NICHD to the UCSF Center for Reproductive Sciences. Work in M.H.’s laboratory was supported by grants from the JDRF and the Leona M. and Harry B. Helmsley Charitable Trust. T.G. was supported by the JDRF and the Leona M. and Harry B. Helmsley Charitable Trust. S.L.D. was partially supported by CIRM. J.S.S. was partially supported by the PhRMA Foundation.

Author information

Authors and Affiliations

Authors

Contributions

Y.O., J.S.S. and M.R-S. conceived the project. J.M.P., K.H., P.D.M. and D.J.R. provided reagents. Z.Q. and J.Y. provided assistance with data analysis. C.H. and S.L.D. carried out the bisulphite sequencing analysis under supervision of J.F.C. T.G. carried out the targeted differentiation to endoderm analysis under supervision of M.H. J.S.S. carried out all of the bioinformatic analyses. Y.O., H.Q. and M.R-S. designed and Y.O. and H.Q. carried out all other experiments with technical assistance from L.B. Y.O, J.S.S. and M.R-S. wrote the manuscript with input from the other authors.

Corresponding authors

Correspondence to Jun S. Song or Miguel Ramalho-Santos.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Supplementary information

Supplementary Information

Supplementary Information (PDF 1694 kb)

Supplementary Information

Supplementary Information (XLS 340 kb)

Supplementary Information

Supplementary Information (XLS 374 kb)

Rights and permissions

Reprints and permissions

About this article

Cite this article

Ohi, Y., Qin, H., Hong, C. et al. Incomplete DNA methylation underlies a transcriptional memory of somatic cells in human iPS cells. Nat Cell Biol 13, 541–549 (2011). https://doi.org/10.1038/ncb2239

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/ncb2239

This article is cited by

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