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.

  • Letter
  • Published:

Maternal imprinting at the H19–Igf2 locus maintains adult haematopoietic stem cell quiescence

Abstract

The epigenetic regulation of imprinted genes by monoallelic DNA methylation of either maternal or paternal alleles is critical for embryonic growth and development1. Imprinted genes were recently shown to be expressed in mammalian adult stem cells to support self-renewal of neural and lung stem cells2,3,4; however, a role for imprinting per se in adult stem cells remains elusive. Here we show upregulation of growth-restricting imprinted genes, including in the H19–Igf2 locus5, in long-term haematopoietic stem cells and their downregulation upon haematopoietic stem cell activation and proliferation. A differentially methylated region upstream of H19 (H19-DMR), serving as the imprinting control region, determines the reciprocal expression of H19 from the maternal allele and Igf2 from the paternal allele1. In addition, H19 serves as a source of miR-675, which restricts Igf1r expression6. We demonstrate that conditional deletion of the maternal but not the paternal H19-DMR reduces adult haematopoietic stem cell quiescence, a state required for long-term maintenance of haematopoietic stem cells, and compromises haematopoietic stem cell function. Maternal-specific H19-DMR deletion results in activation of the Igf2–Igfr1 pathway, as shown by the translocation of phosphorylated FoxO3 (an inactive form) from nucleus to cytoplasm and the release of FoxO3-mediated cell cycle arrest, thus leading to increased activation, proliferation and eventual exhaustion of haematopoietic stem cells. Mechanistically, maternal-specific H19-DMR deletion leads to Igf2 upregulation and increased translation of Igf1r, which is normally suppressed by H19-derived miR-675. Similarly, genetic inactivation of Igf1r partly rescues the H19-DMR deletion phenotype. Our work establishes a new role for this unique form of epigenetic control at the H19–Igf2 locus in maintaining adult stem cells.

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: Defective LT-HSCs in mH19ΔDMR/+ mice.
Figure 2: Compromised HSC function in mH19ΔDMR/+ mice.
Figure 3: Activation of Igf2–FoxO3 signalling in mH19ΔDMR/+ mice.
Figure 4: Igf1r regulation and rescue by genetic blockage of Igf2–Igf1r signalling.

Similar content being viewed by others

Accession codes

Accessions

ArrayExpress

Data deposits

The microarray and RNA-seq data have been deposited in ArrayExpress under accession numbers E-MTAB-1644 and E-MTAB-1628, respectively.

References

  1. Bartolomei, M. S. Genomic imprinting: employing and avoiding epigenetic processes. Genes Dev. 23, 2124–2133 (2009)

    Article  CAS  Google Scholar 

  2. Berg, J. S. et al. Imprinted genes that regulate early mammalian growth are coexpressed in somatic stem cells. PLoS ONE 6, e26410 (2011)

    Article  ADS  CAS  Google Scholar 

  3. Ferrón, S. R. et al. Postnatal loss of Dlk1 imprinting in stem cells and niche astrocytes regulates neurogenesis. Nature 475, 381–385 (2011)

    Article  Google Scholar 

  4. Zacharek, S. J. et al. Lung stem cell self-renewal relies on BMI1-dependent control of expression at imprinted loci. Cell Stem Cell 9, 272–281 (2011)

    Article  CAS  Google Scholar 

  5. DeChiara, T. M., Robertson, E. J. & Efstratiadis, A. Parental imprinting of the mouse insulin-like growth factor II gene. Cell 64, 849–859 (1991)

    Article  CAS  Google Scholar 

  6. Keniry, A. et al. The H19 lincRNA is a developmental reservoir of miR-675 that suppresses growth and Igf1r. Nature Cell Biol. 14, 659–665 (2012)

    Article  CAS  Google Scholar 

  7. Haug, J. S. et al. N-cadherin expression level distinguishes reserved versus primed states of hematopoietic stem cells. Cell Stem Cell 2, 367–379 (2008)

    Article  CAS  Google Scholar 

  8. Yang, L. et al. Identification of LinSca1+kit+CD34+Flt3- short-term hematopoietic stem cells capable of rapidly reconstituting and rescuing myeloablated transplant recipients. Blood 105, 2717–2723 (2005)

    Article  CAS  Google Scholar 

  9. Frost, J. M. & Moore, G. E. The importance of imprinting in the human placenta. PLoS Genet. 6, e1001015 (2010)

    Article  Google Scholar 

  10. Hudson, Q. J., Kulinski, T. M., Huetter, S. P. & Barlow, D. P. Genomic imprinting mechanisms in embryonic and extraembryonic mouse tissues. Heredity 105, 45–56 (2010)

    Article  CAS  Google Scholar 

  11. Thorvaldsen, J. L., Fedoriw, A. M., Nguyen, S. & Bartolomei, M. S. Developmental profile of H19 differentially methylated domain (DMD) deletion alleles reveals multiple roles of the DMD in regulating allelic expression and DNA methylation at the imprinted H19/Igf2 locus. Mol. Cell. Biol. 26, 1245–1258 (2006)

    Article  CAS  Google Scholar 

  12. Lerner, C. & Harrison, D. E. 5-Fluorouracil spares hemopoietic stem cells responsible for long-term repopulation. Exp. Hematol. 18, 114–118 (1990)

    CAS  PubMed  Google Scholar 

  13. Smith, F. M., Garfield, A. S. & Ward, A. Regulation of growth and metabolism by imprinted genes. Cytogenet. Genome Res. 113, 279–291 (2006)

    Article  CAS  Google Scholar 

  14. Kang, H. M., Park, S. & Kim, H. Insulin-like growth factor 2 enhances insulinogenic differentiation of human eyelid adipose stem cells via the insulin receptor. Cell Prolif. 44, 254–263 (2011)

    Article  CAS  Google Scholar 

  15. Zhang, J. et al. PTEN maintains haematopoietic stem cells and acts in lineage choice and leukaemia prevention. Nature 441, 518–522 (2006)

    Article  ADS  CAS  Google Scholar 

  16. Tothova, Z. et al. FoxOs are critical mediators of hematopoietic stem cell resistance to physiologic oxidative stress. Cell 128, 325–339 (2007)

    Article  CAS  Google Scholar 

  17. Klinakis, A. et al. Igf1r as a therapeutic target in a mouse model of basal-like breast cancer. Proc. Natl Acad. Sci. USA 106, 2359–2364 (2009)

    Article  ADS  CAS  Google Scholar 

  18. Zou, P. et al. p57(Kip2) and p27(Kip1) cooperate to maintain hematopoietic stem cell quiescence through interactions with Hsc70. Cell Stem Cell 9, 247–261 (2011)

    Article  CAS  Google Scholar 

  19. Walkley, C. R., Shea, J. M., Sims, N. A., Purton, L. E. & Orkin, S. H. Rb regulates interactions between hematopoietic stem cells and their bone marrow microenvironment. Cell 129, 1081–1095 (2007)

    Article  CAS  Google Scholar 

  20. Kubota, Y., Osawa, M., Jakt, L. M., Yoshikawa, K. & Nishikawa, S. Necdin restricts proliferation of hematopoietic stem cells during hematopoietic regeneration. Blood 114, 4383–4392 (2009)

    Article  CAS  Google Scholar 

  21. Stadtfeld, M. et al. Aberrant silencing of imprinted genes on chromosome 12qF1 in mouse induced pluripotent stem cells. Nature 465, 175–181 (2010)

    Article  ADS  CAS  Google Scholar 

  22. Zhao, Z. et al. Circular chromosome conformation capture (4C) uncovers extensive networks of epigenetically regulated intra- and interchromosomal interactions. Nature Genet. 38, 1341–1347 (2006)

    Article  CAS  Google Scholar 

  23. Varrault, A. et al. Zac1 regulates an imprinted gene network critically involved in the control of embryonic growth. Dev. Cell 11, 711–722 (2006)

    Article  CAS  Google Scholar 

  24. Karlsson, G. et al. Smad4 is critical for self-renewal of hematopoietic stem cells. J. Exp. Med. 204, 467–474 (2007)

    Article  CAS  Google Scholar 

  25. Sugimura, R. et al. Noncanonical wnt signaling maintains hematopoietic stem cells in the niche. Cell 150, 351–365 (2012)

    Article  CAS  Google Scholar 

  26. Perry, J. M. et al. Cooperation between both Wnt/β-catenin and PTEN/PI3K/Akt signaling promotes primitive hematopoietic stem cell self-renewal and expansion. Genes Dev. 25, 1928–1942 (2011)

    Article  CAS  Google Scholar 

  27. Dijke, P. & Heldin, C. H. Smad Signal Transduction: Smads in Proliferation, Differentiation and Disease (Springer, 2006)

    Book  Google Scholar 

  28. Johnson, C. D. et al. The let-7 microRNA represses cell proliferation pathways in human cells. Cancer Res. 67, 7713–7722 (2007)

    Article  CAS  Google Scholar 

  29. Toledano, H., D’Alterio, C., Czech, B., Levine, E. & Jones, D. L. The let-7-Imp axis regulates ageing of the Drosophila testis stem-cell niche. Nature 485, 605–610 (2012)

    Article  ADS  CAS  Google Scholar 

  30. Yu, W. et al. A novel amplification target, DUSP26, promotes anaplastic thyroid cancer cell growth by inhibiting p38 MAPK activity. Oncogene 26, 1178–1187 (2007)

    Article  CAS  Google Scholar 

  31. Thorvaldsen, J. L., Fedoriw, A. M., Nguyen, S. & Bartolomei, M. S. Developmental profile of H19 differentially methylated domain (DMD) deletion alleles reveals multiple roles of the DMD in regulating allelic expression and DNA methylation at the imprinted H19/Igf2 locus. Mol. Cell. Biol. 26, 1245–1258 (2006)

    Article  CAS  Google Scholar 

  32. Dietrich, P., Dragatsis, I., Xuan, S., Zeitlin, S. & Efstratiadis, A. Conditional mutagenesis in mice with heat shock promoter-driven cre transgenes. Mamm. Genome 11, 196–205 (2000)

    Article  CAS  Google Scholar 

  33. Zhang, J. et al. Identification of the haematopoietic stem cell niche and control of the niche size. Nature 425, 836–841 (2003)

    Article  ADS  CAS  Google Scholar 

  34. Haug, J. S. et al. N-cadherin expression level distinguishes reserved versus primed states of hematopoietic stem cells. Cell Stem Cell 2, 367–379 (2008)

    Article  CAS  Google Scholar 

  35. Miller, C. L. & Eaves, C. J. Expansion in vitro of adult murine hematopoietic stem cells with transplantable lympho-myeloid reconstituting ability. Proc. Natl Acad. Sci. USA 94, 13648–13653 (1997)

    Article  ADS  CAS  Google Scholar 

  36. Akashi, K. et al. Transcriptional accessibility for genes of multiple tissues and hematopoietic lineages is hierarchically controlled during early hematopoiesis. Blood 101, 383–389 (2003)

    Article  CAS  Google Scholar 

  37. Bolstad, B. M., Irizarry, R. A., Astrand, M. & Speed, T. P. A comparison of normalization methods for high density oligonucleotide array data based on variance and bias. Bioinformatics 19, 185–193 (2003)

    Article  CAS  Google Scholar 

  38. 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 

  39. Irizarry, R. A. et al. Summaries of Affymetrix GeneChip probe level data. Nucleic Acids Res. 31, e15 (2003)

    Article  Google Scholar 

  40. Smyth, G. K. Linear models and empirical bayes methods for assessing differential expression in microarray experiments. Stat. Appl. Genet. Mol. Biol. 3, Article3 (2004)

    Article  MathSciNet  Google Scholar 

  41. Ema, H. et al. Adult mouse hematopoietic stem cells: purification and single-cell assays. Nature Protocols 1, 2979–2987 (2006)

    Article  CAS  Google Scholar 

  42. Trapnell, C., Pachter, L. & Salzberg, S. L. TopHat: discovering splice junctions with RNA-Seq. Bioinformatics 25, 1105–1111 (2009)

    Article  CAS  Google Scholar 

  43. 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  Google Scholar 

  44. Trapnell, C. et al. Transcript assembly and quantification by RNA-Seq reveals unannotated transcripts and isoform switching during cell differentiation. Nature Biotechnol. 28, 511–515 (2010)

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank M. Hembree, T. Johnson, H. Marshall, B. Lewis, D. Dukes, C. Semerad, J. Park and A. Box for technical support, and members of the Li laboratory for scientific discussion. We thank J. Lu and Y. Huang for communications about miR-675 and Let-7. We thank K. Tannen for editing. This work was supported by the Stowers Institute for Medical Research and by the Department of Biotechnology, Ministry of Science and Technology, Government of India, as an overseas associateship to A. Venkatraman. M. Bartolomei is supported by the National Institutes of Health (GM51279).

Author information

Authors and Affiliations

Authors

Contributions

A.V. performed experiments, analysed data and wrote the manuscript. X.H. provided training, performed transplantations and RNA-seq. F.T., J.T., M.C., L.P., X.Z., A.P., H.L., J.P. M.Z., J.H. and T.C. performed part of the experiments. M.B. contributed the mouse lines. L.L. directed the overall project and co-wrote the manuscript. All authors contributed to reading and editing the manuscript.

Corresponding author

Correspondence to Linheng Li.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Supplementary information

Supplementary Figures

This file contains Supplementary Figures 1-7. (PDF 869 kb)

PowerPoint slides

Rights and permissions

Reprints and permissions

About this article

Cite this article

Venkatraman, A., He, X., Thorvaldsen, J. et al. Maternal imprinting at the H19–Igf2 locus maintains adult haematopoietic stem cell quiescence. Nature 500, 345–349 (2013). https://doi.org/10.1038/nature12303

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nature12303

This article is cited by

Comments

By submitting a comment you agree to abide by our Terms and Community Guidelines. If you find something abusive or that does not comply with our terms or guidelines please flag it as inappropriate.

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