Postnatal loss of Dlk1 imprinting in stem cells and niche astrocytes regulates neurogenesis (original) (raw)

Nature volume 475, pages 381–385 (2011)Cite this article

Subjects

Abstract

The gene for the atypical NOTCH ligand delta-like homologue 1 (Dlk1) encodes membrane-bound and secreted isoforms that function in several developmental processes in vitro and in vivo. Dlk1, a member of a cluster of imprinted genes, is expressed from the paternally inherited chromosome1,2. Here we show that mice that are deficient in Dlk1 have defects in postnatal neurogenesis in the subventricular zone: a developmental continuum that results in depletion of mature neurons in the olfactory bulb. We show that DLK1 is secreted by niche astrocytes, whereas its membrane-bound isoform is present in neural stem cells (NSCs) and is required for the inductive effect of secreted DLK1 on self-renewal. Notably, we find that there is a requirement for Dlk1 to be expressed from both maternally and paternally inherited chromosomes. Selective absence of Dlk1 imprinting in both NSCs and niche astrocytes is associated with postnatal acquisition of DNA methylation at the germ-line-derived imprinting control region. The results emphasize molecular relationships between NSCs and the niche astrocyte cells of the microenvironment, identifying a signalling system encoded by a single gene that functions coordinately in both cell types. The modulation of genomic imprinting in a stem-cell environment adds a new level of epigenetic regulation to the establishment and maintenance of the niche, raising wider questions about the adaptability, function and evolution of imprinting in specific developmental contexts.

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

Access options

Subscribe to this journal

Receive 51 print issues and online access

$199.00 per year

only $3.90 per issue

Buy this article

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

Additional access options:

Similar content being viewed by others

References

  1. Schmidt, J. V., Matteson, P. G., Jones, B. K., Guan, X. J. & Tilghman, S. M. The Dlk1 and Gtl2 genes are linked and reciprocally imprinted. Genes Dev. 14, 1997–2002 (2000)
    CAS PubMed PubMed Central Google Scholar
  2. Takada, S. et al. Delta-like and Gtl2 are reciprocally expressed, differentially methylated linked imprinted genes on mouse chromosome 12. Curr. Biol. 10, 1135–1138 (2000)
    Article CAS PubMed Google Scholar
  3. Riquelme, P. A., Drapeau, E. & Doetsch, F. Brain micro-ecologies: neural stem cell niches in the adult mammalian brain. Phil. Trans. R. Soc. B 363, 123–137 (2008)
    Article PubMed Google Scholar
  4. Ma, D. K., Bonaguidi, M. A., Ming, G. L. & Song, H. Adult neural stem cells in the mammalian central nervous system. Cell Res. 19, 672–682 (2009)
    Article CAS PubMed Google Scholar
  5. Lledo, P. M., Merkle, F. T. & Alvarez-Buylla, A. Origin and function of olfactory bulb interneuron diversity. Trends Neurosci. 31, 392–400 (2008)
    Article CAS PubMed PubMed Central Google Scholar
  6. Song, H., Stevens, C. F. & Gage, F. H. Astroglia induce neurogenesis from adult neural stem cells. Nature 417, 39–44 (2002)
    Article ADS CAS PubMed Google Scholar
  7. Doetsch, F. The glial identity of neural stem cells. Nature Neurosci. 6, 1127–1134 (2003)
    Article CAS PubMed Google Scholar
  8. Környei, Z. et al. Humoral and contact interactions in astroglia/stem cell co-cultures in the course of glia-induced neurogenesis. Glia 49, 430–444 (2005)
    Article PubMed Google Scholar
  9. Moon, Y. S. et al. Mice lacking paternally expressed Pref-1/Dlk1 display growth retardation and accelerated adiposity. Mol. Cell. Biol. 22, 5585–5592 (2002)
    Article CAS PubMed PubMed Central Google Scholar
  10. Abdallah, B. M. et al. Regulation of human skeletal stem cells differentiation by Dlk1/Pref-1. J. Bone Miner. Res. 19, 841–852 (2004)
    Article CAS PubMed Google Scholar
  11. Raghunandan, R. et al. Dlk1 influences differentiation and function of β lymphocytes. Stem Cells Dev. 17, 495–508 (2008)
    Article CAS PubMed PubMed Central Google Scholar
  12. Davis, E. et al. Ectopic expression of DLK1 protein in skeletal muscle of padumnal heterozygotes causes the callipyge phenotype. Curr. Biol. 14, 1858–1862 (2004)
    Article CAS PubMed Google Scholar
  13. Christophersen, N. S. et al. Midbrain expression of Delta-like 1 homologue is regulated by GDNF and is associated with dopaminergic differentiation. Exp. Neurol. 204, 791–801 (2007)
    Article CAS PubMed Google Scholar
  14. Bauer, M. et al. Delta-like 1 participates in the specification of ventral midbrain progenitor derived dopaminergic neurons. J. Neurochem. 104, 1101–1115 (2008)
    Article CAS PubMed Google Scholar
  15. da Rocha, S. T. et al. Gene dosage effects of the imprinted delta-like homologue 1 (Dlk1/Pref1) in development: implications for the evolution of imprinting. PLoS Genet. 5, e1000392 (2009)
    Article PubMed Google Scholar
  16. Li, L., Forman, S. J. & Bhatia, R. Expression of DLK1 in hematopoietic cells results in inhibition of differentiation and proliferation. Oncogene 24, 4472–4476 (2005)
    Article CAS PubMed Google Scholar
  17. Tramontin, A. D., García-Verdugo, J. M., Lim, D. A. & Alvarez-Buylla, A. Postnatal development of radial glia and the ventricular zone (VZ): a continuum of the neural stem cell compartment. Cereb. Cortex 13, 580–587 (2003)
    Article PubMed Google Scholar
  18. Ferrón, S. R. et al. A combined ex/in vivo assay to detect effects of exogenously added factors in neural stem cells. Nature Protocols 2, 849–859 (2007)
    Article PubMed Google Scholar
  19. Kippin, T. E., Martens, D. J. & van der Kooy, D. p21 loss compromises the relative quiescence of forebrain stem cell proliferation leading to exhaustion of their proliferation capacity. Genes Dev. 19, 756–767 (2005)
    Article CAS PubMed PubMed Central Google Scholar
  20. Paik, J. H. et al. FoxOs cooperatively regulate diverse pathways governing neural stem cell homeostasis. Cell Stem Cell 5, 540–553 (2009)
    Article CAS PubMed PubMed Central Google Scholar
  21. Bray, S. J., Takada, S., Harrison, E., Shen, S. C. & Ferguson-Smith, A. C. The atypical mammalian ligand Delta-like homologue 1 (Dlk1) can regulate Notch signalling in Drosophila . BMC Dev. Biol. 8, 11 (2008)
    Article PubMed PubMed Central Google Scholar
  22. Nyfeler, Y. et al. Jagged1 signals in the postnatal subventricular zone are required for neural stem cell self-renewal. EMBO J. 24, 3504–3515 (2005)
    Article CAS PubMed PubMed Central Google Scholar
  23. Andreu-Agulló, C., Morante-Redolat, J. M., Delgado, A. C. & Farinas, I. Vascular niche factor PEDF modulates Notch-dependent stemness in the adult subependymal zone. Nature Neurosci. 12, 1514–1523 (2009)
    Article PubMed Google Scholar
  24. Takada, S. et al. Epigenetic analysis of the Dlk1_–_Gtl2 imprinted domain on mouse chromosome 12: implications for imprinting control from comparison with Igf2_–_H19 . Hum. Mol. Genet. 11, 77–86 (2002)
    Article CAS PubMed Google Scholar
  25. Lin, S. et al. Asymmetric regulation of imprinting on the maternal and paternal chromosomes at the Dlk1_–_Gtl2 imprinted cluster on mouse chromosome 12. Nature Genet. 35, 97–102 (2003)
    Article CAS PubMed Google Scholar
  26. Li, X. et al. A maternal-zygotic effect gene, Zfp57, maintains both maternal and paternal imprints. Dev. Cell 15, 547–557 (2008)
    Article CAS PubMed PubMed Central Google Scholar
  27. Mirzadeh, Z. et al. The subventricular zone en-face: wholemount staining and ependymal flow. J Vis Exp. 39 1938 10.3791/1938 (2010)
    Article Google Scholar
  28. Lie, D. C. et al. Wnt signalling regulates adult hippocampal neurogenesis. Nature 437, 1370–1375 (2005)
    Article ADS CAS PubMed Google Scholar
  29. García-Marqués, J., De Carlos, J. A., Greer, C. A. & López-Mascaraque, L. Different astroglia permissivity controls the migration of olfactory bulb interneuron precursors. Glia 58, 218–230 (2010)
    Article PubMed Google Scholar
  30. Hsieh, J. J. et al. Truncated mammalian Notch1 activates CBF1/RBPJk-repressed genes by a mechanism resembling that of Epstein-Barr virus EBNA2. Mol. Cell. Biol. 16, 952–959 (1996)
    Article CAS PubMed PubMed Central Google Scholar

Download references

Acknowledgements

We are grateful to J. Herbert and E. B. Keverne for sharing expertise during the course of this work. We thank members of the Ferguson-Smith laboratory for discussions and B. Sun, D. Gray, S. Curran, I. Gutteridge, R. Rancourt and X. d’Anglemont de Tassigny for technical assistance. The work was funded by grants from the Medical Research Council and Wellcome Trust to A.C.F.-S. and by grants from Ministerio de Ciencia e Innovación (SAF2008-01006, CB06/05/0086, RD06/0010/0010) and Generalitat Valenciana (Prometeo) to I.F. S.R.F. is a recipient of a University of Cambridge Herchel-Smith Fellowship.

Author information

Author notes

  1. Marika Charalambous and Elizabeth Radford: These authors contributed equally to this work.

Authors and Affiliations

  1. Department of Physiology, Development & Neuroscience, University of Cambridge, Cambridge CB2 3EG, UK,
    Sacri R. Ferrón, Marika Charalambous, Elizabeth Radford, Kirsten McEwen, Eleanor Hind & Anne C. Ferguson-Smith
  2. Department of Molecular Neurobiology, National Institute for Medical Research, Medical Research Council, London NW7 1AA, UK,
    Hendrik Wildner & Francois Guillemot
  3. Departamento de Biología Celular, Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas, Universidad de Valencia, 46100 Burjassot, Spain,
    Jose Manuel Morante-Redolat & Isabel Fariñas
  4. Department of Inorganic and Organic Chemistry and Biochemistry, Medical School, Regional Center for Biomedical Research, University of Castilla-La Mancha, Avenida de Almansa 14, 02006 Albacete, Spain,
    Jorge Laborda
  5. Division of Cellular and Gene Therapies, Cellular and Tissue Therapies Branch, Center for Biologics Evaluation and Research, Food and Drug Administration, Bethesda, Maryland 20892, USA,
    Steven R. Bauer

Authors

  1. Sacri R. Ferrón
    You can also search for this author inPubMed Google Scholar
  2. Marika Charalambous
    You can also search for this author inPubMed Google Scholar
  3. Elizabeth Radford
    You can also search for this author inPubMed Google Scholar
  4. Kirsten McEwen
    You can also search for this author inPubMed Google Scholar
  5. Hendrik Wildner
    You can also search for this author inPubMed Google Scholar
  6. Eleanor Hind
    You can also search for this author inPubMed Google Scholar
  7. Jose Manuel Morante-Redolat
    You can also search for this author inPubMed Google Scholar
  8. Jorge Laborda
    You can also search for this author inPubMed Google Scholar
  9. Francois Guillemot
    You can also search for this author inPubMed Google Scholar
  10. Steven R. Bauer
    You can also search for this author inPubMed Google Scholar
  11. Isabel Fariñas
    You can also search for this author inPubMed Google Scholar
  12. Anne C. Ferguson-Smith
    You can also search for this author inPubMed Google Scholar

Contributions

S.R.F. conceived and performed experiments and developed the project, coordinated collaborations and wrote the manuscript. M.C. conducted methylation experiments, helped to design experiments, contributed ideas and interpreted results. E.R. performed expression analysis, contributed to ideas and interpreted results. K.M. helped with imprinting and methylation studies. H.W. and F.G. designed and performed embryo studies. E.H. helped with imprinting and NOTCH-expression studies. J.M.M.-R. performed the NOTCH-pathway experiments. J.L. and S.R.B. provided the _Dlk1_−/− mouse. I.F. contributed ideas, discussed results and helped to write the manuscript. A.C.F.-S. conceived and developed the project, designed experiments, interpreted results, coordinated collaborations, contributed ideas and funds and wrote the manuscript.

Corresponding author

Correspondence toAnne C. Ferguson-Smith.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Supplementary information

PowerPoint slides

Rights and permissions

About this article

Cite this article

Ferrón, S., Charalambous, M., Radford, E. et al. Postnatal loss of Dlk1 imprinting in stem cells and niche astrocytes regulates neurogenesis.Nature 475, 381–385 (2011). https://doi.org/10.1038/nature10229

Download citation

This article is cited by

Editorial Summary

Selective loss of imprinting in neurogenesis

Life-long neurogenesis is known to occur in some areas of the mammalian adult brain, including in the subventricular zone (SVZ), where interaction between neural precursor cells and resident astrocytes can produced migratory neuroblasts. Here, Anne Ferguson-Smith and colleagues reveal a differential role for the same developmental gene, Dlk1, in neural precursor cells and astrocytes. The gene product, the Notch ligand DLK1, has two isoforms. One is an inductive niche factor secreted by astrocytes, and the other is a membrane-bound isoform required by the neural stem cells themselves to respond to secreted DLK1. Selective changes of imprinting with age modulate the role of Dlk1 in each cell type in mice, regulating neurogenesis when establishing the adult neurogenic niche. The modulation of genomic imprinting in a stem-cell environment adds a previously unrecognized element to epigenetic regulation.

Associated content