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:

HMGA regulates the global chromatin state and neurogenic potential in neocortical precursor cells

Abstract

Neural precursor cells (NPCs) in the mouse neocortex generate various neuronal and glial cell types in a developmental stage–dependent manner. Most NPCs lose their neurogenic potential during development, although the underlying mechanisms of this process are not fully understood. We found that the chromatin of mouse NPCs gradually becomes more condensed and less dynamic on a global scale during neocortical development. Furthermore, we found high mobility group A (HMGA) proteins to be essential for the open chromatin state of NPCs at early developmental stages. Knockdown of HMGA proteins in early-stage NPCs reduced their neurogenic potential. Conversely, overexpression of HMGA proteins conferred neurogenic potential on late-stage NPCs, an effect that was antagonized by coexpression of a histone H1 mutant that inhibits chromatin opening. Thus, HMGA proteins contribute to the neurogenic potential of NPCs in the early stages of neocortical development, possibly through induction of an open chromatin state.

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: Chromatin state in NPCs becomes condensed globally during mouse neocortical development.
Figure 2: HMGA proteins are important for global opening of the chromatin state in early-stage NPCs.
Figure 3: HMGA proteins are essential for the neurogenic potential of early-stage NPCs in vitro.
Figure 4: HMGA proteins are important for the neurogenic potential of NPCs in vivo.
Figure 5: HMGA proteins inhibit astrocyte differentiation of early-stage NPCs in vivo.
Figure 6: Expression of H1cc inhibits the promotion of neuronal differentiation by HMGA overexpression in vivo.

Similar content being viewed by others

Accession codes

Accessions

NCBI Reference Sequence

References

  1. Meshorer, E. et al. Hyperdynamic plasticity of chromatin proteins in pluripotent embryonic stem cells. Dev. Cell 10, 105–116 (2006).

    Article  CAS  Google Scholar 

  2. Gaspar-Maia, A. et al. Chd1 regulates open chromatin and pluripotency of embryonic stem cells. Nature 460, 863–868 (2009).

    Article  CAS  Google Scholar 

  3. Hajkova, P. et al. Chromatin dynamics during epigenetic reprogramming in the mouse germ line. Nature 452, 877–881 (2008).

    Article  CAS  Google Scholar 

  4. Seki, Y. et al. Extensive and orderly reprogramming of genome-wide chromatin modifications associated with specification and early development of germ cells in mice. Dev. Biol. 278, 440–458 (2005).

    Article  CAS  Google Scholar 

  5. Hirabayashi, Y. & Gotoh, Y. Stage-dependent fate determination of neural precursor cells in mouse forebrain. Neurosci. Res. 51, 331–336 (2005).

    Article  CAS  Google Scholar 

  6. Miller, F.D. & Gauthier, A.S. Timing is everything: making neurons versus glia in the developing cortex. Neuron 54, 357–369 (2007).

    Article  CAS  Google Scholar 

  7. Nagao, M. et al. Coordinated control of self-renewal and differentiation of neural stem cells by Myc and the p19ARF-p53 pathway. J. Cell Biol. 183, 1243–1257 (2008).

    Article  CAS  Google Scholar 

  8. Hirabayashi, Y. et al. Polycomb limits the neurogenic competence of neural precursor cells to promote astrogenic fate transition. Neuron 63, 600–613 (2009).

    Article  CAS  Google Scholar 

  9. Naka, H., Nakamura, S., Shimazaki, T. & Okano, H. Requirement for COUP-TFI and II in the temporal specification of neural stem cells in CNS development. Nat. Neurosci. 11, 1014–1023 (2008).

    Article  CAS  Google Scholar 

  10. Sanosaka, T. et al. Identification of genes that restrict astrocyte differentiation of midgestational neural precursor cells. Neuroscience 155, 780–788 (2008).

    Article  CAS  Google Scholar 

  11. Goldberg, A.D., Allis, C.D. & Bernstein, E. Epigenetics: a landscape takes shape. Cell 128, 635–638 (2007).

    Article  CAS  Google Scholar 

  12. Fusco, A. & Fedele, M. Roles of HMGA proteins in cancer. Nat. Rev. Cancer 7, 899–910 (2007).

    Article  CAS  Google Scholar 

  13. Reeves, R. Molecular biology of HMGA proteins: hubs of nuclear function. Gene 277, 63–81 (2001).

    Article  CAS  Google Scholar 

  14. Qian, X. et al. Timing of CNS cell generation: a programmed sequence of neuron and glial cell production from isolated murine cortical stem cells. Neuron 28, 69–80 (2000).

    Article  CAS  Google Scholar 

  15. Sunabori, T. et al. Cell cycle–specific nestin expression coordinates with morphological changes in embryonic cortical neural progenitors. J. Cell Sci. 121, 1204–1212 (2008).

    Article  CAS  Google Scholar 

  16. Waterston, R.H. et al. Initial sequencing and comparative analysis of the mouse genome. Nature 420, 520–562 (2002).

    Article  CAS  Google Scholar 

  17. Catez, F. et al. Network of dynamic interactions between histone H1 and high-mobility-group proteins in chromatin. Mol. Cell. Biol. 24, 4321–4328 (2004).

    Article  CAS  Google Scholar 

  18. Zhao, K., Käs, E., Gonzalez, E. & Laemmli, U.K. SAR-dependent mobilization of histone H1 by HMG-I/Y in vitro: HMG-I/Y is enriched in H1-depleted chromatin. EMBO J. 12, 3237–3247 (1993).

    Article  CAS  Google Scholar 

  19. Nishino, J., Kim, I., Chada, K. & Morrison, S.J. Hmga2 promotes neural stem cell self-renewal in young, but not old, mice by reducing p16Ink4a and p19Arf expression. Cell 135, 227–239 (2008).

    Article  CAS  Google Scholar 

  20. Zhou, X., Benson, K.F., Ashar, H.R. & Chada, K. Mutation responsible for the mouse pygmy phenotype in the developmentally regulated factor HMGI-C. Nature 376, 771–774 (1995).

    Article  CAS  Google Scholar 

  21. Gloster, A. et al. The T alpha 1 alpha-tubulin promoter specifies gene expression as a function of neuronal growth and regeneration in transgenic mice. J. Neurosci. 14, 7319–7330 (1994).

    Article  CAS  Google Scholar 

  22. Sawamoto, K. et al. Direct isolation of committed neuronal progenitor cells from transgenic mice coexpressing spectrally distinct fluorescent proteins regulated by stage-specific neural promoters. J. Neurosci. Res. 65, 220–227 (2001).

    Article  CAS  Google Scholar 

  23. Ramirez-Carrozzi, V.R. et al. A unifying model for the selective regulation of inducible transcription by CpG islands and nucleosome remodeling. Cell 138, 114–128 (2009).

    Article  CAS  Google Scholar 

  24. Hirabayashi, Y. & Gotoh, Y. Epigenetic control of neural precursor cell fate during development. Nat. Rev. Neurosci. 11, 377–388 (2010).

    Article  CAS  Google Scholar 

  25. Frisch, M. et al. In silico prediction of scaffold/matrix attachment regions in large genomic sequences. Genome Res. 12, 349–354 (2002).

    Article  CAS  Google Scholar 

  26. Knoepfler, P.S. et al. Myc influences global chromatin structure. EMBO J. 25, 2723–2734 (2006).

    Article  CAS  Google Scholar 

  27. Takahashi, K. & Yamanaka, S. Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell 126, 663–676 (2006).

    Article  CAS  Google Scholar 

  28. Takahashi, K. et al. Induction of pluripotent stem cells from adult human fibroblasts by defined factors. Cell 131, 861–872 (2007).

    Article  CAS  Google Scholar 

  29. Chang, T.C. et al. Lin-28B transactivation is necessary for Myc-mediated let-7 repression and proliferation. Proc. Natl. Acad. Sci. USA 106, 3384–3389 (2009).

    Article  CAS  Google Scholar 

  30. Yu, J. et al. Induced pluripotent stem cell lines derived from human somatic cells. Science 318, 1917–1920 (2007).

    Article  CAS  Google Scholar 

  31. Heo, I. et al. Lin28 mediates the terminal uridylation of let-7 precursor microRNA. Mol. Cell 32, 276–284 (2008).

    Article  CAS  Google Scholar 

  32. Newman, M.A., Thomson, J.M. & Hammond, S.M. Lin-28 interaction with the Let-7 precursor loop mediates regulated microRNA processing. RNA 14, 1539–1549 (2008).

    Article  CAS  Google Scholar 

  33. Piskounova, E. et al. Determinants of microRNA processing inhibition by the developmentally regulated RNA-binding protein Lin28. J. Biol. Chem. 283, 21310–21314 (2008).

    Article  CAS  Google Scholar 

  34. Rybak, A. et al. A feedback loop comprising lin-28 and let-7 controls pre-let-7 maturation during neural stem-cell commitment. Nat. Cell Biol. 10, 987–993 (2008).

    Article  CAS  Google Scholar 

  35. Viswanathan, S.R., Daley, G.Q. & Gregory, R.I. Selective blockade of microRNA processing by Lin28. Science 320, 97–100 (2008).

    Article  CAS  Google Scholar 

  36. Lee, Y.S. & Dutta, A. The tumor suppressor microRNA let-7 represses the HMGA2 oncogene. Genes Dev. 21, 1025–1030 (2007).

    Article  CAS  Google Scholar 

  37. Mayr, C., Hemann, M.T. & Bartel, D.P. Disrupting the pairing between let-7 and Hmga2 enhances oncogenic transformation. Science 315, 1576–1579 (2007).

    Article  CAS  Google Scholar 

  38. Hirabayashi, Y. et al. The Wnt/beta-catenin pathway directs neuronal differentiation of cortical neural precursor cells. Development 131, 2791–2801 (2004).

    Article  CAS  Google Scholar 

  39. Konno, D. et al. Neuroepithelial progenitors undergo LGN-dependent planar divisions to maintain self-renewability during mammalian neurogenesis. Nat. Cell Biol. 10, 93–101 (2008).

    Article  CAS  Google Scholar 

  40. Miyata, T. & Ogawa, M. Twisting of neocortical progenitor cells underlies a spring-like mechanism for daughter-cell migration. Curr. Biol. 17, 146–151 (2007).

    Article  CAS  Google Scholar 

  41. Ochiai, W. et al. Periventricular notch activation and asymmetric Ngn2 and Tbr2 expression in pair-generated neocortical daughter cells. Mol. Cell. Neurosci. 40, 225–233 (2009).

    Article  CAS  Google Scholar 

  42. Tabata, H. & Nakajima, K. Efficient in utero gene transfer system to the developing mouse brain using electroporation: visualization of neuronal migration in the developing cortex. Neuroscience 103, 865–872 (2001).

    Article  CAS  Google Scholar 

  43. Kamakura, S. et al. Hes binding to STAT3 mediates crosstalk between Notch and JAK-STAT signaling. Nat. Cell Biol. 6, 547–554 (2004).

    Article  CAS  Google Scholar 

  44. Watakabe, A. et al. Comparative analysis of layer-specific genes in Mammalian neocortex. Cereb. Cortex 17, 1918–1933 (2007).

    Article  Google Scholar 

  45. Kuwahara, A. et al. Wnt signaling and its downstream target N-myc regulate basal progenitors in the developing neocortex. Development 137, 1035–1044 (2010).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank C.L. Cepko (Harvard University), T. Matsuda (Kyoto University), T. Kitamura (The University of Tokyo), M. Nakafuku (Cincinnati Children's Hospital Medical Center and University of Cincinnati), H. Song (Johns Hopkins University) and H. Okano (Keio University) for plasmids, K. Ohsumi (Nagoya University) for antibodies, K. Tyssowski and E. Nigh for editing the manuscript, H. Okano for Nes-d4Venus mice, A. Miyajima for FACS, and members of the Gotoh laboratory for helpful discussion. This work was supported by Grants-in-Aid for Scientific Research (A) from the Ministry of Education, Culture, Sports, Science, and Technology (MEXT) of Japan, Innovative Areas “Neural Diversity and Neocortical Organization” of MEXT, by Core Research for Evolutional Science and Technology of the Japan Science and Technology Agency, by the Japan Society for the Promotion of Science, and by the Global COE Program “Integrative Life Science Based on the Study of Biosignaling Mechanisms” of MEXT.

Author information

Authors and Affiliations

Authors

Contributions

Y.K., Y.F., Y.H. and Y.G. designed the study. Y.K. and Y.F. carried out the experiments and analyzed the data. Y.H. supported the experiments. Y.K. and Y.G. wrote the manuscript. Y.G. supervised the study.

Corresponding author

Correspondence to Yukiko Gotoh.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–8 and Supplementary Table 1 (PDF 6220 kb)

Rights and permissions

Reprints and permissions

About this article

Cite this article

Kishi, Y., Fujii, Y., Hirabayashi, Y. et al. HMGA regulates the global chromatin state and neurogenic potential in neocortical precursor cells. Nat Neurosci 15, 1127–1133 (2012). https://doi.org/10.1038/nn.3165

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nn.3165

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