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:

Slowly dividing neural progenitors are an embryonic origin of adult neural stem cells

Abstract

The mechanism by which adult neural stem cells (NSCs) are established during development is unclear. In this study, analysis of cell cycle progression by examining retention of a histone 2B (H2B)-GFP fusion protein revealed that, in a subset of mouse embryonic neural progenitor cells (NPCs), the cell cycle slows between embryonic day (E) 13.5 and E15.5 while other embryonic NPCs continue to divide rapidly. By allowing H2B-GFP expressed at E9.5 to become diluted in dividing cells until the young adult stage, we determined that a majority of NSCs in the young adult subependymal zone (SEZ) originated from these slowly dividing embryonic NPCs. The cyclin-dependent kinase inhibitor p57 is highly expressed in this embryonic subpopulation, and the deletion of p57 impairs the emergence of adult NSCs. Our results suggest that a substantial fraction of adult SEZ NSCs is derived from a slowly dividing subpopulation of embryonic NPCs and identify p57 as a key factor in generating this embryonic origin of adult SEZ NSCs.

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: Slowly dividing NPCs in the embryonic brain.
Figure 2: Emergence of slowly dividing NPCs during embryogenesis.
Figure 3: H2B-GFP-retaining cells in the young adult SEZ.
Figure 4: Young adult NSCs retain H2B-GFP.
Figure 5: Greater p57 expression in slowly dividing embryonic NPCs than in other NPCs.
Figure 6: Conditional p57 deletion reduces slowly dividing embryonic NPCs.
Figure 7: Conditional p57 deletion reduces young adult SEZ NSCs and their progeny.
Figure 8: High p57 expression contributes to the maintenance of NPCs' undifferentiated state.

Similar content being viewed by others

References

  1. Cheung, T.H. & Rando, T.A. Molecular regulation of stem cell quiescence. Nat. Rev. Mol. Cell Biol. 14, 329–340 (2013).

    CAS  PubMed  Google Scholar 

  2. Kriegstein, A. & Alvarez-Buylla, A. The glial nature of embryonic and adult neural stem cells. Annu. Rev. Neurosci. 32, 149–184 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  3. Mirzadeh, Z., Merkle, F.T., Soriano-Navarro, M., Garcia-Verdugo, J.M. & Alvarez-Buylla, A. Neural stem cells confer unique pinwheel architecture to the ventricular surface in neurogenic regions of the adult brain. Cell Stem Cell 3, 265–278 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  4. Kageyama, R., Imayoshi, I. & Sakamoto, M. The role of neurogenesis in olfaction-dependent behaviors. Behav. Brain Res. 227, 459–463 (2012).

    PubMed  Google Scholar 

  5. Imayoshi, I. et al. Roles of continuous neurogenesis in the structural and functional integrity of the adult forebrain. Nat. Neurosci. 11, 1153–1161 (2008).

    CAS  PubMed  Google Scholar 

  6. Ernst, A. et al. Neurogenesis in the striatum of the adult human brain. Cell 156, 1072–1083 (2014).

    CAS  PubMed  Google Scholar 

  7. Ming, G.L. & Song, H. Adult neurogenesis in the mammalian brain: significant answers and significant questions. Neuron 70, 687–702 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  8. Anthony, T.E., Klein, C., Fishell, G. & Heintz, N. Radial glia serve as neuronal progenitors in all regions of the central nervous system. Neuron 41, 881–890 (2004).

    CAS  PubMed  Google Scholar 

  9. Doetsch, F. The glial identity of neural stem cells. Nat. Neurosci. 6, 1127–1134 (2003).

    CAS  PubMed  Google Scholar 

  10. Merkle, F.T., Tramontin, A.D., García-Verdugo, J.M. & Alvarez-Buylla, A. Radial glia give rise to adult neural stem cells in the subventricular zone. Proc. Natl. Acad. Sci. USA 101, 17528–17532 (2004).

    CAS  PubMed  PubMed Central  Google Scholar 

  11. Bowman, A.N., van Amerongen, R., Palmer, T.D. & Nusse, R. Lineage tracing with Axin2 reveals distinct developmental and adult populations of Wnt/β-catenin-responsive neural stem cells. Proc. Natl. Acad. Sci. USA 110, 7324–7329 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  12. 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).

    CAS  PubMed  Google Scholar 

  13. Noctor, S.C., Martínez-Cerdeño, V. & Kriegstein, A.R. Distinct behaviors of neural stem and progenitor cells underlie cortical neurogenesis. J. Comp. Neurol. 508, 28–44 (2008).

    PubMed  PubMed Central  Google Scholar 

  14. Furutachi, S., Matsumoto, A., Nakayama, K.I. & Gotoh, Y. p57 controls adult neural stem cell quiescence and modulates the pace of lifelong neurogenesis. EMBO J. 32, 970–981 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  15. Takahashi, T., Nowakowski, R.S. & Caviness, V.S. The cell cycle of the pseudostratified ventricular epithelium of the embryonic murine cerebral wall. J. Neurosci. 15, 6046–6057 (1995).

    CAS  PubMed  PubMed Central  Google Scholar 

  16. Calegari, F., Haubensak, W., Haffner, C. & Huttner, W.B. Selective lengthening of the cell cycle in the neurogenic subpopulation of neural progenitor cells during mouse brain development. J. Neurosci. 25, 6533–6538 (2005).

    CAS  PubMed  PubMed Central  Google Scholar 

  17. Tumbar, T. et al. Defining the epithelial stem cell niche in skin. Science 303, 359–363 (2004).

    CAS  PubMed  Google Scholar 

  18. Wilson, A. et al. Hematopoietic stem cells reversibly switch from dormancy to self-renewal during homeostasis and repair. Cell 135, 1118–1129 (2008).

    CAS  PubMed  Google Scholar 

  19. Chakkalakal, J.V., Jones, K.M., Basson, M.A. & Brack, A.S. The aged niche disrupts muscle stem cell quiescence. Nature 490, 355–360 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  20. Nonaka-Kinoshita, M. et al. Regulation of cerebral cortex size and folding by expansion of basal progenitors. EMBO J. 32, 1817–1828 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  21. Graham, V., Khudyakov, J., Ellis, P. & Pevny, L. SOX2 functions to maintain neural progenitor identity. Neuron 39, 749–765 (2003).

    CAS  PubMed  Google Scholar 

  22. Tran, V., Lim, C., Xie, J. & Chen, X. Asymmetric division of Drosophila male germline stem cell shows asymmetric histone distribution. Science 338, 679–682 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  23. Corti, S. et al. Isolation and characterization of murine neural stem/progenitor cells based on Prominin-1 expression. Exp. Neurol. 205, 547–562 (2007).

    CAS  PubMed  Google Scholar 

  24. Götz, M. & Huttner, W.B. The cell biology of neurogenesis. Nat. Rev. Mol. Cell Biol. 6, 777–788 (2005).

    PubMed  Google Scholar 

  25. Doetsch, F., Caillé, I., Lim, D.A., García-Verdugo, J.M. & Alvarez-Buylla, A. Subventricular zone astrocytes are neural stem cells in the adult mammalian brain. Cell 97, 703–716 (1999).

    CAS  PubMed  Google Scholar 

  26. Spassky, N. et al. Adult ependymal cells are postmitotic and are derived from radial glial cells during embryogenesis. J. Neurosci. 25, 10–18 (2005).

    CAS  PubMed  PubMed Central  Google Scholar 

  27. Merkle, F.T., Mirzadeh, Z. & Alvarez-Buylla, A. Mosaic organization of neural stem cells in the adult brain. Science 317, 381–384 (2007).

    CAS  PubMed  Google Scholar 

  28. Young, K.M., Fogarty, M., Kessaris, N. & Richardson, W.D. Subventricular zone stem cells are heterogeneous with respect to their embryonic origins and neurogenic fates in the adult olfactory bulb. J. Neurosci. 27, 8286–8296 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  29. Lledo, P.M., Merkle, F.T. & Alvarez-Buylla, A. Origin and function of olfactory bulb interneuron diversity. Trends Neurosci. 31, 392–400 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  30. Doetsch, F., García-Verdugo, J.M. & Alvarez-Buylla, A. Regeneration of a germinal layer in the adult mammalian brain. Proc. Natl. Acad. Sci. USA 96, 11619–11624 (1999).

    CAS  PubMed  PubMed Central  Google Scholar 

  31. Matsumoto, A. et al. p57 is required for quiescence and maintenance of adult hematopoietic stem cells. Cell Stem Cell 9, 262–271 (2011).

    CAS  PubMed  Google Scholar 

  32. Isaka, F. et al. Ectopic expression of the bHLH gene Math1 disturbs neural development. Eur. J. Neurosci. 11, 2582–2588 (1999).

    CAS  PubMed  Google Scholar 

  33. Pateras, I.S., Apostolopoulou, K., Niforou, K., Kotsinas, A. & Gorgoulis, V.G. p57KIP2: “Kip”ing the cell under control. Mol. Cancer Res. 7, 1902–1919 (2009).

    CAS  PubMed  Google Scholar 

  34. Matsumoto, A. et al. Deregulation of the p57–E2F1-p53 axis results in nonobstructive hydrocephalus and cerebellar malformation in mice. Mol. Cell. Biol. 31, 4176–4192 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  35. Joseph, B. et al. p57Kip2 is a repressor of Mash1 activity and neuronal differentiation in neural stem cells. Cell Death Differ. 16, 1256–1265 (2009).

    CAS  PubMed  Google Scholar 

  36. Joseph, B. et al. p57(Kip2) cooperates with Nurr1 in developing dopamine cells. Proc. Natl. Acad. Sci. USA 100, 15619–15624 (2003).

    CAS  PubMed  PubMed Central  Google Scholar 

  37. 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).

    CAS  PubMed  PubMed Central  Google Scholar 

  38. Porlan, E. et al. Transcriptional repression of Bmp2 by p21(Waf1/Cip1) links quiescence to neural stem cell maintenance. Nat. Neurosci. 16, 1567–1575 (2013).

    CAS  PubMed  Google Scholar 

  39. Kueh, H.Y., Champhekar, A., Nutt, S.L., Elowitz, M.B. & Rothenberg, E.V. Positive feedback between PU.1 and the cell cycle controls myeloid differentiation. Science 341, 670–673 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  40. Groszer, M. et al. PTEN negatively regulates neural stem cell self-renewal by modulating G0–G1 cell cycle entry. Proc. Natl. Acad. Sci. USA 103, 111–116 (2006).

    CAS  PubMed  Google Scholar 

  41. Renault, V.M. et al. FoxO3 regulates neural stem cell homeostasis. Cell Stem Cell 5, 527–539 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  42. Mira, H. et al. Signaling through BMPR-IA regulates quiescence and long-term activity of neural stem cells in the adult hippocampus. Cell Stem Cell 7, 78–89 (2010).

    CAS  PubMed  Google Scholar 

  43. Imayoshi, I., Sakamoto, M., Yamaguchi, M., Mori, K. & Kageyama, R. Essential roles of Notch signaling in maintenance of neural stem cells in developing and adult brains. J. Neurosci. 30, 3489–3498 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  44. Kawaguchi, D., Furutachi, S., Kawai, H., Hozumi, K. & Gotoh, Y. Dll1 maintains quiescence of adult neural stem cells and segregates asymmetrically during mitosis. Nat. Commun. 4, 1880 (2013).

    PubMed  Google Scholar 

  45. Mignone, J.L., Kukekov, V., Chiang, A.S., Steindler, D. & Enikolopov, G. Neural stem and progenitor cells in nestin-GFP transgenic mice. J. Comp. Neurol. 469, 311–324 (2004).

    CAS  PubMed  Google Scholar 

  46. López-Juárez, A. et al. Gsx2 controls region-specific activation of neural stem cells and injury-induced neurogenesis in the adult subventricular zone. Genes Dev. 27, 1272–1287 (2013).

    PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank K. Tyssowski for editing the manuscript, F. Calegari (Center for Regenerative Therapies Dresden (CRTD)) for reagents, H. Sugizaki, C. Koga and J. Takeuchi for FACS analysis, A Shitamukai, F. Matsuzaki and Y. Sasai for comments and members of the Gotoh laboratory for discussion. This work was supported by Core Research for Evolutional Science and Technology (CREST) of the Japan Science and Technology Agency.

Author information

Authors and Affiliations

Authors

Contributions

S.F. designed the study, conducted the experimental work and wrote the manuscript. H.M., T.W., H.K., N.Y. and Y. Harada conducted the experimental work. I.I. generated Nestin-NLS-mCherry mice. M.N. provided 9TB-Dox. K.I.N. generated p57-floxed mice. Y. Hirabayashi supervised the project. Y.G. supervised the project and wrote the manuscript.

Corresponding author

Correspondence to Yukiko Gotoh.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Integrated supplementary information

Supplementary Figure 1 Confirmation of the reversibility of H2B-GFP mRNA induction.

(a) Expression of the H2B-GFP transgene was induced at E9.5, and the embryos were examined at E10.5 or E16.5. At E10.5, the telencephalon of Rosa-rtTA/TRE-mCMV-H2B-GFP embryos was dissected and assayed for the level of H2B-GFP mRNA. The telencephalon of E10.5 Rosa-rtTA/TRE-mCMV-H2B-GFP embryos without Dox injection was examined as a negative control. At E16.5, the telencephalon of Rosa-rtTA/TRE-mCMV-H2B-GFP embryos was dissected, and LRCs (the top 10% fraction for H2B-GFP level) and non-LRCs among NPCs (CD133+CD24 population) were isolated by FACS for quantification of H2B-GFP mRNA. (b) Data are means ± SD (n = 3 experiments). Two-tailed Student’s t test; P < 0.001 ((+)Dox E10.5 versus (–)Dox E10.5; (+)Dox E10.5 versus (+)Dox E16.5 LRC; (+)Dox E10.5 versus (+)Dox E16.5 Non-LRC), P = 0.6 ((+)Dox E16.5 LRC versus (+)Dox E16.5 Non-LRC). ***P < 0.001; n.s., not significant (P ≥ 0.05).

Supplementary Figure 2 The distribution of H2B-GFP label-retaining cells in the E17.5 mouse brain.

(a) 9TB-Dox was injected at E9.5 to pregnant mice and brain sections of Rosa-rtTA/TRE-mCMV-H2B-GFP embryos were prepared at E17.5. (b) Immunohistochemistry with an antibody against GFP (green) at different rostrocaudal levels. Nuclei were stained with Hoechst (blue). The boxed regions correspond to the scheme in Supplementary Fig. 3d. Asterisks indicate choroid plexus.

Supplementary Figure 3 The distribution of H2B-GFP label-retaining cells in ventricular zone subregions of the E17.5 mouse brain.

(a) Pregnant mice were injected with 9TB-Dox at E9.5, and brain sections of Rosa-rtTA/TRE-mCMV-H2B-GFP embryos were prepared at E17.5. (b and c) Staining of sections for GFP (green), Pax6 (red, b) and Gsh2 (red, c). Nuclei were stained with Hoechst (blue). The boxed regions in the left panel are shown at higher magnification in the right panels. LRCs (arrowheads) were observed in the ventricular zone of the telencephalon. (d) The distribution of H2B-GFP-retaining NPCs in ventricular zone subregions at different rostrocaudal levels. Ventricular zone subregions are defined as follows: Pax6++Ascl1 as neocortex (NCX), Pax6+Ascl1+ as dorsal lateral ganglionic eminence (dLGE), Pax6Ascl1+ (lateral) as ventral lateral ganglionic eminence (vLGE), and Pax6Ascl1+ (medial) as septum (SE). H2B-GFP-retaining NPCs were distributed throughout the Ascl1+ LGE but were enriched in the Pax6+Ascl1+ dLGE. Data are means ± SEM (n = 3 animals). The scheme corresponds to the boxed regions in Supplementary Fig. 2b. Scale bars: 100 μm in (b and c, left) and 25 μm in (b and c, right).

Supplementary Figure 4 Overlapping distributions of H2B-GFP retention and EdU retention.

(a) Pregnant mice were injected with 9TB-Dox at E9.5 and with EdU (four times at 3-h intervals) at E10.5. Brain sections of Rosa-rtTA/TRE-mCMV-H2B-GFP embryos or pups were prepared at E17.5 or P14. (b) Sections for native fluorescence of GFP (green) and EdU staining (red) at E17.5 (top) and P14 (bottom). Nuclei were stained with Hoechst (blue). Asterisks indicate the choroid plexus. (c) Quantitative analysis of H2B-GFP- and EdU-retaining cells in SEZ subregions at P14. Data are means ± SEM (n = 3 animals). Scale bar: 20 μm.

Supplementary Figure 5 Lack of Dcx and NG2 expression in H2B-GFP label-retaining cells in the ventricular zone of the E17.5 mouse brain.

(a) Pregnant mice were injected with 9TB-Dox at E9.5, and brain sections of Rosa-rtTA/TRE-mCMV-H2B-GFP embryos were prepared at E17.5. (b) Staining for GFP (green) and Dcx (red). Nuclei were stained with Hoechst (blue). The boxed region in the left panel is shown at higher magnification in the right panel. (c) Staining for GFP (green) and NG2 (red). Nuclei were stained with Hoechst (blue). Scale bars: 25 μm in (b, left and c) and 7.5 μm in (b, right).

Supplementary Figure 6 FACS strategy used to isolate the embryonic and postnatal NPCs.

(a) Structure of the Nestin-NLS-mCherry transgene. (b) Sections of the forebrain of E13.5 Nestin-NLS-mCherry transgenic mice. The mCherry fluorescence (red) was localized to the ventricular zone. (c) FACS profiles for isolation of NPCs at various time points after H2B-GFP induction. NPCs (boxes in right panels) were defined as CD133+CD24 population (E10.5) or CD133+mCherry+ population (E13.5, E15.5, E17.5 and P3). (d) FACS profiles of negative control cells at E10.5 (left panel) and E15.5 (right panel). (Related to Fig. 2)

Supplementary Figure 7 The distribution of H2B-GFP induced at various time points.

(a) Pregnant mice were injected with 9TB-Dox at E9.5 (one time), E14.5 (two times at 8-h interval), or E17.5 (two times at 8-h interval), and brain sections of Rosa-rtTA/TRE-mCMV-H2B-GFP animals were prepared 1 day after the injection at E14.5 or E17.5 (E15.5 or E18.5, respectively) or at P14 or P28. (b and c) Immunohistochemistry with an antibody against GFP (green). Nuclei were stained with Hoechst (blue). The boxed regions in (b) are shown at higher magnification in the insets. The successful induction of H2B-GFP by 9TB-Dox at E9.5 and E14.5 was confirmed by its distribution in the neocortex at P14 (c). UL, upper layer; DL, deep layer of the neocortex. Asterisk indicates the choroid plexus. (d) Immunohistochemistry with antibodies against GFP (green) and GFAP (red). Nuclei were stained with Hoechst (blue). Note that when 9TB-Dox was administered at E17.5, only a small fraction (31.7 ± 6.0%, mean ± SEM, n = 3 animals) of SEZ-NSCs (GFAP+Sox2+apical process+ cells) retained H2B-GFP at P28, probably as a result of the slowly cycling embryonic subpopulation having already slowed down the cell cycle by E17.5. Indeed, the labeling by H2B-GFP among cells in the ventricular zone was only partial at E18.5 when 9TB-Dox was administered at E17.5. By contrast, most NPCs in the ventricular zone were H2B-GFP positive at E15.5 when 9TB-Dox was administered at E14.5. Strikingly, 92.0 ± 2.2% (mean ± SEM, n = 3 animals) of GFAP+Sox2+apical process+ SEZ-NSCs in the lateral wall of the SEZ retained H2B-GFP at high levels even at P28 when Dox was administered at E14.5, consistent with the notion that most of the slowly dividing embryonic progenitors slow down the cell cycle between E13.5 and E15.5 and contribute to the major population of adult SEZ-NSCs. Scale bars: 50 μm in (b and d) and 200 μm in (c).

Supplementary Figure 8 H2B-GFP-retaining cells in the young adult brain.

(a) Transient H2B-GFP expression was induced at E9.5 and then allowed to become diluted in dividing cells until postnatal or young adult stages. (b) Distribution of H2B-GFP LRCs in the SEZ at different rostrocaudal levels at P28. Shown images are immunohistochemistry with antibodies against GFP (green) and S100β (red). (c) Immunohistochemistry with an antibody against GFP (green) of P28 Rosa-rtTA/TRE-mCMV-H2B-GFP mouse (left) and its negative control (TRE-mCMV-H2B-GFP, right) brain sections. (d) Immunohistochemistry with an antibody against GFP (green) of P14 Rosa-rtTA/TRE-mCMV-H2B-GFP mouse with Dox (left) or without Dox (right) injection at E9.5. Nuclei were stained with Hoechst (blue). Asterisks indicate choroid plexus. Scale bars: 100 μm in (b and c) and 50 μm in (d).

Supplementary Figure 9 H2B-GFP- and BrdU-retaining cells express p57 at a high level.

(a) Pregnant mice were injected with 9TB-Dox at E9.5, and brain sections of Rosa-rtTA/TRE-mCMV-H2B-GFP embryos were prepared at E17.5. (b) Immunohistochemistry with antibodies against GFP (green) and p57 (red). Nuclei were stained with Hoechst (blue). Arrowheads indicate LRCs expressing p57 at a high level. (c) Pregnant mice were injected with BrdU (four times at 3-h intervals) at E10.5, and brain sections of embryos or pups were prepared at E17.5, E18.5, P0, and P10. (d) Immunohistochemistry with antibodies against BrdU (green) and p57 (red). Nuclei were stained with TO-PRO-3 (blue). The boxed regions in the upper panels are shown at higher magnification in the lower panels. Scale bar: 25 μm.

Supplementary Figure 10 Effects of p57 overexpression.

(a) In utero electroporation (IUE) was performed at E14.5 with expression plasmids for GFP alone (control) or for GFP and p57, and brain sections were subjected to immunohistochemistry at E18.5 (b) or P4 (d). (b) Immunohistochemistry with antibodies against GFP (green), Sox2 (blue), and PCNA (red). Arrowheads indicate GFP+ cells in the ventricular zone. (c) Quantification of the data in (b). Data are means ± SEM (n = 4 and 5 animals for control and p57, respectively). Two-tailed Student’s t test; P = 0.0072. (d) Immunohistochemistry with antibodies against GFP (green), Sox2 (blue), and Dcx (red). (e) Our model. Slowly dividing embryonic NPCs are an embryonic origin of adult NSCs, and p57 is responsible for their emergence. Scale bars: 25 μm in (b) and 20 μm in (d). **P < 0.01.

Supplementary information

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Furutachi, S., Miya, H., Watanabe, T. et al. Slowly dividing neural progenitors are an embryonic origin of adult neural stem cells. Nat Neurosci 18, 657–665 (2015). https://doi.org/10.1038/nn.3989

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nn.3989

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