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:

A large pool of actively cycling progenitors orchestrates self-renewal and injury repair of an ectodermal appendage

Abstract

The classical model of tissue renewal posits that small numbers of quiescent stem cells (SCs) give rise to proliferating transit-amplifying cells before terminal differentiation. However, many organs house pools of SCs with proliferative and differentiation potentials that diverge from this template. Resolving SC identity and organization is therefore central to understanding tissue renewal. Here, using a combination of single-cell RNA sequencing (scRNA-seq), mouse genetics and tissue injury approaches, we uncover cellular hierarchies and mechanisms that underlie the maintenance and repair of the continuously growing mouse incisor. Our results reveal that, during homeostasis, a group of actively cycling epithelial progenitors generates enamel-producing ameloblasts and adjacent layers of non-ameloblast cells. After injury, tissue repair was achieved through transient increases in progenitor-cell proliferation and through direct conversion of Notch1-expressing cells to ameloblasts. We elucidate epithelial SC identity, position and function, providing a mechanistic basis for the homeostasis and repair of a fast-turnover ectodermal appendage.

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

Fig. 1: Identification of distinct cell types by single-cell transcriptomic analysis and delineation of their spatial organization in the incisor epithelium.
Fig. 2: An unbiased catalogue of cell types during amelogenesis.
Fig. 3: Mapping of class 3 cell types to spatial regions of the incisor-epithelium growth region.
Fig. 4: Class 1 cells house progenitor cells that give rise to class 2 and class 3 populations
Fig. 5: Proliferation dynamics of the incisor epithelium growth region.
Fig. 6: Single-cell transcriptomic analysis reveals changes in cell cycle and differentiation program after injury.
Fig. 7: SI cells undergo direct conversion to ameloblasts to contribute to the recovery of injured incisor epithelium.

Similar content being viewed by others

Data availability

The scRNA-seq data that support the findings of this study have been deposited in the Gene Expression Omnibus (GEO) under accession code GSE131204. Source data for figures are provided in Supplementary Table 11. All other data supporting the findings of this study are available from the corresponding authors on reasonable request.

Code availability

The custom code developed for the whole-mount proliferation assay (Fig. 5d–f) is provided at https://github.com/kephale/incisor-cell-segmentation. The code for selected single cell RNAseq data analyses including SPRING visualizations is available as Jupyter notebooks at https:///www.github.com/rapolaszilionis/Sharir_et_al_2019.

References

  1. Ahn, Y. Signaling in tooth, hair, and mammary placodes. Curr. Top. Dev. Biol. 111, 421–459 (2015).

  2. Naveau, A., Seidel, K. & Klein, O. D. Tooth, hair and claw: comparing epithelial stem cell niches of ectodermal appendages. Exp. Cell Res. 325, 96–103 (2014).

    Article  CAS  Google Scholar 

  3. Thesleff, I., Vaahtokari, A. & Partanen, A. M. Regulation of organogenesis. Common molecular mechanisms regulating the development of teeth and other organs. Int. J. Dev. Biol. 39, 35–50 (1995).

    CAS  PubMed  Google Scholar 

  4. Lajtha, L. G. Stem cell concepts. Differentiation 14, 23–33 (1979).

    Article  CAS  Google Scholar 

  5. Cotsarelis, G., Sun, T.-T. & Lavker, R. M. Label-retaining cells reside in the bulge area of pilosebaceous unit: implications for follicular stem cells, hair cycle, and skin carcinogenesis. Cell 61, 1329–1337 (1990).

    Article  CAS  Google Scholar 

  6. Mackenzie, I. C. & Bickenbach, J. R. Label-retaining keratinocytes and langerhans cells in mouse epithelia. Cell Tissue Res. 242, 551–556 (1985).

    Article  CAS  Google Scholar 

  7. Clevers, H. & Watt, F. M. Defining adult stem cells by function, not by phenotype. Annu. Rev. Biochem. 87, 1015–1027 (2018).

    Article  CAS  Google Scholar 

  8. Paul, F. et al. Transcriptional heterogeneity and lineage commitment in myeloid progenitors. Cell 163, 1663–1677 (2015).

    Article  CAS  Google Scholar 

  9. Yang, H., Adam, R. C., Ge, Y., Hua, Z. L. & Fuchs, E. Epithelial-mesenchymal micro-niches govern stem cell lineage choices. Cell 169, 483–496 (2017).

    Article  CAS  Google Scholar 

  10. Ness, A. R. Eruption rates of impeded and unimpeded mandibular incisors of the adult laboratory mouse. Arch. Oral Biol. 10, 439–451 (1965).

    Article  CAS  Google Scholar 

  11. Warshawsky, H. & Smith, C. E. Morphological classification of rat incisor ameloblasts. Anat. Rec. 179, 423–445 (1974).

  12. Harada, H. et al. Localization of putative stem cells in dental epithelium and their association with notch and FGF signaling. J. Cell Biol. 147, 105–120 (1999).

    Article  CAS  Google Scholar 

  13. Morrison, S. J., Uchida, N. & Weissman, I. L. The biology of hematopoietic stem cells. Annu. Rev. Cell Dev. Biol. 11, 35–71 (1995).

    Article  CAS  Google Scholar 

  14. Hu, J. K.-H., Mushegyan, V. & Klein, O. D. On the cutting edge of organ renewal: identification, regulation, and evolution of incisor stem cells. Genesis 52, 79–92 (2014).

  15. Biehs, B. et al. BMI1 represses Ink4a/Arf and Hox genes to regulate stem cells in the rodent incisor. Nat. Cell Biol. 15, 846–852 (2013).

    Article  CAS  Google Scholar 

  16. Juuri, E. et al. Sox2 + stem cells contribute to all epithelial lineages of the tooth via Sfrp5 + progenitors. Dev. Cell 23, 317–328 (2012).

    Article  CAS  Google Scholar 

  17. Seidel, K. et al. Resolving stem and progenitor cells in the adult mouse incisor through gene co-expression analysis. eLife 6, e24712 (2017).

    Article  Google Scholar 

  18. Seidel, K. et al. Hedgehog signaling regulates the generation of ameloblast progenitors in the continuously growing mouse incisor. Dev. Camb. Engl. 137, 3753–3761 (2010).

    CAS  Google Scholar 

  19. Weinreb, C., Wolock, S. & Klein, A. M. SPRING: a kinetic interface for visualizing high dimensional single-cell expression data. Bioinformatics 34, 1246–1248 (2018).

    Article  CAS  Google Scholar 

  20. Butler, A., Hoffman, P., Smibert, P., Papalexi, E. & Satija, R. Integrating single-cell transcriptomic data across different conditions, technologies, and species. Nat. Biotechnol. 36, 411–420 (2018).

    Article  CAS  Google Scholar 

  21. Weinreb, C., Wolock, S., Tusi, B. K., Socolovsky, M. & Klein, A. M. Fundamental limits on dynamic inference from single-cell snapshots. Proc. Natl Acad. Sci. USA 115, E2467–E2476 (2018).

    Article  CAS  Google Scholar 

  22. Nanci, A. Ten Cate’s Oral Histology 8th edn (Elsevier, 2012).

  23. La Manno, G. et al. RNA velocity of single cells. Nature 560, 494–498 (2018).

    Article  Google Scholar 

  24. Herman, J. S., Sagar & Grün, D. FateID infers cell fate bias in multipotent progenitors from single-cell RNA-seq data. Nat. Methods 15, 379–386 (2018).

    Article  CAS  Google Scholar 

  25. Foudi, A. et al. Analysis of histone 2B-GFP retention reveals slowly cycling hematopoietic stem cells. Nat. Biotechnol. 27, 84–90 (2009).

    Article  CAS  Google Scholar 

  26. Doupé, D. P., Klein, A. M., Simons, B. D. & Jones, P. H. The ordered architecture of murine ear epidermis is maintained by progenitor cells with random fate. Dev. Cell 18, 317–323 (2010).

    Article  Google Scholar 

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

    Article  CAS  Google Scholar 

  28. 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 

  29. Stange, D. E. et al. Differentiated Troy + chief cells act as reserve stem cells to generate all lineages of the stomach epithelium. Cell 155, 357–368 (2013).

    Article  CAS  Google Scholar 

  30. Liu, H., Yan, X., Pandya, M., Luan, X. & Diekwisch, T. G. H. Daughters of the enamel organ: development, fate, and function of the stratum intermedium, stellate reticulum, and outer enamel epithelium. Stem Cells Dev. 25, 1580–1590 (2016).

    Article  CAS  Google Scholar 

  31. Harada, H. et al. Stratum intermedium lineage diverges from ameloblast lineage via notch signaling. Biochem. Biophys. Res. Commun. 340, 611–616 (2006).

    Article  CAS  Google Scholar 

  32. Jheon, A. H. et al. Inhibition of notch signaling during mouse incisor renewal leads to enamel defects. J. Bone Miner. Res. 31, 152–162 (2016).

    Article  CAS  Google Scholar 

  33. Voehringer, D., Liang, H.-E. & Locksley, R. M. Homeostasis and effector function of lymphopenia-induced ‘memory-like’ T cells in constitutively T cell-depleted mice. J. Immunol. 180, 4742–4753 (2008).

    Article  CAS  Google Scholar 

  34. Kaukua, N. et al. Glial origin of mesenchymal stem cells in a tooth model system. Nature 513, 551–554 (2014).

    Article  CAS  Google Scholar 

  35. Hu, J. K.-H. et al. An FAK-YAP-mTOR signaling axis regulates stem cell-based tissue renewal in mice. Cell Stem Cell 21, 91–106 (2017).

    Article  CAS  Google Scholar 

  36. Zhao, H. et al. Secretion of Shh by a neurovascular bundle niche supports mesenchymal stem cell homeostasis in the adult mouse incisor. Cell Stem Cell 14, 160–173 (2014).

    Article  CAS  Google Scholar 

  37. Klein, A. M., Nakagawa, T., Ichikawa, R., Yoshida, S. & Simons, B. D. Mouse germ line stem cells undergo rapid and stochastic turnover. Cell Stem Cell 7, 214–224 (2010).

    Article  CAS  Google Scholar 

  38. Snippert, H. J. et al. Intestinal crypt homeostasis results from neutral competition between symmetrically dividing Lgr5 stem cells. Cell 143, 134–144 (2010).

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  40. Leung, Y. et al. Bifunctional ectodermal stem cells around the nail display dual fate homeostasis and adaptive wounding response toward nail regeneration. Proc. Natl Acad. Sci. USA 111, 15114–15119 (2014).

    Article  CAS  Google Scholar 

  41. Wabik, A. & Jones, P. H. Switching roles: the functional plasticity of adult tissue stem cells. EMBO J. 34, 1164–1179 (2015).

    Article  CAS  Google Scholar 

  42. Blanpain, C. & Fuchs, E. Plasticity of epithelial stem cells in tissue regeneration. Science 344, 1242281 (2014).

    Article  Google Scholar 

  43. Dell’Orso, S. et al. Single cell analysis of adult skeletal muscle stem cells in homeostatic and regenerative conditions. Development 146, dev174177 (2019).

    Article  Google Scholar 

  44. Gadye, L. et al. Injury activates transient olfactory stem cell states with diverse lineage capacities. Cell Stem Cell 21, 775–790 (2017).

    Article  CAS  Google Scholar 

  45. Joost, S. et al. Single-cell transcriptomics of traced epidermal and hair follicle stem cells reveals rapid adaptations during wound healing. Cell Rep. 25, 585–597 (2018).

    Article  CAS  Google Scholar 

  46. Plasschaert, L. W. et al. A single-cell atlas of the airway epithelium reveals the CFTR-rich pulmonary ionocyte. Nature 560, 377–381 (2018).

    Article  CAS  Google Scholar 

  47. Nusse, Y. M. et al. Parasitic helminths induce fetal-like reversion in the intestinal stem cell niche. Nature 559, 109–113 (2018).

    Article  CAS  Google Scholar 

  48. Velten, L. et al. Human haematopoietic stem cell lineage commitment is a continuous process. Nat. Cell Biol. 19, 271–281 (2017).

    Article  CAS  Google Scholar 

  49. Metscher, B. D. MicroCT for comparative morphology: simple staining methods allow high-contrast 3D imaging of diverse non-mineralized animal tissues. BMC Physiol. 9, 11 (2009).

    Article  Google Scholar 

  50. Djomehri, S. I. et al. Mineral density volume gradients in normal and diseased human tissues. PloS ONE 10, e0121611 (2015).

    Article  Google Scholar 

  51. Hu, J. K.-H., McGlinn, E., Harfe, B. D., Kardon, G. & Tabin, C. J. Autonomous and nonautonomous roles of hedgehog signaling in regulating limb muscle formation. Genes Dev. 26, 2088–2102 (2012).

    Article  CAS  Google Scholar 

  52. Chavez, M. G. et al. Isolation and culture of dental epithelial stem cells from the adult mouse incisor. J. Vis. Exp. 87, e51266 (2014).

  53. Wolf, F. A., Angerer, P. & Theis, F. J. SCANPY: large-scale single-cell gene expression data analysis. Genome Biol. 19, 15 (2018).

    Article  Google Scholar 

  54. Zemmour, D. et al. Single-cell gene expression reveals a landscape of regulatory T cell phenotypes shaped by the TCR. Nat. Immunol. 19, 291–301 (2018).

    Article  CAS  Google Scholar 

  55. Zilionis, R. et al. Single-cell transcriptomics of human and mouse lung cancers reveals conserved myeloid populations across individuals and species. Immunity 50, 1317–1334 (2019).

    Article  Google Scholar 

  56. Jaitin, D. A. et al. Massively parallel single-cell RNA-seq for marker-free decomposition of tissues into cell types. Science 343, 776–779 (2014).

    Article  CAS  Google Scholar 

  57. Hu, J. C.-C. et al. Enamel defects and ameloblast-specific expression in Enam knock-out/lacZ knock-in mice. J. Biol. Chem. 283, 10858–10871 (2008).

    Article  CAS  Google Scholar 

  58. Smith, C. E., Hu, Y., Hu, J. C.-C. & Simmer, J. P. Ultrastructure of early amelogenesis in wild-type, Amelx -/-, and Enam -/- mice: enamel ribbon initiation on dentin mineral and ribbon orientation by ameloblasts. Mol. Genet. Genomic Med. 4, 662–683 (2016).

    Article  CAS  Google Scholar 

  59. Tusi, B. K. et al. Population snapshots predict early haematopoietic and erythroid hierarchies. Nature 555, 54–60 (2018).

    Article  CAS  Google Scholar 

  60. Klein, A. M. et al. Droplet barcoding for single-cell transcriptomics applied to embryonic stem cells. Cell 161, 1187–1201 (2015).

    Article  CAS  Google Scholar 

  61. Mootha, V. K. et al. PGC-1α-responsive genes involved in oxidative phosphorylation are coordinately downregulated in human diabetes. Nat. Genet. 34, 267–273 (2003).

    Article  CAS  Google Scholar 

  62. Subramanian, A. et al. Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles. Proc. Natl Acad. Sci. USA 102, 15545–15550 (2005).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank A. Rathnayake, R. D’Urso, N. Wang, B. Hoehn and S. Alto for technical assistance, L. Jimenez-Rojo for mouse surgeries, K. Lindquist for consulting on GSEA analyses and J. Bush as well as members of the O.D.K. and A.M.K. laboratories for helpful discussion. This work was supported by the NIH (R35-DE026602 and R01-DE027620 to O.D.K). A.S. was funded by the NIH (K08-DE026219). J.H. was funded by the NIH (K99-DE025874). A.M.K. was funded by the NIH (5R33CA212697). UCSF Parnassus Flow Cytometry Core is supported by the NIH (P30DK063720 and 1S10OD021822-01). UCSF Core Center for Musculoskeletal Biology and Medicine is supported by the NIH (P30AR066262).

Author information

Authors and Affiliations

Authors

Contributions

A.S. conceived the study, designed and performed experiments, and analysed data. P.M., D.C.-A., M.W., T.W., J.H., L.E., K.H, and P.P. performed experiments and analysed data. R.Z., K.K. and T.M. analysed data. C.W.S. contributed essential reagents. O.D.K. and A.M.K. directed the study and wrote the paper with A.S., P.M., R.Z., and J.H.

Corresponding authors

Correspondence to Allon M. Klein or Ophir D. Klein.

Ethics declarations

Competing interests

A.M.K. is a founder of 1CellBio. C.W.S. is employed by Genentech.

Additional information

Publisher’s note: Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Integrated supplementary information

Supplementary Figure 1 The incisor epithelium is in steady state between 8 and 16 weeks of age.

a,b, In vivo micro-computed tomography (µCT) images of mouse hemi-mandibles immediately after a small notch was made in the left incisor (a) and 4 days after (b). Boxed areas (orange) are enlarged on the right. The change in distance between the notch and the tooth’s labial surface (red and green lines) corresponds to the amount of enamel growth in 4 days. Scale bars, 3 mm. c, µCT images showing the left hemi-mandible at different ages and the associated changes in size of the incisor growth region. Scale bars, 5 mm in left panels; 100 μm in right panels. d, Immunostaining for BrdU in the incisor growth region. Mice were dosed three times with BrdU over 3 days before harvesting to assess cell division. Nuclei were counterstained with DAPI. Scale bars, 100 μm. e, Schematic models showing broadly distributed cycling cells in the incisor growth region of young mice (<5 weeks, top) versus the restriction of cycling cells in the IEE and the adjacent SR in older mice (bottom). f, TUNEL staining revealed few (1-2 cells/section) apoptotic cells in cervical loops from 8 and 16 weeks old mice. Yellow arrowheads mark TUNEL signals. Scale bars, 100 μm. g, Quantification of the rate of incisor growth per week and the incisor growth region volume, both assessed by µCT. N=3 animals. h, Percentage of BrdU+ cells/section. Dashed lines outline epithelium. N=3 animals. Scatter plots indicate the mean of samples and error bars represent ± S.D. of the mean. Normally distributed data were analyzed using parametric tests including one-way ANOVA with Tukey–Kramer post hoc test. Significance was taken as P<0.05 with a confidence interval of 95%. * P< 0.05, ** P < 0.01, ** P < 0.01, *** P < 0.001.

Supplementary Figure 2 Previously reported markers are broadly expressed in the incisor growth region.

RNAscope in situ hybridization in mouse incisor growth region for Sox2 (a), Gli1 (d), Bmi1 (g) and Lrig1 (j); transcripts are visualized as red dots. Enlargements of the boxed insets from the dorsal (green) and ventral (orange) OEE, and the middle portion of the IEE (yellow) regions for each marker are shown on the right. Schematic view of the incisor growth region highlights the expression domains of the above genes. Sox2 (a’), Gli1 (d’) and Bmi1 (g’) are expressed throughout the proximal dental epithelium, including the OEE, IEE and the SR, whereas Lrig1 (j’) is present almost exclusively in the OEE and the OSR adjacent to the OEE. Representative images of RFP (Sox2, Lrig1 and Bmi1) or βgal (Gli1) immunostaining in the incisor growth region of Sox2CreER;R26tdTomato (b), Bmi1CreER;R26tdTomato (h), Lrig1CreER;R26tdTomato (k) and Gli1CreER;R26lacZ (e) mice 24 hours (top) or 21 days (bottom) after a single, low-dose tamoxifen injection. The initial labelling of all these markers was consistent with our in situ hybridization results, indicating that the progeny of Sox2, Gli1 and Bmi1 cells may originate from cells in any of the above regions. In contrast, cells labelled with Lrig1 were found exclusively in the OEE and condensed SR. Green, yellow and orange arrowheads in 24 hours chase mark cells in the dorsal, ventral OEE and IEE, respectively. Dashed lines outline incisor epithelium. SPRING plots of the entire control (top right) and of Class 1 after regressing out the cell cycle effect (right bottom), overlaid with gene expression of the above markers (Sox2 (c), Gli1 (f), Bmi1 (i) and Lrig1 (l); Color scale as in Fig. 1g. Scale bars, 100 μm.

Supplementary Figure 3 Extended analysis of the transcriptomic signatures of the incisor epithelium during homeostasis.

a, b, Flow cytometry gating used for sorting the control (a) and recovery (b) incisor epithelial cells. Experiment performed once. c, SPRING plot of the entire control dataset before filtering. d, Examples of genes specific to populations identified as mesenchymal (dark red) and immune (light blue) cells. e, SPRING plot of the control incisor epithelium dataset after removal of mesenchymal and immune cells. f, SPRING plots overlaid with gene expression patterns for the markers described in Fig. 1h. g, SPRING plots overlaid with gene expression patterns for the markers described in Fig. 2c–f. h, SPRING plots overlaid with gene expression patterns for the markers described in Fig. 3.

Supplementary Figure 4 Quantitative analysis of incisor epithelial kinetics using the TetOn-H2B-GFP system.

a, Frontal images and µCT rendering of the hemi-mandible from control (top), tetOff (middle) and tetON (bottom)-H2B-GFP mice. tetOff (middle) mice possessed chalky white incisors with blunted tips and missing enamel (yellow arrowhead), as compared to the yellow, sharp and intact incisors in controls (top) and TetOn-H2B-GFP (bottom) mice. µCT analysis shows that enamel is compromised (pink arrowheads) in the tetOff-H2B-GFP mice. Coronal sections at a region distal to the first molar [red dotted lines in (b)] show reduced density (blue arrowheads) and defective enamel (red arrowhead) in TetOff-H2B-GFP mice. Scale bars, 5 mm (in a- 3D),1 mm (in coronal section). b, Quantification of the incisor volume (top) and enamel density (bottom) assessed by µCT. N=3 animals. Bar charts indicate the mean of samples and error bars represent ± S.D. of the mean. Data were analyzed using unpaired t-test with Welch's correction. Significance was taken as P<0.05 with a confidence interval of 95%. * P< 0.05. c, Immunostaining for GFP in the incisor growth region. Prior to Dox induction, the incisor epithelium in TetOn-H2B-GFP is GFP-negative. Scale bar, 100 µm. d, Immunostaining for GFP in the incisor growth region of TetOn-H2B-GFP mice pulsed (Dox) between postnatal day (P) 2 and 4 and chased for 8 weeks. The OEE and the adjacent SR maintained high levels of GFP, which persisted for at least 8 weeks. Scale bars, 100 µm. e, Immunostaining for PAX7 (red) and GFP (green) in the anterior tibialis muscle of TetOn-H2B-GFP mice. Scale bars, 10 µm. f, Immunostaining for GFP in the incisor growth region of TetOn-H2B-GFP mice pulsed (Dox) for 21 days starting at 8 weeks of age and chased for 6 (top) and 12 (bottom) months. The IEE and the rest of the proliferating regions of the incisor epithelium are negative for GFP (yellow insets), while the lower regions of the OEE and the adjacent SR (orange insets) maintain high levels of GFP. The layers of cells ventral to the SI and the distal ameloblasts layer (pink insets) also contain GFP-positive cells. Scale bars, 100 µm.

Supplementary Figure 5 Extended analysis of the transcriptomic signatures of the incisor epithelium during recovery.

a, TUNEL (red) and EdU (green) staining revealed high levels of apoptotic cell death in the cycling regions of the incisor epithelium during 5FU treatment (day 2), while the non-cycling regions remained unaffected. Yellow (IEE) and orange (OEE) boxed area are enlarged on the bottom. Scale bars, 50 µm. b, c, SPRING plots combining control and recovering incisor epithelial cell dataset. Colors are the same as depicted in Fig. 4g, h. d, Top 20 enriched genes for each of the cell populations defined in the injury condition (top map) show similar expression patterns (annotated in doted green vertical lines) as their counterparts in control (bottom map). Cells are colored as in Fig. 1d for control (bottom right), and by classification results (see “Defining cell populations observed in single-cell RNAseq data” in methods) for injury (top right). e-g, SPRING plot shown in panel c with the two conditions displayed side-by-side and colored by gene expression of markers shown Fig. 4i-k: cycling cell markers Ccnb1 and Birc5, pre-ameloblasts markers Igfbpl1 and Ank2, and injury-enriched markers Sfrp5 and Cldn10. h, I, Equivalents of Supplementary Fig. 3e, f for Class 1 cells in injury.

Supplementary Figure 6 Examination of the effect of 5FU treatment on Notch signaling.

a, Notch1 activity as assessed by NICD immunostaining. Scale bars, 100 μm. b, RNAscope in situ hybridization shows changes in expression patterns of Ank2 (Class 2) and Ccnb1 (Class1) during recovery from 5FU. Scale bars, 100 μm. c, RNAscope in situ hybridization shows changes in expression patterns of Notch1 and target genes during recovery from 5FU. Scale bars, 100 μm. d, Heatmap showing the average expression of Notch signaling pathway components per Class 3 populations in control and recovery day 3 states.

Supplementary Figure 7 Assessment of the effect of 5FU treatment on Notch1-expressing cells.

a, Schematic of 5FU and tamoxifen (Tam) treatment schedule. Notch1CreER;R26tdTomato mice were dosed three times with Tam to lineage-trace Notch1-expressing cells and their descendants in steady state and during recovery from 5FU injury. b, Immunostaining for tdTomato (red) in control (left) and 5FU-treated (right two panels) mice analyzed 5 days and 21 days after the Tam induction. Scale bar,100 µm. c, Schematic of mechanical injury schedule. d, The distal portion of the incisor of Notch1CreER;R26mTmG mice was mechanically severed and the proximal region grown under the kidney capsule of a host mouse to lineage-trace Notch1-expressing cells and their descendants in steady state and during recovery from mechanical injury. Intact incisors were used as control. Bottom panel: Confocal images of control (left) and injured (right) mice analyzed 10 days after the treatment. Cells that underwent Cre recombination were permanently labeled with membrane GFP (mG). Very few cells reached the ameloblast layer in control (yellow arrowheads), while numerous traced cells reached the ameloblast layer during recovery from mechanical injury (yellow arrowheads). Scale bars,100 µm. e, TUNEL (red) and GFP (mG;green) staining revealed high levels of apoptotic cell death in the SI region of the incisor epithelium of Notch1CreER; RosaDTA/+ 2 days after TAM induction. Scale bar,100 µm. f, Schematic of the hierarchy of the three cell classes during homeostasis and injury repair. Actively cycling IEE cells (blue) generate both the enamel-producing ameloblasts (grey) and the adjacent non-ameloblast epithelial cells (orange). During injury repair, additional progenitors enter cell cycle, and SI cells are converted to become ameloblasts.

Supplementary information

Supplementary Information

Supplementary Figures 1–7, titles and legends for Supplementary tables.

Reporting Summary

Supplementary Table 1

Top 100 enriched genes per cluster in the control dataset. Fold changes are indicated for each gene, and all values are ordered per cluster.

Supplementary Table 2

Top 20 enriched genes in the control cells after the cell cycle effect was regressed out. Fold changes are indicated for each gene and all values are ordered per cluster.

Supplementary Table 3

Expression matrix of genes in the ameloblast trajectory.

Supplementary Table 4

Expression matrix of key markers used to identify cell populations in the non-ameloblast epithelium.

Supplementary Table 5

Summary of the GSEA for each cluster of the control dataset. Results are presented as individual tabs for each cluster.

Supplementary Table 6

Top 20 enriched genes in the clusters of the recovery dataset. Fold changes are indicated for each gene, and all values are ordered per cluster.

Supplementary Table 7

GSEA of control and recovery datasets.

Supplementary Table 8

Top 20 enriched genes in the recovering dataset after the cell cycle effect was regressed out. Fold changes are indicated for each gene and all values are ordered per cluster.

Supplementary Table 9

Expression matrix for genes of the Notch signaling pathway in control and recovery datasets.

Supplementary Table 10

Hyperlinks to Interactive explorers of resulting SPRING plots.

Supplementary Table 11

Source data.

Supplementary Video 1

Two-photon time-lapse live imaging shows Notch1-expressing cells in the ameloblast region of control explant.

Supplementary Video 2

Two-photon time-lapse live imaging shows Notch1-expressing cells in the ameloblast region of 5FU treated explant.

Supplementary Video 3

Two-photon time-lapse live imaging shows Notch1-expressing cells in the IEE of control explant.

Supplementary Video 4

Two-photon time-lapse live imaging shows Notch1-expressing cells in the IEE of 5FU treated explant.

Supplementary Note

Additional methods and theory.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Sharir, A., Marangoni, P., Zilionis, R. et al. A large pool of actively cycling progenitors orchestrates self-renewal and injury repair of an ectodermal appendage. Nat Cell Biol 21, 1102–1112 (2019). https://doi.org/10.1038/s41556-019-0378-2

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41556-019-0378-2

This article is cited by

Search

Quick links

Nature Briefing: Translational Research

Sign up for the Nature Briefing: Translational Research newsletter — top stories in biotechnology, drug discovery and pharma.

Get what matters in translational research, free to your inbox weekly. Sign up for Nature Briefing: Translational Research