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:

Stem cell plasticity enables hair regeneration following Lgr5+ cell loss

Abstract

Under injury conditions, dedicated stem cell populations govern tissue regeneration. However, the molecular mechanisms that induce stem cell regeneration and enable plasticity are poorly understood. Here, we investigate stem cell recovery in the context of the hair follicle to understand how two molecularly distinct stem cell populations are integrated. Utilizing diphtheria-toxin-mediated cell ablation of Lgr5+ (leucine-rich repeat-containing G-protein-coupled receptor 5) stem cells, we show that killing of Lgr5+ cells in mice abrogates hair regeneration but this is reversible. During recovery, CD34+ (CD34 antigen) stem cells activate inflammatory response programs and start dividing. Pharmacological attenuation of inflammation inhibits CD34+ cell proliferation. Subsequently, the Wnt pathway controls the recovery of Lgr5+ cells and inhibition of Wnt signalling prevents Lgr5+ cell and hair germ recovery. Thus, our study uncovers a compensatory relationship between two stem cell populations and the underlying molecular mechanisms that enable hair follicle regeneration.

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: Labelling and deletion of Lgr5+ bulge and hair germ stem cells.
Figure 2: Ablation of Lgr5+ cells inhibits telogen-to-anagen transition and hair growth.
Figure 3: Discontinuation of Lgr5+ cell ablation restores hair growth.
Figure 4: CD34+ stem cell activation and inflammation signature.
Figure 5: Attenuation of inflammation inhibits CD34+ stem cell activation.
Figure 6: CD34+ stem cells give rise to Lgr5+ cells.
Figure 7: BMP and Wnt signalling play opposite roles during Lgr5+ cell recovery.
Figure 8: Wnt signalling is required for Lgr5+ cell recovery.

Similar content being viewed by others

Accession codes

Primary accessions

Gene Expression Omnibus

References

  1. Hsu, Y. C., Pasolli, H. A. & Fuchs, E. Dynamics between stem cells, niche, and progeny in the hair follicle. Cell 144, 92–105 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  2. Mascre, G. et al. Distinct contribution of stem and progenitor cells to epidermal maintenance. Nature 489, 257–262 (2012).

    CAS  PubMed  Google Scholar 

  3. Ritsma, L. et al. Intestinal crypt homeostasis revealed at single-stem-cell level by in vivo live imaging. Nature 507, 362–365 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  4. Rompolas, P. & Greco, V. Stem cell dynamics in the hair follicle niche. Semin. Cell Dev. Biol. 25–26, 34–42 (2014).

    PubMed  Google Scholar 

  5. Levy, V., Lindon, C., Harfe, B. D. & Morgan, B. A. Distinct stem cell populations regenerate the follicle and interfollicular epidermis. Dev. Cell 9, 855–861 (2005).

    CAS  PubMed  Google Scholar 

  6. Page, M. E., Lombard, P., Ng, F., Gottgens, B. & Jensen, K. B. The epidermis comprises autonomous compartments maintained by distinct stem cell populations. Cell Stem Cell 13, 471–482 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  7. Ito, M. et al. Stem cells in the hair follicle bulge contribute to wound repair but not to homeostasis of the epidermis. Nat. Med. 11, 1351–1354 (2005).

    CAS  PubMed  Google Scholar 

  8. Plikus, M. V. et al. Epithelial stem cells and implications for wound repair. Semin. Cell Dev. Biol. 23, 946–953 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  9. Schepeler, T., Page, M. E. & Jensen, K. B. Heterogeneity and plasticity of epidermal stem cells. Development 141, 2559–2567 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  10. Barker, N. et al. Identification of stem cells in small intestine and colon by marker gene Lgr5. Nature 449, 1003–1007 (2007).

    CAS  PubMed  Google Scholar 

  11. Jaks, V. et al. Lgr5 marks cycling, yet long-lived, hair follicle stem cells. Nat. Genet. 40, 1291–1299 (2008).

    CAS  PubMed  Google Scholar 

  12. Junttila, M. R. et al. Targeting LGR5+ cells with an antibody-drug conjugate for the treatment of colon cancer. Sci. Transl. Med. 7, 314ra186 (2015).

    PubMed  Google Scholar 

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

    CAS  PubMed  Google Scholar 

  14. Yan, K. S. et al. The intestinal stem cell markers Bmi1 and Lgr5 identify two functionally distinct populations. Proc. Natl Acad. Sci. USA 109, 466–471 (2012).

    CAS  PubMed  Google Scholar 

  15. Tian, H. et al. A reserve stem cell population in small intestine renders Lgr5-positive cells dispensable. Nature 478, 255–259 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  16. Metcalfe, C., Kljavin, N. M., Ybarra, R. & de Sauvage, F. J. Lgr5+ stem cells are indispensable for radiation-induced intestinal regeneration. Cell Stem Cell 14, 149–159 (2014).

    CAS  PubMed  Google Scholar 

  17. Trempus, C. S. et al. Enrichment for living murine keratinocytes from the hair follicle bulge with the cell surface marker CD34. J. Invest. Dermatol. 120, 501–511 (2003).

    CAS  PubMed  Google Scholar 

  18. Rompolas, P. et al. Live imaging of stem cell and progeny behaviour in physiological hair-follicle regeneration. Nature 487, 496–499 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  19. Blanpain, C., Lowry, W. E., Geoghegan, A., Polak, L. & Fuchs, E. Self-renewal, multipotency, and the existence of two cell populations within an epithelial stem cell niche. Cell 118, 635–648 (2004).

    CAS  PubMed  Google Scholar 

  20. Genander, M. et al. BMP signaling and its pSMAD1/5 target genes differentially regulate hair follicle stem cell lineages. Cell Stem Cell 15, 619–633 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  21. Greco, V. et al. A two-step mechanism for stem cell activation during hair regeneration. Cell Stem Cell 4, 155–169 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  22. Huelsken, J., Vogel, R., Erdmann, B., Cotsarelis, G. & Birchmeier, W. β-Catenin controls hair follicle morphogenesis and stem cell differentiation in the skin. Cell 105, 533–545 (2001).

    CAS  PubMed  Google Scholar 

  23. Kandyba, E. et al. Competitive balance of intrabulge BMP/Wnt signaling reveals a robust gene network ruling stem cell homeostasis and cyclic activation. Proc. Natl Acad. Sci. USA 110, 1351–1356 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  24. Kobielak, K., Stokes, N., de la Cruz, J., Polak, L. & Fuchs, E. Loss of a quiescent niche but not follicle stem cells in the absence of bone morphogenetic protein signaling. Proc. Natl Acad. Sci. USA 104, 10063–10068 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  25. Zhang, J. et al. Bone morphogenetic protein signaling inhibits hair follicle anagen induction by restricting epithelial stem/progenitor cell activation and expansion. Stem Cells 24, 2826–2839 (2006).

    CAS  PubMed  Google Scholar 

  26. Muller-Rover, S. et al. A comprehensive guide for the accurate classification of murine hair follicles in distinct hair cycle stages. J. Invest. Dermatol. 117, 3–15 (2001).

    CAS  PubMed  Google Scholar 

  27. Mardaryev, A. N. et al. Lhx2 differentially regulates Sox9, Tcf4 and Lgr5 in hair follicle stem cells to promote epidermal regeneration after injury. Development 138, 4843–4852 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  28. Jensen, K. B. et al. Lrig1 expression defines a distinct multipotent stem cell population in mammalian epidermis. Cell Stem Cell 4, 427–439 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  29. Muller-Rover, S. et al. E- and P-cadherin expression during murine hair follicle morphogenesis and cycling. Exp. Dermatol. 8, 237–246 (1999).

    CAS  PubMed  Google Scholar 

  30. Wang, L., Siegenthaler, J. A., Dowell, R. D. & Yi, R. Foxc1 reinforces quiescence in self-renewing hair follicle stem cells. Science 351, 613–617 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  31. Balli, D. et al. Foxm1 transcription factor is required for lung fibrosis and epithelial-to-mesenchymal transition. EMBO J. 32, 231–244 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  32. Hinata, K., Gervin, A. M., Jennifer Zhang, Y. & Khavari, P. A. Divergent gene regulation and growth effects by NF-κB in epithelial and mesenchymal cells of human skin. Oncogene 22, 1955–1964 (2003).

    CAS  PubMed  Google Scholar 

  33. Bouchard, C., Staller, P. & Eilers, M. Control of cell proliferation by Myc. Trends Cell Biol. 8, 202–206 (1998).

    CAS  PubMed  Google Scholar 

  34. Lawrence, T. The nuclear factor NF-κB pathway in inflammation. Cold Spring Harb. Perspect. Biol. 1, a001651 (2009).

    PubMed  PubMed Central  Google Scholar 

  35. Robarge, K. D. et al. GDC-0449—A potent inhibitor of the hedgehog pathway. Bioorg. Med. Chem. Lett. 19, 5576–5581 (2009).

    CAS  PubMed  Google Scholar 

  36. Del Pozo Martin, Y. et al. Mesenchymal cancer cell-stroma crosstalk promotes niche activation, epithelial reversion, and metastatic colonization. Cell Rep. 13, 2456–2469 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  37. Liu, J. et al. Targeting Wnt-driven cancer through the inhibition of Porcupine by LGK974. Proc. Natl Acad. Sci. USA 110, 20224–20229 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  38. Horsley, V., Aliprantis, A. O., Polak, L., Glimcher, L. H. & Fuchs, E. NFATc1 balances quiescence and proliferation of skin stem cells. Cell 132, 299–310 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  39. Plikus, M. V. et al. Self-organizing and stochastic behaviors during the regeneration of hair stem cells. Science 332, 586–589 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  40. Plikus, M. V. et al. Cyclic dermal BMP signalling regulates stem cell activation during hair regeneration. Nature 451, 340–344 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  41. Rendl, M., Lewis, L. & Fuchs, E. Molecular dissection of mesenchymal-epithelial interactions in the hair follicle. PLoS Biol. 3, e331 (2005).

    PubMed  PubMed Central  Google Scholar 

  42. Karin, M. & Clevers, H. Reparative inflammation takes charge of tissue regeneration. Nature 529, 307–315 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  43. Perez-Moreno, M. et al. p120-catenin mediates inflammatory responses in the skin. Cell 124, 631–644 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

  44. Kopp, E. & Ghosh, S. Inhibition of NF-κB by sodium salicylate and aspirin. Science 265, 956–959 (1994).

    CAS  PubMed  Google Scholar 

  45. Yin, M. J., Yamamoto, Y. & Gaynor, R. B. The anti-inflammatory agents aspirin and salicylate inhibit the activity of IκB kinase-β. Nature 396, 77–80 (1998).

    CAS  PubMed  Google Scholar 

  46. Hsu, Y. C., Li, L. & Fuchs, E. Transit-amplifying cells orchestrate stem cell activity and tissue regeneration. Cell 157, 935–949 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  47. Langton, A. K., Herrick, S. E. & Headon, D. J. An extended epidermal response heals cutaneous wounds in the absence of a hair follicle stem cell contribution. J. Invest. Dermatol. 128, 1311–1318 (2008).

    CAS  PubMed  Google Scholar 

  48. Tetteh, P. W. et al. Replacement of lost Lgr5-positive stem cells through plasticity of their enterocyte-lineage daughters. Cell Stem Cell 18, 203–213 (2016).

    CAS  PubMed  Google Scholar 

  49. Rompolas, P., Mesa, K. R. & Greco, V. Spatial organization within a niche as a determinant of stem-cell fate. Nature 502, 513–518 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  50. Claudinot, S., Nicolas, M., Oshima, H., Rochat, A. & Barrandon, Y. Long-term renewal of hair follicles from clonogenic multipotent stem cells. Proc. Natl Acad. Sci. USA 102, 14677–14682 (2005).

    CAS  PubMed  PubMed Central  Google Scholar 

  51. Jensen, U. B. et al. A distinct population of clonogenic and multipotent murine follicular keratinocytes residing in the upper isthmus. J. Cell Sci. 121, 609–617 (2008).

    CAS  PubMed  Google Scholar 

  52. Ito, M., Kizawa, K., Hamada, K. & Cotsarelis, G. Hair follicle stem cells in the lower bulge form the secondary germ, a biochemically distinct but functionally equivalent progenitor cell population, at the termination of catagen. Differentiation 72, 548–557 (2004).

    PubMed  Google Scholar 

  53. Kishimoto, J., Burgeson, R. E. & Morgan, B. A. Wnt signaling maintains the hair-inducing activity of the dermal papilla. Genes Dev. 14, 1181–1185 (2000).

    CAS  PubMed  PubMed Central  Google Scholar 

  54. Choi, Y. S. et al. Distinct functions for Wnt/β-catenin in hair follicle stem cell proliferation and survival and interfollicular epidermal homeostasis. Cell Stem Cell 13, 720–733 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  55. Adam, R. C. et al. Pioneer factors govern super-enhancer dynamics in stem cell plasticity and lineage choice. Nature 521, 366–370 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  56. Yu, Z. et al. The extent of the uptake of plasmid into the skin determines the immune responses induced by a DNA vaccine applied topically onto the skin. J. Pharm. Pharmacol. 63, 199–205 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  57. Brown, R. L. & Greenhalgh, D. G. Mouse models to study wound closure and topical treatment of infected wounds in healing-impaired and normal healing hosts. Wound Repair Regen. 5, 198–204 (1997).

    CAS  PubMed  Google Scholar 

  58. Lafkas, D. et al. Therapeutic antibodies reveal Notch control of transdifferentiation in the adult lung. Nature 528, 127–131 (2015).

    CAS  PubMed  Google Scholar 

  59. Wu, T. D. & Nacu, S. Fast and SNP-tolerant detection of complex variants and splicing in short reads. Bioinformatics 26, 873–881 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  60. Law, C. W., Chen, Y., Shi, W. & Smyth, G. K. voom: precision weights unlock linear model analysis tools for RNA-seq read counts. Genome Biol. 15, R29 (2014).

    PubMed  PubMed Central  Google Scholar 

  61. Ritchie, M. E. et al. limma powers differential expression analyses for RNA-sequencing and microarray studies. Nucleic Acids Res. 43, e47 (2015).

    PubMed  PubMed Central  Google Scholar 

  62. Srinivasan, K. et al. Untangling the brain’s neuroinflammatory and neurodegenerative transcriptional responses. Nat. Commun. 7, 11295 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  63. Yaari, G., Bolen, C. R., Thakar, J. & Kleinstein, S. H. Quantitative set analysis for gene expression: a method to quantify gene set differential expression including gene–gene correlations. Nucleic Acids Res. 41, e170 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  64. 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).

    CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We are grateful to the Animal Unit, particularly W. Ortiz, the Center for Advanced Light Microscopy, especially L. Komuves, and the Fluorescence Activated Cell Sorting (FACS) laboratory at Genentech for technical help and advice. We thank P. Sotiropoulou at the Universite libre de Bruxelles for her advice on CD34+ cell FACS sorting. We thank H. Sharpe and C. Metcalfe for critical reading of the manuscript.

Author information

Authors and Affiliations

Authors

Contributions

J.D.H. designed and conducted the majority of the experiments, performed the data analyses and wrote the manuscript. B.B. carried out the ISH and DAB stainings and co-wrote the manuscript. A.V.K., N.M.K., F.d.S.e.M. and B.A. helped with the in vivo experiments. H.K. analysed the ISH results. Z.M. managed the library generation and RNA-sequencing. R.P. analysed the RNA-sequencing data. F.J.d.S. supervised the project and co-wrote the manuscript.

Corresponding author

Correspondence to Frederic J. de Sauvage.

Ethics declarations

Competing interests

All authors are employees of Genentech, Inc. and some own shares of Roche.

Integrated supplementary information

Supplementary Figure 1 Lgr5+ cell ablation in the skin of the Lgr5DTR/+ mouse model.

(a) Schematic representation of the Lgr5DTR/+ mouse model expressing a diphtheria toxin receptor (DTR)/enhanced green fluorescent protein (eGFP) fusion construct in the endogenous Lgr5 locus. Lgr5+ cells are depicted in green and are ablated upon diphtheria toxin treatment (DT). (b) Immunofluorescence co-stainings for GFP and the apoptosis marker cleaved Caspase 3 (Casp3) in Lgr5+/+ (wt) and Lgr5DTR/+ mice 24 h after administration of one dose of DT (100 μg kg−1 bodyweight) or 0.9% NaCl. Scale bars, 20 μm. (c) H&E staining on skin from untreated 3.5 week old Lgr5DTR/+ mice. Rectangle marks region shown below in high magnification. Scale bars, 50 μm. (d) Immunofluorescence staining for the hair germ stem cell marker P-Cad on skin from untreated 3.5 week old Lgr5DTR/+ mice. Asterisks mark autofluorescence from hair shafts. Scale bars, 50 μm. Where visible, the dermal papilla is separated from the hair follicle epithelium by a dotted line.

Supplementary Figure 2 Deletion of Lgr5+ cells during anagen results in the collapse of the bulb.

(a) Immunofluorescence staining for CD34 and GFP on Lgr5DTR/+ skin during anagen phase of the hair cycle. (b) Schematic depicting the dosing regimen of 5 week old Lgr5DTR/+ mice treated every 48 h with DT (100 μg kg−1 bodyweight) or 0.9% NaCl. DT/NaCl treatment was discontinued after three doses and tissue was harvested at d1 or d11. Rectangular bars show hair cycle stages of control mice at the indicated age. Ana, Anagen; Cat, Catagen; Tel, Telogen. (c,e) H&E staining on (c) d1 and (e) d11 skin from Lgr5DTR/+ mice treated with DT or NaCl. Rectangles mark regions shown below in high magnification. (d,f) Immunofluorescence staining for CD34 on (d) d1 and (f) d11 skin from Lgr5DTR/+ mice treated with DT or NaCl. Where visible, the dermal papilla is separated from the hair follicle epithelium by a dotted line. Bu, bulge. Scale bars, 50 μm.

Supplementary Figure 3 Lgr5+ cells are not required for wound healing.

(a) Schematic depicting the dosing regimen of 3.5 week old Lgr5+/+ and Lgr5DTR/+ mice treated every 48 h with DT (100 μg kg−1 bodyweight) or 0.9% NaCl. Two full-thickness wounds (6 mm diameter) were induced on d1 of the experiment and wound healing was analyzed at d8. (b) Brightfield images and H&E stainings on d8 skin from wounded Lgr5+/+ and Lgr5DTR/+ mice treated with DT or NaCl. Scale bars, 50 μm. (c) Quantification of the wound area (in cm2) at d8 on the back of Lgr5+/+ and Lgr5DTR/+ mice treated with DT or NaCl. Each data point represents the results from one mouse. n = 4 mice. Center values, mean; Error bars, s.e.m.; n.s., not significant (unpaired t-test).

Supplementary Figure 4 Lgr5+ cell deletion delays immediate but not long-term anagen induction.

(a) Schematic depicting the dosing regimen of 8 week old Lgr5+/+ and Lgr5DTR/+ mice in telogen treated every 48 h with DT (100 μg kg−1 bodyweight) or 0.9% NaCl. Hair plucking was performed 24 h after the last DT or saline injection. DT and NaCl treatment was discontinued after three doses and tissue was harvested at d25. Rectangular bar shows hair cycle stage of control mice at the indicated age. Tel, Telogen. (b) H&E staining on d25 skin from Lgr5+/+ and Lgr5DTR/+ mice treated with 3 doses of DT or NaCl. (c) Schematic showing the dosing regimen of 8 week old Lgr5+/+ and Lgr5DTR/+ mice in telogen treated every 48 h with DT (100 μg kg−1 bodyweight) or 0.9% NaCl. DT and NaCl treatment was discontinued after three doses and tissue was harvested at d14 and at d31. Rectangular bars show hair cycle stages of control mice at the indicated age. Ana, Anagen; Tel, Telogen. (d) H&E staining on d14 skin of Lgr5+/+ and Lgr5DTR/+ mice treated with 3 doses of DT or NaCl. (e) Immunofluorescence staining for CD34 and P-Cad on d14 skin from Lgr5+/+ and Lgr5DTR/+ mice treated with 3 doses of DT or NaCl. (f) H&E staining on d31 skin from Lgr5+/+ and Lgr5DTR/+ mice treated with 3 doses of DT or NaCl. Where visible, the dermal papilla is separated from the hair follicle epithelium by a dotted line. In (b,d,f), scale bars, 50 μm. In (e), scale bars, 20 μm.

Supplementary Figure 5 CD34+ cell divisions during Lgr5+ cell recovery.

(af) Immunofluorescence co-staining for CD34 and the cell division marker phospho-Histone H3 (pH3) on skin from Lgr5DTR/+ mice treated with 3 doses of (a) NaCl or (bf) DT at (a,b) d1, (c) d2, (d) d4, (e) d7 and (f) d11. Yellow arrows point out pH3+ cells. Asterisks mark autofluorescence from hair shafts. Where visible, the dermal papilla is separated from the hair follicle epithelium by a dotted line. Scale bars, 20 μm.

Supplementary Figure 6 CD34+ cell proliferation does not depend on developmental pathways.

(a) Gene set activity in DT-treated versus saline treated samples for selected gene sets associated with developmental pathways BMP, Wnt and Hh, unchanged in CD34+ hair follicle stem cells of DT-treated Lgr5DTR/+ mice at d1. For each gene set the mean and 95% confidence intervals are plotted and coloured according to their false discovery rate (FDR)-adjusted P-values when compared to zero. n = 3 mice. (b) Schematic showing the dosing regimen of 8 week old Lgr5DTR/+ mice treated with three doses of DT (100 μg kg−1 bodyweight) every 48 h. Starting 24 h before the first DT administration, mice were treated with either vehicle control (MCT), vismodegib (Vismo, 75 mg kg−1 bodyweight), LDN-193189 (LDN, 35 mg kg−1 bodyweight) or LGK-974 (LGK, 5 mg kg−1 bodyweight) twice daily (b.i.d.). Rectangular bars show hair cycle stage of control mice at the indicated age. Tel, Telogen. (c) Immunofluorescence co-staining for CD34 and the proliferation marker Ki67 on d1 skin from DT-treated Lgr5DTR/+ mice that received either MCT (upper left), Vismo (upper right), LDN (lower left) or LGK (lower right). (d) Histogram depicting the percentage of Ki67+ cells among CD34+ stem cells at d1 of MCT, Vismo, LDN or LGK treated Lgr5DTR/+ mice (DT-treated). n = 3 mice. Center values, mean; Error bars, s.e.m. (e) Gli1 in situ hybridisation on d1 hair follicles of DT treated Lgr5DTR/+ mice that received either MCT or Vismo. (f) 3,3′-Diaminobenzidine (DAB) staining for phospho-SMAD1/5 (p-SMAD1/5) on d1 hair follicles from DT treated Lgr5DTR/+ mice that received either MCT or LDN. (g) Axin2 in situ hybridisation on d1 hair follicles of DT treated Lgr5DTR/+ mice that received either MCT or LGK. Scale bars, 20 μm.

Supplementary Figure 7 Wnt inhibition blocks hair germ recovery, but has no effect on the physiological hair germ.

(a,b) DAB staining for (a) p-SMAD1/5, (b) β-Catenin (β-Cat) on d7 skin of Lgr5DTR/+ mice during the Lgr5GFP cell recovery phase. In (b), counterstain: haematoxylin. HG, Hair germ. Scale bars, 20 μm. (c) Wnt5a (upper panels), Wnt10b (lower panels) in situ hybridisation on hair follicles of saline or DT treated Lgr5DTR/+ mice at d7 of Lgr5GFP cell recovery. Scale bars, 10 μm. (d) Dosing regimen of 8 week old Lgr5DTR/+ mice treated with DT (100 μg kg−1 bodyweight, every 48 h), discontinued after three doses and starting 24 h later, mice were treated twice daily (b.i.d.) periorally with MCT (control), vismodegib (Vismo, 75 mg kg−1 bodyweight), LDN-193189 (LDN, 35 mg kg−1 bodyweight) or LGK-974 (LGK, 5 mg kg−1 bodyweight). Skin harvest: d11. Rectangular bar: hair cycle stage of control mice at indicated age. Tel, Telogen. (e) GFP staining on d11 skin from Lgr5DTR/+ mice treated with DT and MCT, Vismo, LDN or LGK. Scale bars, 20 μm. (f) Number of GFP+ cells per hair follicle at d11 in Lgr5DTR/+ mice treated with DT and MCT (n = 4 mice), Vismo (n = 5 mice), LDN (n = 3 mice) or LGK (n = 3 mice). Red circles, individual data points; Center values, mean; Error bars, s.e.m.; n.s., not significant, P = 0.0285 (unpaired t-test). (g) DAB staining for p-SMAD1/5 on skin from DT treated Lgr5DTR/+ mice at d11 that received either MCT or LGK. Scale bars, 20 μm. (h) P-Cad and GFP co-staining on skin from non-DT administered Lgr5DTR/+ mice treated with MCT or LGK-974 (5 mg kg−1 bodyweight) for 11 days b.i.d. Scale bars, 20 μm. (i) Taqman qRT-PCR analysis for Lgr5 and GFP transcript levels (relative to Actb) in skin from non-DT administered Lgr5DTR/+ mice treated with MCT (control) or LGK-974 b.i.d. for 11 days. Histogram: Fold change normalised to mRNA levels in MCT treated Lgr5DTR/+ control mice. n = 4 mice. Center values, mean; Error bars, s.e.m; n.s., not significant (unpaired t-test). Where visible, the dermal papilla is separated from the hair follicle epithelium by a dotted line.

Supplementary information

Supplementary Information

Supplementary Information (PDF 19761 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Hoeck, J., Biehs, B., Kurtova, A. et al. Stem cell plasticity enables hair regeneration following Lgr5+ cell loss. Nat Cell Biol 19, 666–676 (2017). https://doi.org/10.1038/ncb3535

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/ncb3535

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