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:

In vivo transcriptional governance of hair follicle stem cells by canonical Wnt regulators

Abstract

Hair follicle stem cells (HFSCs) regenerate hair in response to Wnt signalling. Here, we unfold genome-wide transcriptional and chromatin landscapes of β-catenin–TCF3/4–TLE circuitry, and genetically dissect their biological roles within the native HFSC niche. We show that during HFSC quiescence, TCF3, TCF4 and TLE (Groucho) bind coordinately and transcriptionally repress Wnt target genes. We also show that β-catenin is dispensable for HFSC viability, and if TCF3/4 levels are sufficiently reduced, it is dispensable for proliferation. However, β-catenin is essential to activate genes that launch hair follicle fate and suppress sebocyte fate determination. TCF3/4 deficiency mimics Wnt–β-catenin-dependent activation of these hair follicle fate targets; TCF3 overexpression parallels their TLE4-dependent suppression. Our studies unveil TCF3/4–TLE histone deacetylases as a repressive rheostat, whose action can be relieved by Wnt–β-catenin signalling. When TCF3/4 and TLE levels are high, HFSCs can maintain stemness, but remain quiescent. When these levels drop or when Wnt–β-catenin levels rise, this balance is shifted and hair regeneration initiates.

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: HFSCs are Wnt-responsive, but do not require β-catenin to exist in a quiescent, undifferentiated state.
Figure 2: Bulge HFSCs can proliferate in vitro without β-catenin.
Figure 3: Engrafted β-catenin-deficient bulge HFSCs make only epidermis and not hair.
Figure 4: β-catenin-deficient bulge HFSCs can become activated in their native niche, but progress to make the wrong fate choice.
Figure 5: β-catenin influences expression of Wnt-responsive genes in bulge HFSCs that are first activated in the hair germ at anagen onset.
Figure 6: Co-occupancy of TCF3 and TCF4 on the bulge HFSC genome.
Figure 7: Within the stem cell niche, elevated TCF3 impairs bulge HFSC activation, while loss-of-TCF3/4 lowers the activation threshold.
Figure 8: TLE proteins act as co-repressors of TCF3/4 in quiescent bulge HFSCs.

Similar content being viewed by others

References

  1. Clevers, H. & Nusse, R. Wnt–β-catenin signaling and disease. Cell 149, 1192–1205 (2012).

    Article  CAS  PubMed  Google Scholar 

  2. Willert, K. & Nusse, R. β-catenin: a key mediator of Wnt signaling. Curr. Opin. Genet. Dev. 8, 95–102 (1998).

    Article  CAS  PubMed  Google Scholar 

  3. Korinek, V. et al. Depletion of epithelial stem-cell compartments in the small intestine of mice lacking Tcf-4. Nat. Genet. 19, 379–383 (1998).

    Article  CAS  PubMed  Google Scholar 

  4. Sato, T. & Clevers, H. Growing self-organizing mini-guts from a single intestinal stem cell: mechanism and applications. Science 340, 1190–1194 (2013).

    Article  CAS  PubMed  Google Scholar 

  5. Willert, K. et al. Wnt proteins are lipid-modified and can act as stem cell growth factors. Nature 423, 448–452 (2003).

    Article  CAS  PubMed  Google Scholar 

  6. Sato, N., Meijer, L., Skaltsounis, L., Greengard, P. & Brivanlou, A.H. Maintenance of pluripotency in human and mouse embryonic stem cells through activation of Wnt signaling by a pharmacological GSK-3-specific inhibitor. Nat. Med. 10, 55–63 (2004).

    Article  CAS  PubMed  Google Scholar 

  7. DasGupta, R. & Fuchs, E. Multiple roles for activated LEF/TCF transcription complexes during hair follicle development and differentiation. Development 126, 4557–4568 (1999).

    CAS  PubMed  Google Scholar 

  8. Merrill, B. J., Gat, U., DasGupta, R. & Fuchs, E. Tcf3 and Lef1 regulate lineage differentiation of multipotent stem cells in skin. Genes Dev. 15, 1688–1705 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Wray, J. et al. Inhibition of glycogen synthase kinase-3 alleviates Tcf3 repression of the pluripotency network and increases embryonic stem cell resistance to differentiation. Nat. Cell Biol. 13, 838–845 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Lyashenko, N. et al. Differential requirement for the dual functions of β-catenin in embryonic stem cell self-renewal and germ layer formation. Nat. Cell Biol. 13, 753–761 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Yi, F., Pereira, L. & Merrill, B.J. Tcf3 functions as a steady-state limiter of transcriptional programs of mouse embryonic stem cell self-renewal. Stem Cells 26, 1951–1960 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Ten Berge, D. et al. Embryonic stem cells require Wnt proteins to prevent differentiation to epiblast stem cells. Nat. Cell Biol. 13, 1070–1075 (2011).

    Article  CAS  PubMed  Google Scholar 

  13. Shy, B. R. et al. Regulation of Tcf7l1 DNA binding and protein stability as principal mechanisms of Wnt–β-catenin signaling. Cell Rep. 4, 1–9 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Yi, F. et al. Opposing effects of Tcf3 and Tcf1 control Wnt stimulation of embryonic stem cell self-renewal. Nat. Cell Biol. 13, 762–70 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Merrill, B. J. et al. Tcf3: a transcriptional regulator of axis induction in the early embryo. Development 131, 263–274 (2004).

    Article  CAS  PubMed  Google Scholar 

  16. Gat, U., DasGupta, R., Degenstein, L. & Fuchs, E. De novo hair follicle morphogenesis and hair tumors in mice expressing a truncated β-catenin in skin. Cell 95, 605–614 (1998).

    Article  CAS  PubMed  Google Scholar 

  17. Nguyen, H. et al. Tcf3 and Tcf4 are essential for long-term homeostasis of skin epithelia. Nat. Genet. 41, 1068–1075 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Nguyen, H., Rendl, M. & Fuchs, E. Tcf3 governs stem cell features and represses cell fate determination in skin. Cell 127, 171–183 (2006).

    Article  CAS  PubMed  Google Scholar 

  19. Wu, C.I. et al. Function of Wnt–β-catenin in counteracting Tcf3 repression through the Tcf3-β-catenin interaction. Development 139, 2118–2129 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Van de Wetering, M. et al. The β-catenin/TCF-4 complex imposes a crypt progenitor phenotype on colorectal cancer cells. Cell 111, 241–250 (2002).

    Article  CAS  PubMed  Google Scholar 

  21. Korinek, V. et al. Constitutive transcriptional activation by a β-catenin-Tcf complex in APC-/- colon carcinoma. Science 275, 1784–1787 (1997).

    Article  CAS  PubMed  Google Scholar 

  22. Angus-Hill, M. L., Elbert, K. M., Hidalgo, J. & Capecchi, M. R. T-cell factor 4 functions as a tumor suppressor whose disruption modulates colon cell proliferation and tumorigenesis. Proc. Natl Acad. Sci. USA 108, 4914–4919 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Tang, W. et al. A genome-wide RNAi screen for Wnt–β-catenin pathway components identifies unexpected roles for TCF transcription factors in cancer. Proc. Natl Acad. Sci. USA 105, 9697–9702 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Hoffman, J. A., Wu, C. I. & Merrill, B. J. Tcf7l1 prepares epiblast cells in the gastrulating mouse embryo for lineage specification. Development 140, 1665–1675 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Trompouki, E. et al. Lineage regulators direct BMP and Wnt pathways to cell-specific programs during differentiation and regeneration. Cell 147, 577–589 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Verzi, M. P. et al. TCF4 and CDX2, major transcription factors for intestinal function, converge on the same cis-regulatory regions. Proc. Natl Acad. Sci. USA 107, 15157–15162 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Boj, S. F. et al. Diabetes risk gene and Wnt effector Tcf7l2/TCF4 controls hepatic response to perinatal and adult metabolic demand. Cell 151, 1595–1607 (2012).

    Article  CAS  PubMed  Google Scholar 

  28. Cole, M. F., Johnstone, S. E., Newman, J. J., Kagey, M. H. & Young, R. A. Tcf3 is an integral component of the core regulatory circuitry of embryonic stem cells. Genes Dev. 22, 746–755 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Cavallo, R. A. et al. Drosophila Tcf and Groucho interact to repress Wingless signalling activity. Nature 395, 604–608 (1998).

    Article  CAS  PubMed  Google Scholar 

  30. Brantjes, H., Roose, J., van De Wetering, M. & Clevers, H. All Tcf HMG box transcription factors interact with Groucho-related co-repressors. Nucleic Acids Res. 29, 1410–1419 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Hurlstone, A. & Clevers, H. T-cell factors: turn-ons and turn-offs. EMBO J. 21, 2303–2311 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Chen, G. & Courey, A. J. Groucho/TLE family proteins and transcriptional repression. Gene 249, 1–16 (2000).

    Article  CAS  PubMed  Google Scholar 

  33. Liu, C. et al. Control of β-catenin phosphorylation/degradation by a dual-kinase mechanism. Cell 108, 837–847 (2002).

    Article  CAS  PubMed  Google Scholar 

  34. Lo, M. C., Gay, F., Odom, R., Shi, Y. & Lin, R. Phosphorylation by the β-catenin/MAPK complex promotes 14-3-3-mediated nuclear export of TCF/POP-1 in signal-responsive cells in C. elegans . Cell 117, 95–106 (2004).

    Article  CAS  PubMed  Google Scholar 

  35. Hikasa, H. et al. Regulation of TCF3 by Wnt-dependent phosphorylation during vertebrate axis specification. Dev. Cell 19, 521–532 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Park, M. H. et al. Phosphorylation of β-catenin at serine 663 regulates its transcriptional activity. Biochem. Biophys. Res. Commun. 419, 543–549 (2012).

    Article  CAS  PubMed  Google Scholar 

  37. Blanpain, C. & Fuchs, E. Epidermal homeostasis: a balancing act of stem cells in the skin. Nat. Rev. Mol. Cell Biol. 10, 207–217 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. 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).

    Article  CAS  PubMed  Google Scholar 

  39. Millar, S. E. Molecular mechanisms regulating hair follicle development. J. Invest. Dermatol. 118, 216–225 (2002).

    Article  CAS  PubMed  Google Scholar 

  40. Schmidt-Ullrich, R. & Paus, R. Molecular principles of hair follicle induction and morphogenesis. Bioessays 27, 247–261 (2005).

    Article  CAS  PubMed  Google Scholar 

  41. 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  PubMed  Google Scholar 

  42. Lustig, B. et al. Negative feedback loop of Wnt signaling through upregulation of conductin/axin2 in colorectal and liver tumors. Mol. Cell Biol. 22, 1184–1193 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Lowry, W. E. et al. Defining the impact of β-catenin/Tcf transactivation on epithelial stem cells. Genes Dev. 19, 1596–1611 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Posthaus, H. et al. β-Catenin is not required for proliferation and differentiation of epidermal mouse keratinocytes. J. Cell Sci. 115, 4587–4595 (2002).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Chen, T. et al. An RNA interference screen uncovers a new molecule in stem cell self-renewal and long-term regeneration. Nature 485, 104–108 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Zhou, P., Byrne, C., Jacobs, J. & Fuchs, E. Lymphoid enhancer factor 1 directs hair follicle patterning and epithelial cell fate. Genes Dev. 9, 700–713 (1995).

    Article  CAS  PubMed  Google Scholar 

  49. Lien, W. H. et al. Genome-wide maps of histone modifications unwind in vivo chromatin states of the hair follicle lineage. Cell Stem Cell 9, 219–232 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Landt, S. G. et al. ChIP-seq guidelines and practices of the ENCODE and modENCODE consortia. Genome Res. 22, 1813–1831 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Zhang, Y. et al. Model-based analysis of ChIP-Seq (MACS). Genome Biol. 9, R137 (2008).

    Article  PubMed  PubMed Central  Google Scholar 

  52. Beronja, S., Livshits, G., Williams, S. & Fuchs, E. Rapid functional dissection of genetic networks via tissue-specific transduction and RNAi in mouse embryos. Nat. Med. 16, 821–827 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Ito, M. et al. Wnt-dependent de novo hair follicle regeneration in adult mouse skin after wounding. Nature 447, 316–320 (2007).

    Article  CAS  PubMed  Google Scholar 

  54. Srinivas, S. et al. Cre reporter strains produced by targeted insertion of EYFP and ECFP into the ROSA26 locus. BMC Dev. Biol. 1, 4 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Kaufman, C. K. et al. GATA-3: an unexpected regulator of cell lineage determination in skin. Genes Dev. 17, 2108–22 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Nowak, J. A., Polak, L., Pasolli, H. A. & Fuchs, E. Hair follicle stem cells are specified and function in early skin morphogenesis. Cell Stem Cell 3, 33–43 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Langmead, B., Trapnell, C., Pop, M. & Salzberg, S. L. Ultrafast and memory-efficient alignment of short DNA sequences to the human genome. Genome Biol. 10, R25 (2009).

    Article  PubMed  PubMed Central  Google Scholar 

  58. Bailey, T. L. et al. MEME SUITE: tools for motif discovery and searching. Nucleic Acids Res. 37, W202–W208 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Machanick, P. & Bailey, T. L. MEME-ChIP: motif analysis of large DNA datasets. Bioinformatics 27, 1696–1697 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Trapnell, C., Pachter, L. & Salzberg, S. L. TopHat: discovering splice junctions with RNA-Seq. Bioinformatics 25, 1105–1111 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Trapnell, C. et al. Transcript assembly and quantification by RNA-Seq reveals unannotated transcripts and isoform switching during cell differentiation. Nat. Biotechnol. 28, 511–555 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank S. Dewell for assistance in high-throughput sequencing (RU Genomics Resource Center), X. Guo for help in ChIP-seq analysis (Zheng laboratory), and S. Mazel, L. Li, S. Semova and S. Tadesse for FACS sorting (RU FACS facility). We also thank Fuchs’ laboratory members N. Stokes, D. Oristian and A. Aldeguer for assistance in mouse research (Fuchs laboratory); A. Rodriguez-Folgueras for assistance in confocal microscopy; and T. Chen, A. Rodriguez-Folgueras, S. Luo and B. Keyes for discussions and comments. W-H.L. was supported by a Harvey L. Karp Postdoctoral Fellowship and a Jane Coffin Child Fellowship. E.F. is an HHMI Investigator. This work was supported by a grant (to E.F.) from the NIH/NIAMS (R01AR31737) and partially by NIH/NIMH (R21MH099452, R01MH073164, D.Z.).

Author information

Authors and Affiliations

Authors

Contributions

W-H.L. and E.F. designed experiments, analysed data and wrote the paper. W-H.L. performed all experiments, collected all data and prepared the figures. L.P. carried out the grafting and lentiviral injection procedure. M.L. analysed RNA-seq data. D.Z. performed ChIP-seq and other bioinformatics analyses. K.L. assisted in performing some experiments during the revision. E.F. supervised the study.

Corresponding author

Correspondence to Elaine Fuchs.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Integrated supplementary information

Supplementary Figure 1 Hair cycle.

(a) During the resting phase (telogen), HFSCs residing in the bulge (Bu) remain in quiescence as the terminally differentiated inner bulge cells express high levels of inhibitory signals (e.g. BMP, FGF18). At the onset of the regenerative phase (anagen), activated HFSCs located in hair germ (HG) proliferate and initiate HF regeneration in response to the activating cues (e.g. BMP inhibitors and Wnts) produced from crosstalk with the underlying mesenchymal stimulus, referred to as the dermal papilla (DP). Soon after, HFSCs in the bulge are also activated. Some activated HFSCs move downward from the bulge along the outer layer of HFs (ORS), creating an inverse gradient of proliferative cells that fuel the continued production of most proliferative, transient amplifying matrix cells (Mx) at the base of full anagen HFs. In response to high levels of Wnt signaling, matrix progenitors in the pre-cortex region (Pre-co) terminally differentiate to form the hair shaft (HS). At the end of anagen, HFs enter a destructive phase (catagen) and the matrix and much of the lower part of the HF undergo apoptosis. As the epithelial strand regresses, DP is drawn upward towards the bulge/HG and the HF re-enters telogen. IFE, interfollicular epidermis; SG, sebaceous gland.

Supplementary Figure 2 Wnt-responsive Bu-HFSCs resemble HG HFSCs that first get activated and proliferate at the transition.

(a) FACS-Gal sorting scheme to purify early anagen Bu-HFSCs. After sorting for Integrin α6 and CD34 surface expression, Bu-HFSCs were further fractionated into Wnt-responsive (LacZ+) and non-responsive (LacZ-) populations. (b) EdU and immunolabeling show that HG cells at the bulge base and in closest proximity to the dermal papilla (DP) stimulus, are the first to proliferate at the telogen→anagen transition. They expand and give rise to the transit-amplifying (TA) matrix; several days later, cells within the bulge begin to proliferate (arrow). Scale bar represents 50 μm.

Supplementary Figure 3 YFP(+) β-cat cKO HFSCs fail to activate at anagen onset, but remain expressing HFSC stemness genes.

(a) Immunolabelling shows that at telogen→anagen when the first sign of proliferation (arrow) is seen in YFP(+) control hair germ cells, YFP(+) β-cat cKO HFSCs remain quiescent. Quantifications of EdU-labeling experiments are at bottom. Scale bar represents 50 μm. Data are reported as average + SD. n=3; **, p<0.01. (b) FACS analysis for YFP(+)/CD34(+) Bu-HFSCs reveals comparable percentages of control (β-cat Het) and β-cat cKO (β-cat cKO) Bu-HFSCs among HFs from eight different pairs of littermates. n=8; NS, not significant. (c) Real-time PCR analysis for HFSC stemness genes. FACS-purified telogen-phase Bu-HFSCs from Het and β-cat cKO mice (10d post RU486-induced ablation). Values are normalized to β-cat Het Bu-HFSC mRNAs. Data are reported as average + SD. n=3. Error bars for qPCR calculated from technical triplicates. Similar results were reproduced in 3 independent experiments.

Supplementary Figure 4 Genes that are differentially expressed depending upon β-catenin status.

(a) qPCR of telogen-phase Bu-HFSCs confirms RNA-seq data from Telo→Ana (see Fig. 5) showing that upon β-catenin ablation, a cohort of genes are also downregulated in quiescent Bu-HFSCs lacking β-catenin. (b) qPCR confirms that upon β-catenin ablation, a smaller subset of genes are upregulated at Telo→Ana within Bu-HFSCs. Note that fatty acid metabolism genes are elevated in β-cat cKO Bu-HFSCs. (c) Depilation-activated β-cat cKO HFSCs show similar gene changes to those seen in Fig. 5b and 4b. Values in (a)-(c) are normalized to β-cat Het Bu-HFSC mRNAs. Data are reported as average + SD. n=3; *, p<0.05. Error bars for qPCR calculated from technical triplicates. Similar results were reproduced in (a) 3, (b) 3 and (c) 2 independent experiments.

Supplementary Figure 5 TCF3 and TCF4 co-target genes.

(a) LEF1 and TCF1 are expressed and nuclear in adult anagen HFs. Mx, matrix; Pre-Co, pre-cortex. Scale bar represents 50 μm. (b) The specificity and quality of TCF3 and TCF4 antibodies were validated by their pull-down ability in HFSC lysates relative to those pull-down with Tcf7l1-null or Tcf7l2-null skin cells, and to IgG controls. Immunoblotting analyses of immunoprecipitations reveal that sufficient amounts of TCF3 and TCF4 proteins in HFSC lysates were pulled down relative to IgG controls. (c) Genomic distribution of TCF3/4 ChIP-seq co-peaks relative to RefSeq transcripts. Promoter: within ±2kb of transcription start site. Downstream: within ±2kb of transcription end site. Gene proximal: within upstream 2-50kb of transcription start site. Gene distal: beyond upstream 50kb of transcription start site. (d) Comparison of the observed TCF3/4-bound peak distribution to the predicted distribution based on the genomic representation of each compartment. (e) Putative transcription factor binding motifs were discovered from de novo motif analysis using MEME-ChIP software59. The percentages of motif occurrences within TCF3/4-bound peaks versus random genomic sequences are indicated for each motif as determined by the program MAST (p<0.0005). p-values are determined by compared to the percentage observed in random sequences. In addition to Lef1/TCF motif, NFATc1 was the top known motif found at TCF3/4-bound peaks. (f) Most enriched gene ontology for the overlapping targets (n=1,126) of TCF3 targets in mESCs (n=3,591; green)28, and TCF3/4 targets in HFSCs (n=3,387; blue).

Supplementary Figure 6 Gain- and loss-of-function for TCF3 and TCF4.

(a) TCF3 overexpression. qPCR (left) and immunoblotting (right) analyses showing elevated TCF3 mRNA and protein in Bu-HFSCs following Doxy induction of K14rtTA/TRE-mycTCF3 mice. Values are normalized to K14rtTA Bu-HFSC mRNAs. Rps16, mRNA control; α-tubulin; protein loading control. (b) Depletion of TCF3 and TCF4 in HFSCs. E18.5 back skins of K15CrePGR/Tcf7l1fl/fl/Tcf7l2−/−/Rosa26−Y FPfl/stop/fl embryos were grafted onto nude mice, and 7 weeks later, when HFs were in telogen, RU486 was administered to generate double knockout (dKO) HFSCs. qPCR analysis with FACS-isolated Bu-HFSCs from RU486-treated K15CrePGR/Tcf7l1+/fl/Tcf7l2+/−/Rosa26−Y FPfl/stop/fl (TCF3/4 Het) and K15CrePGR/Tcf7l1fl/fl/Tcf7l2−/−/Rosa26−Y FPfl/stop/fl (TCF3/4 dKO) grafted skins. (Left) qPCR analysis demonstrates Tcf3 and TCF4 depletion. (Right) Immunostaining of grafts demonstrates Tcf4 depletion and Cre activation (YFP+). (c) Elevated TCF3 prohibits depilation-induced HFSC activation. Doxy was given throughout. One day after depilation, EdU was administered for 24 hours to identify proliferating cells. (Left) Immunostaining for EdU and Bu-HFSC marker CD34. (Right) Quantifications of EdU-labeling experiments. Note that HFs with increased TCF3 display reduced HFSC activation. Scale bars in (b) and (c) represent 50 μm. Data in (a)-(c) are reported as average + SD. n=3; **, p<0.01. Error bars for qPCR calculated from technical triplicates. Similar results were reproduced in (a) 5 and (b) 3 independent experiments.

Supplementary Figure 7 Co-occupancy of TCF3, TCF4 and TLE in quiescent bulge HFSCs.

(a) TLE occupies bulge HFSC DNA locations that are also bound by TCF3 and TCF4. Density maps of TCF3 (blue), TCF4 (orange), TLE (purple) ChIP-seq reads (x-axis, centered on TCF3 peak summits) at TCF3-bound peaks (Y-axis), broken into peaks in promoter and enhancer regions. (b) Overexpression of TLE4 in HFSCs. (Left) Lentiviral construct carrying Doxy-inducible TRE-Tle4 and H2BRFP genes was introduced to E9.5 K14rtTA embryos by in utero infection52, and at adulthood, Doxy was administered according to schematic. (Middle) A transduced (RFP+) HF. Scale bar represents 50 μm. (Right) qPCR analysis of FACS-isolated RFP(+) Bu-HFSCs from Doxy-induced mice. Note elevated Tle4 mRNA levels relative to control. Data are reported as average + SD. n=3; *, p<0.05. Error bars for qPCR calculated from technical triplicates. Similar results were reproduced in 3 independent experiments.

Supplementary Figure 8 Full scan of immunoblots.

Full scan of cropped immunoblot images in Figs, 2b, 8a, 5b and 6a are shown.

Supplementary information

Supplementary Information

Supplementary Information (PDF 2967 kb)

Supplementary Table 1

Supplementary Information (XLS 100 kb)

Supplementary Table 2

Supplementary Information (XLS 32 kb)

Supplementary Table 3

Supplementary Information (XLS 17 kb)

Rights and permissions

Reprints and permissions

About this article

Cite this article

Lien, WH., Polak, L., Lin, M. et al. In vivo transcriptional governance of hair follicle stem cells by canonical Wnt regulators. Nat Cell Biol 16, 179–190 (2014). https://doi.org/10.1038/ncb2903

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/ncb2903

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