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:

Escape of hair follicle stem cells causes stem cell exhaustion during aging

Subjects

Abstract

Stem cell (SC) exhaustion is a hallmark of aging. However, the process of SC depletion during aging has not been observed in live animals, and the underlying mechanism contributing to tissue deterioration remains obscure. We find that, in aged mice, epithelial cells escape from the hair follicle (HF) SC compartment to the dermis, contributing to HF miniaturization. Single-cell RNA-seq and assay for transposase-accessible chromatin using sequencing (ATAC-seq) reveal reduced expression of cell adhesion and extracellular matrix genes in aged HF-SCs, many of which are regulated by Foxc1 and Nfatc1. Deletion of Foxc1 and Nfatc1 recapitulates HF miniaturization and causes hair loss. Live imaging captures individual epithelial cells migrating away from the SC compartment and HF disintegration. This study illuminates a hitherto unknown activity of epithelial cells escaping from their niche as a mechanism underlying SC reduction and tissue degeneration. Identification of homeless epithelial cells in aged tissues provides a new perspective for understanding aging-associated diseases.

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: Aging HFs are characterized by escaped epithelial cells.
Fig. 2: scRNA-seq reveals reduced cell adhesion in aged HF-SCs.
Fig. 3: Downregulation of Foxc1 and Nfatc1 in aged HF-SCs.
Fig. 4: Genetic deletion of Foxc1 and Nfatc1 causes premature hair loss.
Fig. 5: Transcriptomic analysis of dKO HF-SCs.
Fig. 6: Single-cell open-chromatin analysis of HF-SCs reveals the role of FOXC1 and NFATC1.
Fig. 7: Time-lapse imaging captures HF-SCs escaping from the niche in live animals.

Similar content being viewed by others

Data availability

All sequencing data were deposited to NCBI–GEO SuperSeries under accession number GSE133648.

References

  1. Campisi, J. et al. From discoveries in ageing research to therapeutics for healthy ageing. Nature 571, 183–192 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. López-Otín, C., Blasco, M. A., Partridge, L., Serrano, M. & Kroemer, G. The hallmarks of aging. Cell 153, 1194–1217 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  3. Dall’Ara, E. et al. Longitudinal imaging of the ageing mouse. Mech. Ageing Dev. 160, 93–116 (2016).

    Article  PubMed  Google Scholar 

  4. Pittet, M. J. & Weissleder, R. Intravital imaging. Cell 147, 983–991 (2011).

    Article  CAS  PubMed  Google Scholar 

  5. Florian, M. C. et al. A canonical to non-canonical Wnt signalling switch in haematopoietic stem-cell ageing. Nature 503, 392–396 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Ge, Y. et al. The aging skin microenvironment dictates stem cell behavior. Proc. Natl Acad. Sci. USA 117, 5339–5350 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Lay, K., Kume, T. & Fuchs, E. FOXC1 maintains the hair follicle stem cell niche and governs stem cell quiescence to preserve long-term tissue-regenerating potential. Proc. Natl Acad. Sci. USA 113, E1506–E1515 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Matsumura, H. et al. Hair follicle aging is driven by transepidermal elimination of stem cells via COL17A1 proteolysis. Science 351, aad4395 (2016).

    Article  PubMed  CAS  Google Scholar 

  9. Sinha, M. et al. Restoring systemic GDF11 levels reverses age-related dysfunction in mouse skeletal muscle. Science 344, 649–652 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. 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).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Rossi, D. J. et al. Deficiencies in DNA damage repair limit the function of haematopoietic stem cells with age. Nature 447, 725–729 (2007).

    Article  CAS  PubMed  Google Scholar 

  12. Janzen, V. et al. Stem-cell ageing modified by the cyclin-dependent kinase inhibitor p16INK4a. Nature 443, 421–426 (2006).

    Article  CAS  PubMed  Google Scholar 

  13. Sharpless, N. E. & DePinho, R. A. How stem cells age and why this makes us grow old. Nat. Rev. Mol. Cell Biol. 8, 703–713 (2007).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  15. Cho, I. J. et al. Mechanisms, hallmarks, and implications of stem cell quiescence. Stem Cell Rep. 12, 1190–1200 (2019).

    Article  CAS  Google Scholar 

  16. Nakamura-Ishizu, A., Takizawa, H. & Suda, T. The analysis, roles and regulation of quiescence in hematopoietic stem cells. Development 141, 4656–4666 (2014).

    Article  CAS  PubMed  Google Scholar 

  17. Yi, R. Concise review: mechanisms of quiescent hair follicle stem cell regulation. Stem Cells 35, 2323–2330 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  18. Cheng, T. et al. Hematopoietic stem cell quiescence maintained by p21cip1/waf1. Science 287, 1804–1808 (2000).

    Article  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).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Tomasetti, C. & Vogelstein, B. Variation in cancer risk among tissues can be explained by the number of stem cell divisions. Science 347, 78–81 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Garza, L. A. et al. Prostaglandin D2 inhibits hair growth and is elevated in bald scalp of men with androgenetic alopecia. Sci. Transl. Med. 4, 126ra34 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  22. Shimomura, Y. et al. APCDD1 is a novel Wnt inhibitor mutated in hereditary hypotrichosis simplex. Nature 464, 1043–1047 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Pineda, C. M. et al. Intravital imaging of hair follicle regeneration in the mouse. Nat. Protoc. 10, 1116–1130 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Chen, C.-C. et al. Regenerative hair waves in aging mice and extra-follicular modulators follistatin, dkk1, and sfrp4. J. Invest. Dermatol. 134, 2086–2096 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Stuart, T. et al. Comprehensive integration of single-cell data. Cell 177, 1888–1902 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Salzer, M. C. et al. Identity noise and adipogenic traits characterize dermal fibroblast aging. Cell 175, 1575–1590 (2018).

    Article  CAS  PubMed  Google Scholar 

  28. Fujiwara, H. et al. The basement membrane of hair follicle stem cells is a muscle cell niche. Cell 144, 577–589 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Cao, J. et al. The single-cell transcriptional landscape of mammalian organogenesis. Nature 566, 496–502 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Lee, J. et al. Runx1 and p21 synergistically limit the extent of hair follicle stem cell quiescence in vivo. Proc. Natl Acad. Sci. USA 110, 4634–4639 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Osorio, K. M. et al. Runx1 modulates developmental, but not injury-driven, hair follicle stem cell activation. Development 135, 1059–1068 (2008).

    Article  CAS  PubMed  Google Scholar 

  32. 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).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Keyes, B. E. et al. Nfatc1 orchestrates aging in hair follicle stem cells. Proc. Natl Acad. Sci. USA 110, E4950–E4959 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Lien, W.-H. et al. In vivo transcriptional governance of hair follicle stem cells by canonical Wnt regulators. Nat. Cell Biol. 16, 179–190 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Besson, V. et al. PW1 gene/paternally expressed gene 3 (PW1/Peg3) identifies multiple adult stem and progenitor cell populations. Proc. Natl Acad. Sci. USA 108, 11470–11475 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Kimura-Ueki, M. et al. Hair cycle resting phase is regulated by cyclic epithelial FGF18 signaling. J. Invest. Dermatol. 132, 1338–1345 (2012).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Zhang, Y. V., Cheong, J., Ciapurin, N., McDermitt, D. J. & Tumbar, T. Distinct self-renewal and differentiation phases in the niche of infrequently dividing hair follicle stem cells. Cell Stem Cell 5, 267–278 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  41. 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 

  42. Morris, R. J. et al. Capturing and profiling adult hair follicle stem cells. Nat. Biotechnol. 22, 411–417 (2004).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. 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 

  45. Vidal, V. P. et al. Sox9 is essential for outer root sheath differentiation and the formation of the hair stem cell compartment. Curr. Biol. 15, 1340–1351 (2005).

    Article  CAS  PubMed  Google Scholar 

  46. Buenrostro, J. D. et al. Single-cell chromatin accessibility reveals principles of regulatory variation. Nature 523, 486–490 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Buenrostro, J. D., Giresi, P. G., Zaba, L. C., Chang, H. Y. & Greenleaf, W. J. Transposition of native chromatin for fast and sensitive epigenomic profiling of open chromatin, DNA-binding proteins and nucleosome position. Nat. Methods 10, 1213–1218 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Naik, S. et al. Inflammatory memory sensitizes skin epithelial stem cells to tissue damage. Nature 550, 475–480 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Rhee, H., Polak, L. & Fuchs, E. Lhx2 maintains stem cell character in hair follicles. Science 312, 1946–1949 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Fan, X. et al. Single cell and open chromatin analysis reveals molecular origin of epidermal cells of the skin. Dev. Cell 47, 21–37 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Kadaja, M. et al. SOX9: a stem cell transcriptional regulator of secreted niche signaling factors. Genes Dev. 28, 328–341 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Klein, R. H. et al. GRHL3 binding and enhancers rearrange as epidermal keratinocytes transition between functional states. PLoS Genet. 13, e1006745 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  53. de Guzman Strong, C. et al. Lipid defect underlies selective skin barrier impairment of an epidermal-specific deletion of Gata-3. J. Cell Biol. 175, 661–670 (2006).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  54. Pliner, H. A. et al. Cicero predicts cis-regulatory DNA interactions from single-cell chromatin accessibility data. Mol. Cell 71, 858–871 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Huang, X. Z. et al. Inactivation of the integrin β6 subunit gene reveals a role of epithelial integrins in regulating inflammation in the lung and skin. J. Cell Biol. 133, 921–928 (1996).

    Article  CAS  PubMed  Google Scholar 

  56. Trempus, C. S. et al. CD34 expression by hair follicle stem cells is required for skin tumor development in mice. Cancer Res. 67, 4173–4181 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Lay, K. et al. Stem cells repurpose proliferation to contain a breach in their niche barrier. eLife 7, e41661 (2018).

  58. Driskell, R. R. et al. Distinct fibroblast lineages determine dermal architecture in skin development and repair. Nature 504, 277–281 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Corces, M. R. et al. An improved ATAC-seq protocol reduces background and enables interrogation of frozen tissues. Nat. Methods 14, 959–962 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Kim, D., Langmead, B. & Salzberg, S. L. HISAT: a fast spliced aligner with low memory requirements. Nat. Methods 12, 357–360 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Li, H. et al. The sequence alignment/map format and SAMtools. Bioinformatics 25, 2078–2079 (2009).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  62. Anders, S., Pyl, P. T. & Huber, W. HTSeq—a Python framework to work with high-throughput sequencing data. Bioinformatics 31, 166–169 (2015).

    Article  CAS  PubMed  Google Scholar 

  63. Love, M. I., Huber, W. & Anders, S. Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol. 15, 550 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  64. Zhou, Y. et al. Metascape provides a biologist-oriented resource for the analysis of systems-level datasets. Nat. Commun. 10, 1523 (2019).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  65. 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  PubMed  PubMed Central  Google Scholar 

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

    Article  PubMed  PubMed Central  Google Scholar 

  67. Langmead, B. & Salzberg, S. L. Fast gapped-read alignment with Bowtie 2. Nat. Methods 9, 357–359 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  69. Heinz, S. et al. Simple combinations of lineage-determining transcription factors prime cis-regulatory elements required for macrophage and B cell identities. Mol. Cell 38, 576–589 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. 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 

  71. Quinlan, A. R. & Hall, I. M. BEDTools: a flexible suite of utilities for comparing genomic features. Bioinformatics 26, 841–842 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Robinson, J. T. et al. Integrative Genomics Viewer. Nat. Biotechnol. 29, 24–26 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Ye, T. et al. seqMINER: an integrated ChIP-seq data interpretation platform. Nucleic Acids Res. 39, e35 (2011).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank T. Cech (University of Colorado Boulder, HHMI), K. Green and R. Lavker (Northwestern University) for comments; J. Siegenthaler (University of Colorado School of Medicine) for Foxc1fl/fl and Foxc1-LacZ mice; E. Fuchs (Rockefeller University, HHMI) for Krt14-H2BGFP and Krt14-Cre mice; D. Sheppard (University of California San Francisco) for Itgb6 -KO mice; H. Fujiwara (RIKEN, Japan) for anti-NPNT and anti-EGFL6 antibodies; M. Allen (University of Colorado Boulder) for discussing bioinformatic analysis; J. Orth (University of Colorado Boulder), J. Lopez and R. Goldmeyer (Olympus) for two-photon imaging; and all members of the Yi laboratory for suggestions. Research reported in this publication was supported by the National Institute of Arthritis and Musculoskeletal and Skin Diseases of the National Institutes of Health under award numbers AR059697, AR066703 and AR071435 (to R.Y.) and by the National Institutes of Health under award number GM125871 (to R.D.). C.Z. was supported by an NCI Predoctoral to Postdoctoral Fellow Transition Award (F99CA253738). The content is solely the responsibility of the authors and does not necessarily represent the official views of the National Institutes of Health.

Author information

Authors and Affiliations

Authors

Contributions

R.Y. conceived the study. C.Z., D.W. and R.Y. designed experiments. C.Z. carried out most experiments and computational analysis with assistance from D.W. for IF staining and imaging. J.W. performed two-photon imaging for some control experiments. L.W. helped to analyze scATAC-seq data. W.Q. generated the Itgb6-KO mouse and provided samples. T.K. generated Foxc1 mouse models. R.D. supervised computational analysis. R.Y. and R.D. were co-mentors to C.Z. R.Y. wrote the manuscript with input from all authors.

Corresponding author

Correspondence to Rui Yi.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Peer review information Nature Aging thanks the anonymous reviewers for their contribution to the peer review of this work.

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

Extended data

Extended Data Fig. 1 Live imaging of escaped cells in aging hair follicles.

a, Two-photon longitudinal tracking of hair follicles in young mice during the anagen to telogen hair cycle. Red numbers designate the same hair follicle in each image. Red dotted lines annotate the bulge region. Scale bar, 50μm. b, Two-photon intravital imaging of hair follicles from young (left panel) and old (middle and right panels) mice. White arrowheads point to miniaturized hair follicles and cells located outside of the HF-SC compartments. Red dotted lines outline miniaturized hair follicles. Scale bar, 50μm. c, Boxplot of the percentage of miniaturized hair follicles, quantified from 3-D scan of live animals. (mean ± s.d.= 0 ± 0 (young), 14 ± 17 (old); n=205 HFs from 5 young mice; n=327 HFs from 3 old mice). d, Representative images of hair follicles with KRT5 and activated caspase 3 (acCas3) signals in young (6~8mo) and old (20mo) mice. (n=50 hair follicles from young mice; n=62 hair follicles from old mice, 3 pairs of mice). Scale bar, 20μm. e-f, Boxplot of number of acCas3+ HFSCs(e) and HG(f) per hair follicle (HFSCs: mean ± s.d.= 0.058 ± 0.31 (young), 0.048 ± 0.22 (old); HG: mean ± s.d.= 0.077 ± 0.33 (young), 0.065 ± 0.31 (old); n=50 hair follicles from young mice; n=62 hair follicles from old mice, 3 pairs of mice). g, 3-D view of hair follicles in 24mo old mice. White arrowheads point to numerous escaped epithelial cells scattering in the dermis. Scale bar, 50μm. h, 3-D view of β4 integrin immunofluorescence signals in 24mo old mice. White arrowheads point to HF-SCs with protruding integrin signals in the new bulge side. Scale bar, 20μm. Data in c, e, f are two-sided t-test.

Source data

Extended Data Fig. 2 Quality control and clustering of single-cell RNA-seq data from young and old mice.

a, Quality control and filtering of single cells from old and young samples. Cells were filtered with detected genes numbers (200<nFeature_RNA<5000), transcripts numbers (nCount_RNA) and mitochondrial percentage (percent.mt < 10). b, Track plot of marker genes for each cluster. c, Table shows cluster names, cell numbers and percentage of cells for each cluster after filtering of old and young scRNAseq data.

Extended Data Fig. 3 Single-cell transcriptomic analysis of old and young skin samples.

a-b, UMAP re-clustering and projection of IFE cells, color coded by sample identity (a) and cluster identity (b). c-d, Feature plots of marker genes for basal progenitor cells (Krt14, Krt5) and suprabasal cells (Krt1, Krt10). e-f, Highly enriched GO terms for upregulated genes in old HFSCs (e) and downregulated genes in old IFE (f). g, Violin plots of selected gens in young and old HF-SC and IFE cell clusters. h, Feature plot of monocle3 pseudotime score of hair follicle cells from old and young mice. i, Violin plot of HFSCs pseudotime score in young and old samples. j, Highly enriched GO terms for HFSCs in the ground state with lower pseudotime score (<5.5).

Extended Data Fig. 4 Transcriptional activity of Foxc1 locus in 15mo old mouse skin.

a, Transcriptional activity of Foxc1 locus (Foxc1-LacZ knockin) is detected in anagen bulge but not detected in telogen bulge. Scale bar, 20μm. b, Transcriptional activity of Foxc1 locus (Foxc1-LacZ knockin) is not detected in telogen bulge. Scale bar, 20μm. c, Robust transcriptional activity of Foxc1 locus (Foxc1-LacZ knockin) is detected in both bulge and IRS regions of anagen hair follicles. Scale bar, 20μm.

Extended Data Fig. 5 Hair follicle miniaturization and loss in dKO mice.

a, Krt5 and Ki67 staining of hair follicles at early anagen (anagen III, P25) in control and dKO mice, arrowheads indicate Ki67+ HF-SCs, Right panel, quantification of Ki67+ HF-SCs per hair follicle (n=30 hair follicles from 3 pairs of mice). Scale bar, 20μm. b, Ki67 staining of hair follicles at late anagen (anagen V-VI) in control, Foxc1 cKO, Nfatc1 cKO, and dKO mice, arrowheads indicate Ki67+ HF-SCs. Scale bar, 20μm. c, Images of hair coat in the same control and dKO mice on P41 (left panel) and P64 (right panel), the right half of back skin was shaved on P41 and imaged again on P64. d, Images of hair coat of control and dKO mice at ~16mo old. e, Two-photon images of hair follicles in control and dKO mice at 12mo old, red arrowheads point to escaped cells outside of hair follicles. Scale bar, 70μm. f, Boxplot of the percentage of miniaturized hair follicles in young, old and dKO mice. (mean ± s.d.= 0 ± 0 (young), 14 ± 17 (old), 77 ± 25 (dKO); 5 young mice; 3 old mice; 5 dKO mice). g, Representative Two-Photon images for the quantification of HFSCs, red asterisks mark HFSCs(g). h, Boxplot of the number of HFSCs per HF in different samples. (mean ± s.d.= 19.33 ± 2.44 (young), 15.33 ± 4.04 (old), 10.95 ± 3.97 (dKO);91HFs, 5 young mice; 122 HFs, 3 old mice; 58 HFs, 5 dKO mice). i, Longitudinal tracking of dKO hair follicles over 26 days. Red numbers indicate the identical hair follicles in each image. Scale bar, 70μm. j, Boxplot of the percentage of HFs undergo regeneration, degeneration and quiescence in dKO samples (mean ± s.d.= 70 ± 24 (regeneration), 25.23 ± 25 (degeneration), 4.12 ± 7.49 (quiescence); 78 HFs from more than 3 mice were tracked for at least 16 days). Data in a, f, h are two-sided t-test.

Source data

Extended Data Fig. 6 Quality control and clustering of single-cell RNA-seq data from control and dKO.

a, Quality control and filtering of single cells from both control and dKO samples at P38. Cells were filtered with detected genes numbers (200<nFeature_RNA<5000), transcripts numbers (nCount_RNA) and mitochondrial percentage (percent.mt < 10). b, Track plot of marker genes for each cluster. c, Table shows cluster names, cell numbers and percentage of cells for each cluster after filtering of both control and dKO single-cell RNA-seq data.

Extended Data Fig. 7 Isolation and transcriptomic analysis of Foxc1 and Nfatc1 single KO and induced dKO HF-SCs.

a, Immunofluorescence staining of Foxc1 and Nfatc1 in Krt15-CrePR induced dKO hair follicles. tdT is tdTomato signals from ROSA26-LSL-tdT allele, indicating Cre+ dKO HF-SCs. Red arrowheads indicate HFSCs without Foxc1 and Nfatc1 signals, white arrowheads indicate inner bulge region, which is negative for tdT. Scale bar, 20μm. b, Highly enriched GO terms of upregulated genes and selected differentially expressed in induced dKO HF-SCs. c, Gating strategy for flow cytometry analysis. d-f, Flow cytometry analysis and quantification of HF-SCs during the first anagen (P28-P31) in control, Krt14-Cre-mediated dKO hair follicles (d), Foxc1 cKO and Nfatc1 cKO hair follicles (e). The rectangle regions are CD34-APChi and Cd49f-PE hi HF-SC populations. Representative plots for 3~5 sets of experiments are shown. f, Flow cytometry analysis of Krt15-CrePR-mediated dKO hair follicles with ROSA26-LSL-tdT allele to mark Cre+ dKO cells. The rectangle regions are CD34-APChi and tdTomatohi populations. Representative plots for 5 sets of experiments are shown.

Extended Data Fig. 8 Single-cell ATAC analysis of Foxc1 and Nfatc1 controlled open chromatin in HF-SCs.

a, tSNE plots of control and dKO total epithelial cells (Krt14-H2bGFP+). The HF-SC populations in each sample are highlighted in blue color and circled. The selected populations show the strongest open chromatin signatures of Cd34, the marker for HF-SC, and the weakest signatures of Gata6, a differentiation marker. b, K-means clustering of control and dKO open chromatin regions from aggregated scATAC-seq data from the HF-SC populations. Cluster 8 is reduced in dKO and cluster 10 is enhanced in dKO. c, Top 3 most highly enriched transcription factor motifs in cluster 8. d-f, Aggregated scATAC-seq tracks of Actg1 (d), Npnt (e) and Col6a1/2 (f) loci annotated with FOXC1 and NFATC1 motifs. Location of FOXC1 (green marks) and NFATC1 (red marks) motifs are indicated. Arrows point to HF-SC-specific open chromatin regions that are lost in dKO and the dashed rectangles mark the TSS of Actg1, Npnt and Col6a1/2, respectively.

Extended Data Fig. 9 Enhancer-promoter interactions are inferred by using aggregated Cicero scores computed from scATAC-seq.

a-f, Enhancer-promoter interactions of Actg1 (a), Bmp2 (b), Krt14 (c), Col6a1 (d), Fgf18 (e) and Itgb6 (f) are illustrated in ctrl IFE, dKO IFE, ctrl HF-SC and dKO HF-SC. The aggregated Cicero score is calculated by the summation of Cicero scores of all enhancer-promoter interactions to the TSS region of each gene. The vertical lines mark the TSS and the dashed line indicates the cutoff of Cicero score (0.2) used for calculation.

Extended Data Fig. 10 Deletion of Itgb6 does not lead to premature hair loss.

a, Hair coat is normal in both control and Itgb6 KO animals at ~9-mo old. b, HF-SC compartment is normal in both control and Itgb6 KO animals at ~9-mo old. Scale bar, 20μm.

Supplementary information

Supplementary Information

Legends and captions for Supplementary Tables and Videos

Reporting Summary

Supplementary Tables

Supplementary Tables 1–9.

Supplementary Video 1

An animated illustration identifies an escaped epithelial cell (KRT5+) in the dermis. The movie is related to Fig. 1h.

Supplementary Video 2

An animated illustration shows a number of abnormally located, escaped cells outside of the bulge region of a regressing HF. The movie is related to Fig. 4j.

Supplementary Video 3

Cellular activities of telogen HFs in control mice. A 5-h time-lapse movie of telogen HFs from control mice (P45). The time interval of each time point was 6 min. No discernible cellular activities including migration and cell division were observed. The entire sequences were looped twice during movie production. The movie is related to Fig. 7a.

Supplementary Video 4

A 5.8-h time-lapse movie of anagen HFs from control mice (P75). The time interval of each time point was 6 min. Limited cellular activities such as migration were observed mostly in HF-SCs (red arrowhead) and IRSs (yellow arrowhead) of growing HFs. The movie is related to Fig. 7b.

Supplementary Video 5

Cellular activities of anagen HFs in dKO mice. A 2.5-h time-lapse movie of anagen HFs from dKO mice. The time interval of each time point was 6 min. Robust cell migration activities were observed. In the upper HF, ORS progenitor cells migrate downward along the surface of the HF; IRS differentiated cells (elongated nuclei) migrate upward in the middle of the HF. In the lower HF, ORS progenitor cells migrate horizontally along the surface of the HF. The entire sequences were looped twice during movie production.

Supplementary Video 6

Escape of HF-SCs in dKO mice. A 4-h time-lapse movie of anagen HFs from dKO mice. The time interval of each time point was 6 min. HF-SCs escaped from a defective niche as described in the following sequence: (1) in the first 30 min, an HF-SC (blue arrow) detached from the bulge region; (2) in the first 2 h, a pair of HF-SCs (red and green arrows) escaped together from the bulge region; they went through a small orifice in the BM, judging by the altered morphology of the nuclei during escape; (3) in the final 30 min, an escaped HF-SC (red arrow) migrated ~16 μm further away from the HF; (4) more HF-SCs followed the escaping cells and migrated toward the breach point. The move is related to Fig. 7c.

Supplementary Video 7

Cellular activities in a disintegrating HF. A 2.5-h time-lapse movie of a miniaturized HF from a dKO mouse. The time interval of each time point was 6 min. Two HF cells (green arrowheads) disintegrated and were released into the dermis. An HF cell (red circle) was dividing in the same miniaturized HF. An escaped epithelial cell (yellow arrowhead) migrated away from the HF. The miniaturized HF was next to an SG and already moved upward. The movie is related to Fig. 7e.

Supplementary Video 8

Cell migration in a miniaturized HF. A 2.5-h time-lapse movie of a miniaturized HF from a dKO mouse. The time interval of each time point was 6 min. Less than 20 cells remained in the HF located in the upper portion of the movie. One cell (red arrowhead) migrated downward, away from the HF. The entire sequences were looped twice during movie production.

Supplementary Video 9

Escaped HF cells scattered in the dermis. A 3.2-h time-lapse movie of disintegrating HFs from dKO (~15-mo-old) mice. The time interval of each time point was 6 min. In the HF located in the lower right of the movie, epithelial cells were released from the HF to the dermis. In the upper part of the movie, a disintegrating HF had loosely packed HF cells, with several of them scattering in the nearby dermis. The entire sequences were looped twice during movie production. The movie is related to Fig. 7f.

Source data

Source Data Fig. 1

Statistical source data for Fig. 1c–e.

Source Data Fig. 3

Statistical source data for Fig. 3b,c.

Source Data Fig. 4

Statistical source data for Fig. 4h,i.

Source Data Extended Data Fig. 1

Statistical source data for Extended Data Fig. 1c,e,f.

Source Data Extended Data Fig. 5

Statistical source data for Extended Data Fig. 5f,h,j.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Zhang, C., Wang, D., Wang, J. et al. Escape of hair follicle stem cells causes stem cell exhaustion during aging. Nat Aging 1, 889–903 (2021). https://doi.org/10.1038/s43587-021-00103-w

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s43587-021-00103-w

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