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:

Glucocorticoid signaling and regulatory T cells cooperate to maintain the hair-follicle stem-cell niche

Abstract

Maintenance of tissue homeostasis is dependent on the communication between stem cells and supporting cells in the same niche. Regulatory T cells (Treg cells) are emerging as a critical component of the stem-cell niche for supporting their differentiation. How Treg cells sense dynamic signals in this microenvironment and communicate with stem cells is mostly unknown. In the present study, by using hair follicles (HFs) to study Treg cell–stem cell crosstalk, we show an unrecognized function of the steroid hormone glucocorticoid in instructing skin-resident Treg cells to facilitate HF stem-cell (HFSC) activation and HF regeneration. Ablation of the glucocorticoid receptor (GR) in Treg cells blocks hair regeneration without affecting immune homeostasis. Mechanistically, GR and Foxp3 cooperate in Treg cells to induce transforming growth factor β3 (TGF-β3), which activates Smad2/3 in HFSCs and facilitates HFSC proliferation. The present study identifies crosstalk between Treg cells and HFSCs mediated by the GR–TGF-β3 axis, highlighting a possible means of manipulating Treg cells to support tissue regeneration.

This is a preview of subscription content, access via your institution

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: Impaired hair regeneration in GR cKO mice.
Fig. 2: GR signaling is required for Treg cell-facilitated HFSC activation and proliferation.
Fig. 3: GR signaling induces TGF-β3 expression by Treg cells.
Fig. 4: GR and Foxp3 cooperate to regulate TGF-β3 expression in Treg cells.
Fig. 5: Treg cell GR deficiency drives defective activation of TGF-β3:pSmad2/3 signaling in HFSCs.
Fig. 6: TGF-β3 produced by Treg cells promotes HFSC proliferation and hair regeneration.

Similar content being viewed by others

Data availability

Sequence data (RNA-seq and ChIP–seq) have been deposited in the Gene Expression Omnibus under the accession no. GSE183808. Source data are provided with this paper.

References

  1. Josefowicz, S. Z., Lu, L. F. & Rudensky, A. Y. Regulatory T cells: mechanisms of differentiation and function. Annu. Rev. Immunol. 30, 531–564 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Sakaguchi, S. et al. Regulatory T cells and human disease. Annu. Rev. Immunol. 38, 541–566 (2020).

    Article  CAS  PubMed  Google Scholar 

  3. Munoz-Rojas, A. R. & Mathis, D. Tissue regulatory T cells: regulatory chameleons. Nat. Rev. Immunol. https://doi.org/10.1038/s41577-021-00519-w (2021).

  4. Feuerer, M. et al. Lean, but not obese, fat is enriched for a unique population of regulatory T cells that affect metabolic parameters. Nat. Med. 15, 930–939 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Cipolletta, D. et al. PPAR-gamma is a major driver of the accumulation and phenotype of adipose tissue Treg cells. Nature 486, 549–553 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Bapat, S. P. et al. Depletion of fat-resident Treg cells prevents age-associated insulin resistance. Nature 528, 137–141 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Li, Y. et al. Insulin signaling establishes a developmental trajectory of adipose regulatory T cells. Nat. Immunol. 22, 1175–1185 (2021).

    Article  CAS  PubMed  Google Scholar 

  8. Ali, N. et al. Regulatory T cells in skin facilitate epithelial stem cell differentiation. Cell 169, 1119–1129.e1111 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Mathur, A. N. et al. Treg-cell control of a CXCL5–IL-17 inflammatory axis promotes hair-follicle-stem-cell differentiation during skin-barrier repair. Immunity 50, 655–667 e654 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Burzyn, D. et al. A special population of regulatory T cells potentiates muscle repair. Cell 155, 1282–1295 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Saxena, A. et al. Regulatory T cells are recruited in the infarcted mouse myocardium and may modulate fibroblast phenotype and function. Am. J. Physiol. Heart Circ. Physiol. 307, H1233–H1242 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Arpaia, N. et al. A distinct function of regulatory T cells in tissue protection. Cell 162, 1078–1089 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Hirata, Y. et al. CD150high bone marrow Tregs maintain gematopoietic stem cell quiescence and immune privilege via adenosine. Cell Stem Cell 22, 445–453.e445 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Ito, M. et al. Brain regulatory T cells suppress astrogliosis and potentiate neurological recovery. Nature 565, 246–250 (2019).

    Article  CAS  PubMed  Google Scholar 

  15. Naik, S., Larsen, S. B., Cowley, C. J. & Fuchs, E. Two to tango: dialog between immunity and stem cells in health and disease. Cell 175, 908–920 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Biton, M. et al. T helper cell cytokines modulate intestinal stem cell renewal and differentiation. Cell 175, 1307–1320.e1322 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Li, J. et al. Regulatory T-cells regulate neonatal heart regeneration by potentiating cardiomyocyte proliferation in a paracrine manner. Theranostics 9, 4324–4341 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Zacchigna, S. et al. Paracrine effect of regulatory T cells promotes cardiomyocyte proliferation during pregnancy and after myocardial infarction. Nat. Commun. 9, 2432 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  19. Cain, D. W. & Cidlowski, J. A. Immune regulation by glucocorticoids. Nat. Rev. Immunol. 17, 233–247 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Kadmiel, M. & Cidlowski, J. A. Glucocorticoid receptor signaling in health and disease. Trends Pharmacol. Sci. 34, 518–530 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Weikum, E. R., Knuesel, M. T., Ortlund, E. A. & Yamamoto, K. R. Glucocorticoid receptor control of transcription: precision and plasticity via allostery. Nat. Rev. Mol. Cell Biol. 18, 159–174 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Kim, D. et al. Anti-inflammatory roles of glucocorticoids are mediated by Foxp3+ regulatory T cells via a miR-342-dependent mechanism. Immunity https://doi.org/10.1016/j.immuni.2020.07.002 (2020).

  23. Taves, M. D., Gomez-Sanchez, C. E. & Soma, K. K. Extra-adrenal glucocorticoids and mineralocorticoids: evidence for local synthesis, regulation, and function. Am. J. Physiol. Endocrinol. Metab. 301, E11–E24 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. 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).

    Article  CAS  PubMed  Google Scholar 

  25. Vukelic, S. et al. Cortisol synthesis in epidermis is induced by IL-1 and tissue injury. J. Biol. Chem. 286, 10265–10275 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Rubtsov, Y. P. et al. Regulatory T cell-derived interleukin-10 limits inflammation at environmental interfaces. Immunity 28, 546–558 (2008).

    Article  CAS  PubMed  Google Scholar 

  27. Tronche, F. et al. Disruption of the glucocorticoid receptor gene in the nervous system results in reduced anxiety. Nat. Genet. 23, 99–103 (1999).

    Article  CAS  PubMed  Google Scholar 

  28. Franckaert, D. et al. Promiscuous Foxp3-cre activity reveals a differential requirement for CD28 in Foxp3+ and Foxp3 T cells. Immunol. Cell Biol. 93, 417–423 (2015).

    Article  CAS  PubMed  Google Scholar 

  29. Bittner-Eddy, P. D., Fischer, L. A. & Costalonga, M. Cre-loxP reporter mouse reveals stochastic activity of the Foxp3 promoter. Front. Immunol. 10, 2228 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Wu, D., Huang, Q., Orban, P. C. & Levings, M. K. Ectopic germline recombination activity of the widely used Foxp3-YFP-Cre mouse: a case report. Immunology 159, 231–241 (2020).

    Article  CAS  PubMed  Google Scholar 

  31. Rocamora-Reverte, L. et al. Glucocorticoid receptor-deficient Foxp3+ regulatory T cells fail to control experimental inflammatory bowel disease. Front. Immunol. 10, 472 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Stenn, K. S. & Paus, R. Controls of hair follicle cycling. Physiol. Rev. 81, 449–494 (2001).

    Article  CAS  PubMed  Google Scholar 

  33. Nagao, K. et al. Stress-induced production of chemokines by hair follicles regulates the trafficking of dendritic cells in skin. Nat. Immunol. 13, 744–752 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. 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 

  35. 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).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. 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).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Lay, K. et al. Stem cells repurpose proliferation to contain a breach in their niche barrier. eLife https://doi.org/10.7554/eLife.41661 (2018).

  38. Novak, J. S. S., Baksh, S. C. & Fuchs, E. Dietary interventions as regulators of stem cell behavior in homeostasis and disease. Genes Dev. 35, 199–211 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Flores, A. et al. Lactate dehydrogenase activity drives hair follicle stem cell activation. Nat. Cell Biol. 19, 1017–1026 (2017).

    Article  CAS  PubMed  Google Scholar 

  40. Ahmed, A., Almohanna, H., Griggs, J. & Tosti, A. Genetic hair disorders: a review. Dermatol. Ther. 9, 421–448 (2019).

    Article  Google Scholar 

  41. Malhotra, N. et al. RORalpha-expressing T regulatory cells restrain allergic skin inflammation. Sci. Immunol. https://doi.org/10.1126/sciimmunol.aao6923 (2018).

  42. Kalekar, L. A. et al. Regulatory T cells in skin are uniquely poised to suppress profibrotic immune responses. Sci. Immunol. https://doi.org/10.1126/sciimmunol.aaw2910 (2019).

  43. Nosbaum, A. et al. Cutting edge: regulatory T cells facilitate cutaneous wound healing. J. Immunol. 196, 2010–2014 (2016).

    Article  CAS  PubMed  Google Scholar 

  44. Shimba, A. et al. Glucocorticoids drive diurnal oscillations in T cell distribution and responses by inducing interleukin-7 receptor and CXCR4. Immunity 48, 286–298 e286 (2018).

    Article  CAS  PubMed  Google Scholar 

  45. Hou, R., Denisenko, E., Ong, H. T., Ramilowski, J. A. & Forrest, A. R. R. Predicting cell-to-cell communication networks using NATMI. Nat. Commun. 11, 5011 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Oshimori, N. & Fuchs, E. Paracrine TGF-beta signaling counterbalances BMP-mediated repression in hair follicle stem cell activation. Cell Stem Cell 10, 63–75 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Rahmani, W. et al. Macrophages promote wound-induced hair follicle regeneration in a CX3CR1- and TGF-beta1-dependent manner. J. Invest. Dermatol. 138, 2111–2122 (2018).

    Article  CAS  PubMed  Google Scholar 

  48. Foitzik, K., Paus, R., Doetschman, T. & Dotto, G. P. The TGF-beta2 isoform is both a required and sufficient inducer of murine hair follicle morphogenesis. Dev. Biol. 212, 278–289 (1999).

    Article  CAS  PubMed  Google Scholar 

  49. Gilbert, L. A. et al. CRISPR-mediated modular RNA-guided regulation of transcription in eukaryotes. Cell 154, 442–451 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Cameron, J., Martino, P., Nguyen, L. & Li, X. Cutting edge: CRISPR-based transcriptional regulators reveal transcription-dependent establishment of epigenetic memory of Foxp3 in regulatory T cells. J. Immunol. 205, 2953–2958 (2020).

    Article  CAS  PubMed  Google Scholar 

  51. Loo, C. S. et al. A genome-wide CRISPR screen reveals a role for the non-canonical nucleosome-remodeling BAF complex in Foxp3 expression and regulatory T cell function. Immunity 53, 143–157 e148 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. 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 

  53. Mullen, A. C. & Wrana, J. L. TGF-beta family signaling in embryonic and somatic stem-cell renewal and differentiation. Cold Spring Harb. Perspect. Biol. https://doi.org/10.1101/cshperspect.a022186 (2017).

  54. Clavel, C. et al. Sox2 in the dermal papilla niche controls hair growth by fine-tuning BMP signaling in differentiating hair shaft progenitors. Dev. Cell 23, 981–994 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Doetschman, T. et al. Generation of mice with a conditional allele for the transforming growth factor beta3 gene. Genesis 50, 59–66 (2012).

    Article  CAS  PubMed  Google Scholar 

  56. Weirather, J. et al. Foxp3+ CD4+ T cells improve healing after myocardial infarction by modulating monocyte/macrophage differentiation. Circ. Res. 115, 55–67 (2014).

    Article  CAS  PubMed  Google Scholar 

  57. Kim, J. M., Rasmussen, J. P. & Rudensky, A. Y. Regulatory T cells prevent catastrophic autoimmunity throughout the lifespan of mice. Nat. Immunol. 8, 191–197 (2007).

    Article  CAS  PubMed  Google Scholar 

  58. Slominski, R. M. et al. Extra-adrenal glucocorticoid biosynthesis: implications for autoimmune and inflammatory disorders. Genes Immun. 21, 150–168 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Choi, S. et al. Corticosterone inhibits GAS6 to govern hair follicle stem-cell quiescence. Nature 592, 428–432 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Hengge, U. R., Ruzicka, T., Schwartz, R. A. & Cork, M. J. Adverse effects of topical glucocorticosteroids. J. Am. Acad. Dermatol. 54, 1–15 (2006). quiz 16-18.

    Article  PubMed  Google Scholar 

  61. Rieckmann, M. et al. Myocardial infarction triggers cardioprotective antigen-specific T helper cell responses. J. Clin. Invest. 129, 4922–4936 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Liston, A. et al. Differentiation of regulatory Foxp3+ T cells in the thymic cortex. Proc. Natl. Acad. Sci. USA 105, 11903–11908 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Nowak, J. A. & Fuchs, E. Isolation and culture of epithelial stem cells. Methods Mol. Biol. 482, 215–232 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Andrews, S. FastQC: a quality control tool for high throughput sequence data. BioInformatics http://www.bioinformatics.babraham.ac.uk/projects/fastqc (2010).

  65. Dobin, A. et al. STAR: ultrafast universal RNA-seq aligner. Bioinformatics 29, 15–21 (2013).

    Article  CAS  PubMed  Google Scholar 

  66. 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 

  67. 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  Google Scholar 

  68. 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  Google Scholar 

  69. 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  PubMed  PubMed Central  Google Scholar 

  70. Robinson, M. D., McCarthy, D. J. & Smyth, G. K. edgeR: a Bioconductor package for differential expression analysis of digital gene expression data. Bioinformatics 26, 139–140 (2010).

    Article  CAS  PubMed  Google Scholar 

  71. Robinson, M. D. & Oshlack, A. A scaling normalization method for differential expression analysis of RNA-seq data. Genome Biol. 11, R25 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

  72. Kent, W. J. et al. The human genome browser at UCSC. Genome Res. 12, 996–1006 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank T. Hua and C. Gordon for mouse colony management, C. O’Connor for assistance in flow cytometry, U. Manor for assistance in confocal microscopy, N. Hah for assistance in RNA-seq and ChIP–seq experiments, X. Li (Tufts) for CRISPRi vectors, M. Kurita, J. C. I. Belmonte, N. He and R. M. Evans for helpful discussion, and S. P. Bapat (University of California, San Francisco), L. F. Lu (UCSD), C. Wu (National Cancer Institute (NCI)) and T. Mann for reviewing the manuscript. Z.L. was supported by a NOMIS fellowship. J.Y. and M.N.S were supported by the National Institutes of Health (grant nos.: NCI CCSG P30-014195, NIA P01-AG073084, NIA-NMG RF1-AG064049 and NIA P30-AG068635) and the Leona M. and Harry B. Helmsley Charitable Trust. Y.Z. was supported by the NOMIS Foundation, the Crohn’s and Colitis Foundation, the Leona M. and Harry B. Helmsley Charitable Trust and the National Institutes of Health (grant nos. R01-AI107027, R01-AI1511123, R21-AI154919 and S10-OD023689). This work was also supported by the NCI-funded Salk Institute Cancer Center Core Facilities (grant no. P30-CA014195).

Author information

Authors and Affiliations

Authors

Contributions

Z.L. and Y.Z. conceived the project. Z.L. provided the methodology. Z.L., X.H. and Y.L. carried out the investigations. H.L. and Y.Z. provided the resources. Z.L., J.Y. and M.N.S. carried out the formal analysis. Z.L. and J.Y. curated the data. Y.Z. supervised the project. Y.Z. acquired the funding. Z.L. and Y.Z. wrote the original draft of the paper. Z.L. and Y.Z. wrote, edited and reviewed the paper.

Corresponding author

Correspondence to Ye Zheng.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature Immunology thanks Ming Li and the other, anonymous, reviewer(s) for their contribution to the peer review of this work. N. Bernard was the primary editor on this article and managed its editorial process and peer review in collaboration with the rest of the editorial team.

Additional information

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

Extended data

Extended Data Fig. 1 Normal development and function of Treg cells in GR cKO mice.

a, b, Flow cytometric analysis and quantification of CD4+ T cells and CD8+ T cells (a), and CD25+Foxp3 Treg progenitors, CD25Foxp3+ Treg progenitors, CD25+Foxp3+ Treg cells (b) in the thymus of 2-month-old WT and GR cKO mice (n = 5 mice per condition). NS, P > 0.05. c, d, Flow cytometric analysis and quantification of CD4+ T cells and CD8+ T cells (c) and Foxp3+ Treg cells (d) in the spleen of 6-month-old WT (n = 7) and GR cKO (n = 8) mice. NS, P > 0.05. e, f, g, Flow cytometric analysis and quantification of CD44highCD62Llow activated/memory CD4+ T cells (e), IFNγ or IL-17 producing CD4+ T cells and CD8+ T cells (f), IL-5 or IL-13 producing CD4+ T cells (g) in the spleen of 6-month-old WT (n = 7) and GR cKO (n = 8) mice. NS, P > 0.05. h, Suppression of proliferation of wild-type naive CD4+ T responder cells (Tresp) by WT and GR cKO Treg cells in an in vitro suppression assay (n = 3 biologically independent replicates per condition). NS, P > 0.05. i, Measurement of WT and GR cKO Treg cells suppressive function in a T cell transfer-induced colitis model by body weight loss (n = 7 mice per condition). NS, P > 0.05. Statistical analysis was performed using a two-tailed unpaired Student’s t-test. Data are represented as mean ± SEM. Representative data of two (a-i) independent experiments are shown.

Source data

Extended Data Fig. 2 Specific deletion of GR in Treg cells.

a, b, Quantification of Foxp3+ Treg cells (a), IFNγ or IL-17 producing in CD4+ T cells and CD8+ T cells (b) from the skin of WT (n = 6) and GR cKO mice (n = 5). NS, P > 0.05. c, d, Flow cytometric analysis and quantification of the expression of GR protein in Treg cells, CD4+ Teff cells, CD8+ T cells, dermal γδT cells, DETC from the skin of WT and GR cKO mice (n = 8 mice per group). FMO: Fluorescence minus one control. ****P < 0.0001; NS, P > 0.05. Statistical analysis was performed using a two-tailed unpaired Student’s t-test. Data are represented as mean ± SEM. Representative data of two (a-d) independent experiments are shown.

Source data

Extended Data Fig. 3 Transcriptomic analysis of HFSCs isolated from WT and GR cKO mice.

a, Immunofluorescence staining of Ki67 in HFs from WT and GR cKO mice 3 days post-depilation (n = 3 mice per condition). Arrows indicate hair germ location. Scale bars, 50 μm. b, EdU was intraperitoneally injected on day 2 post-depilation. Immunofluorescence staining of EdU in HFs from WT and GR cKO mice 3 days post-depilation (n = 3 mice per condition). Scale bars, 50 μm. c, RT-qPCR comparison of genes related HFSC proliferation between HFSCs from WT and GR cKO mice 4 days post-depilation (n = 5 mice per condition). **P = 0.0035 (Cdk1); *P = 0.047 (Cdk4); ****P < 0.0001 (Cdca3); ***P = 0.0005 (E2f8); ****P < 0.0001 (Bub1b); ***P = 0.0008 (Ccnd1). d, Heatmap of genes related to HFSC differentiation between HFSCs isolated from WT and GR cKO mice 4 days after depilation. e, Gene ontology analysis of genes down-regulated in HFSCs from WT relative to GR cKO mice. f, GSEA plot for the “Hallmarks – Abnormal hair growth” signature and heatmap of related gene expression in HFSCs from WT and GR cKO mice. Statistical analysis was performed using a two-tailed unpaired Student’s t-test. Data are represented as mean ± SEM. Representative data of three (a-b) or two (c) independent experiments are shown.

Source data

Extended Data Fig. 4 Gating strategy for flow cytometric analysis of skin lymphoid and myeloid cells.

a, Flow cytometric gating strategy to identify skin lymphoid cell lineages, including DETC, dermal γδT cells, CD4+ Teff cells, Treg cells, CD8+ T cells, and the production of proinflammatory cytokines IL-17 and IFNγ by these cells. b, Flow cytometric gating strategy to identify skin myeloid cell lineages, including neutrophils, eosinophils, macrophages and dendritic cells.

Extended Data Fig. 5 Normal immune homeostasis in the skin of GR cKO mice.

WT and GR cKO mice (n = 8 mice per condition) were depilated to induce hair regeneration. 5 days post-depilation, skin immune cell populations were analyzed by FACS. a, b, Representative flow cytometric plots of Treg cells and quantification of Treg cell number, Foxp3+ Treg cells ratio and Foxp3 protein level in the skin of WT and GR cKO mice (n = 8 mice per condition). NS, P > 0.05. c, Quantification of proinflammatory cytokine IFNγ and IL-17 production in DETCs and dermal γδ T cells, CD4+ Teff cells, and CD8+ T cells (n = 8 mice per condition). NS, P > 0.05. d, Quantification of cell numbers of DETCs, dermal γδ T cells, CD4+ Teff cells, and CD8+ T cells, neutrophils, eosinophils, macrophages, and dendritic cells in the skin (n = 8 mice per condition). NS, P > 0.05. e, Representative H&E staining of skin on day 5 post-depilation. Scale bars, 100 μm. Statistical analysis was performed using a two-tailed unpaired Student’s t-test. Data are represented as mean ± SEM. Representative data of two (a-d) independent experiments are shown.

Source data

Extended Data Fig. 6 Transcriptomic analysis of WT and GR cKO skin-resident Treg cells.

a, b, Comparison of Treg cells signature genes (a) and genes associated with Treg cell function in the skin (b) between WT (n = 3) and GR cKO (n = 5) skin-resident Treg cells one day post-depilation. NS, P > 0.05. c, RT-qPCR analysis of the expression of Jag1 in skin Treg cells from WT (n = 3) and GR cKO (n = 4) mice one day post-depilation. **P = 0.0046. d, e, Flow cytometric analysis and quantification of the Jagged-1 protein in skin Treg cells from WT (n = 5) and GR cKO (n = 4) mice. NS, P > 0.05. f, Comparison of the Expression of Notch target genes in HFSCs between WT and GR cKO mice (n = 3 mice per condition). NS, P > 0.05. g, Summary of acquired data from NATMI, showing changes of all the differential pair weight of Tgfb3-Tgfbr and Jag1-Notch between WT and GR cKO mice. Statistical analysis was performed using a two-tailed unpaired Student’s t-test. Data are represented as mean ± SEM. Representative data of two (c-e) independent experiments are shown.

Source data

Extended Data Fig. 7 Induction of TGF-β3 expression by dexamethasone in Treg cells is dependent on the Tgfb3 enhancers.

a, RT-qPCR analysis of the expression of Tgfb1 and Il7r in WT (n = 3) and GR cKO (n = 4) skin-resident Treg cells one day post-depilation. NS, P > 0.05; **P = 0.0015. b, Schematic for CRISPR knockout of GR/Foxp3-bound peaks by CRISPR-Cas9. c, RT-qPCR analysis of the expression of Tgfb3 and Ttll5 after CRISPR knockout of indicated GR- and Foxp3- bound sites in Treg cells (n = 3 biologically independent replicates per condition). For Tgfb3 (up): ***P = 0.0003 (Tgfb3 pro); **P = 0.0018 (Tgfb3 intron); **P = 0.0094 (Ttll5 Peak1); **P = 0.0071 (Ttll5 Peak2); **P = 0.0094 (Ttll5 peak3). For Ttll5 (bottom): NS, P > 0.05; *P = 0.0272 (Tgfb3 intron); *P = 0.0205 (Ttll5 Peak3). Statistical analysis was performed using a two-tailed unpaired Student’s t-test. Data are represented as mean ± SEM. Representative data of two (a, c) independent experiments are shown.

Source data

Extended Data Fig. 8 HFSCs from GR cKO mice show an enrichment of BMP/pSmad1/5 signaling.

a, Immunofluorescence staining and quantification of pSmad1/5 and pSmad2/3 in HFs from WT and GR cKO mice before hair depilation (n = 10 HFs from 3 mice per condition). Scale bars, 50 μm. NS, P > 0.05. b, Immunofluorescence staining and quantification of pSmad1/5 and pSmad2/3 in HFs from WT and GR cKO mice 3 days post-depilation (n = 10 HFs from 3 mice per condition). Scale bars, 50 mm. ****P < 0.0001. c, GSEA plot for the BMP-responsive genes in HFSCs (Genander et al, 2014) from WT to GR cKO mice isolated 4 days post-depilation. d, RT-qPCR analysis of the expression of genes encoding ligands for Wnt and BMP pathways in the skin from WT and GR cKO mice one day post-depilation (n = 5 mice per condition). NS, P > 0.05. Statistical analysis was performed using a two-tailed unpaired Student’s t-test. Data are represented as mean ± SEM. Representative data of three (a, b) or two (d) independent experiments are shown.

Source data

Extended Data Fig. 9 Intradermal injection of TGF-β3 in GR cKO mice results in activation of pSmad2/3 signaling and hair follicle regeneration.

a, Surface view of the BSA or TGF-β3 injected area in WT mice on day 20 post-depilation. Circle in the dashed line indicated injection sites. b, Representative H&E staining and quantification of HF length at the BSA or TGF-β3 injected area of the skin sections from GR cKO mice (n = 25 HFs from 4 mice per condition). The whole images were reconstructed from two adjacent images using stitching plugins from Image J. The area between two dashed lines was the injection site. Scale bars, 500 μm. ****P < 0.0001 c, Immunofluorescence staining and quantification of pSmad1/5 and pSmad2/3 at the BSA or TGF-β3 injected area from GR cKO mice (n = 20 HFs from 5 mice per condition). Scale bars, 50 μm. ****P < 0.0001. Statistical analysis was performed using a two-tailed unpaired Student’s t-test. Data are represented as mean ± SEM. Representative data of three independent experiments are shown.

Source data

Extended Data Fig. 10 Normal T cell development and distribution in TGF-β3 cKO mice.

a, b, Flow cytometric analysis and quantification of CD4+ T cells and CD8+ T cells (a), and Foxp3+ Treg cells (b) in the thymus of 2-month-old WT and TGF-β3 cKO mice (n = 5 mice per condition). NS, P > 0.05. c, d, Flow cytometric analysis and quantification of CD4+ T cells and CD8+ T cells (c), Foxp3+ Treg cells (d) in the spleen of 2-month-old WT and TGF-β3 cKO mice (n = 5 mice per condition). NS, P > 0.05. e, Flow cytometric analysis and quantification CD44highCD62Llow activated/memory T cells in the spleen of WT and TGF-β3 cKO mice (n = 5 mice per condition). Data are mean ± SEM. NS, P > 0.05. Statistical analysis was performed using a two-tailed unpaired Student’s t-test. Data are represented as mean ± SEM. Representative data of two (a-e) independent experiments are shown.

Source data

Supplementary information

Reporting Summary

Supplementary Tables

Supplementary Tables 1–3.

Source data

Source Data Fig. 1

Statistical source data.

Source Data Fig. 2

Statistical source data.

Source Data Fig. 3

Statistical source data.

Source Data Fig. 4

Statistical source data.

Source Data Fig. 5

Statistical source data.

Source Data Fig. 6

Statistical source data.

Source Data Extended Data Fig. 1

Statistical source data.

Source Data Extended Data Fig. 2

Statistical source data.a

Source Data Extended Data Fig. 3

Statistical source data.

Source Data Extended Data Fig. 5

Statistical source data.

Source Data Extended Data Fig. 6

Statistical source data.

Source Data Extended Data Fig. 7

Statistical source data.

Source Data Extended Data Fig. 8

Statistical source data.

Source Data Extended Data Fig. 9

Statistical source data.

Source Data Extended Data Fig. 10

Statistical source data.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Liu, Z., Hu, X., Liang, Y. et al. Glucocorticoid signaling and regulatory T cells cooperate to maintain the hair-follicle stem-cell niche. Nat Immunol 23, 1086–1097 (2022). https://doi.org/10.1038/s41590-022-01244-9

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41590-022-01244-9

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