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:

Niche stiffening compromises hair follicle stem cell potential during ageing by reducing bivalent promoter accessibility

Abstract

Tissue turnover requires activation and lineage commitment of tissue-resident stem cells (SCs). These processes are impacted by ageing, but the mechanisms remain unclear. Here, we addressed the mechanisms of ageing in murine hair follicle SCs (HFSCs) and observed a widespread reduction in chromatin accessibility in aged HFSCs, particularly at key self-renewal and differentiation genes, characterized by bivalent promoters occupied by active and repressive chromatin marks. Consistent with this, aged HFSCs showed reduced ability to activate bivalent genes for efficient self-renewal and differentiation. These defects were niche dependent as the transplantation of aged HFSCs into young recipients or synthetic niches restored SC functions. Mechanistically, the aged HFSC niche displayed widespread alterations in extracellular matrix composition and mechanics, resulting in mechanical stress and concomitant transcriptional repression to silence promoters. As a consequence, increasing basement membrane stiffness recapitulated age-related SC changes. These data identify niche mechanics as a central regulator of chromatin state, which, when altered, leads to age-dependent SC exhaustion.

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: Age-induced HFSC exhaustion is associated with decreased chromatin accessibility.
Fig. 2: HFSC ageing results in the silencing of bivalent promoters.
Fig. 3: Ageing leads to niche-dependent compromised activation of bivalent genes and loss of SC potential.
Fig. 4: Widespread biochemical alterations in the ECM induce niche stiffening to control SC potential.
Fig. 5: Increased BM stiffness compromises HFSC maintenance in vivo.
Fig. 6: Mechanical stress suppresses transcription to silence bivalent promoters.

Similar content being viewed by others

Data availability

Sequencing data that support the findings of this study have been deposited at the Gene Expression Omnibus (GEO) under the accession code GSE148619. Proteomics data have been deposited to the ProteomeXchange Consortium through the PRIDE partner repository51 under the dataset identifier PXD018352. Previously published sequencing data14 that were reanalysed here are available under the accession code GSE31239. Source data are provided with this paper. All other data supporting the findings of this study are available from the corresponding author on reasonable request.

References

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Signer, R. A. & Morrison, S. J. Mechanisms that regulate stem cell aging and life span. Cell Stem Cell 12, 152–165 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Voigt, P., Tee, W. W. & Reinberg, D. A double take on bivalent promoters. Genes Dev. 27, 1318–1338 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Harikumar, A. & Meshorer, E. Chromatin remodeling and bivalent histone modifications in embryonic stem cells. EMBO Rep. 16, 1609–1619 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Hsu, Y. C., Li, L. & Fuchs, E. Emerging interactions between skin stem cells and their niches. Nat. Med. 20, 847–856 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Doles, J., Storer, M., Cozzuto, L., Roma, G. & Keyes, W. M. Age-associated inflammation inhibits epidermal stem cell function. Genes Dev. 26, 2144–2153 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Giangreco, A., Qin, M., Pintar, J. E. & Watt, F. M. Epidermal stem cells are retained in vivo throughout skin aging. Aging Cell 7, 250–259 (2008).

    Article  CAS  PubMed  Google Scholar 

  8. Keyes, B. E. et al. Nfatc1 orchestrates aging in hair follicle stem cells. Proc. Natl Acad. Sci. USA 110, E4950-9 (2013).

    Article  PubMed  CAS  Google Scholar 

  9. 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 

  10. 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 

  11. Saha, P. & Mishra, R. K. Heterochromatic hues of transcription—the diverse roles of noncoding transcripts from constitutive heterochromatin. FEBS J. 286, 4626–4641 (2019).

    Article  CAS  PubMed  Google Scholar 

  12. Brunet, A. & Rando, T. A. Interaction between epigenetic and metabolism in aging stem cells. Curr. Opin. Cell Biol. 45, 1–7 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. 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 

  14. 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 

  15. 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. Differ. Res. Biol. diversity 72, 548–557 (2004).

    Article  Google Scholar 

  16. 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 

  17. Chacon-Martinez, C. A., Klose, M., Niemann, C., Glauche, I. & Wickstrom, S. A. Hair follicle stem cell cultures reveal self-organizing plasticity of stem cells and their progeny. EMBO J. 36, 151–164 (2017).

    Article  CAS  PubMed  Google Scholar 

  18. 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 

  19. Ansorge, H. L. et al. Type XIV collagen regulates fibrillogenesis: premature collagen fibril growth and tissue dysfunction in null mice. J. Biol. Chem. 284, 8427–8438 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Bradshaw, A. D. et al. SPARC-null mice display abnormalities in the dermis characterized by decreased collagen fibril diameter and reduced tensile strength. J. Invest. Dermatol. 120, 949–955 (2003).

    Article  CAS  PubMed  Google Scholar 

  21. Guimaraes, C. F., Gasperini, L., Marques, A. P. & Reis, R. L. The stiffness of living tissues and its implications for tissue engineering. Nat. Rev. Mater. 5, 351–370 (2020).

    Article  Google Scholar 

  22. Langton, A. K., Graham, H. K., Griffiths, C. E. M. & Watson, R. E. B. Ageing significantly impacts the biomechanical function and structural composition of skin. Exp. Dermatol. 28, 981–984 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Mao, J. R. et al. Tenascin-X deficiency mimics Ehlers-Danlos syndrome in mice through alteration of collagen deposition. Nat. Genet. 30, 421–425 (2002).

    Article  CAS  PubMed  Google Scholar 

  24. Murrell, M., Oakes, P. W., Lenz, M. & Gardel, M. L. Forcing cells into shape: the mechanics of actomyosin contractility. Nat. Rev. Mol. Cell Biol. 16, 486–498 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Le, H. Q. et al. Mechanical regulation of transcription controls Polycomb-mediated gene silencing during lineage commitment. Nat. Cell Biol. 18, 864–875 (2016).

    Article  CAS  PubMed  Google Scholar 

  26. Vinogradova, M. et al. An inhibitor of KDM5 demethylases reduces survival of drug-tolerant cancer cells. Nat. Chem. Biol. 12, 531–538 (2016).

    Article  CAS  PubMed  Google Scholar 

  27. Conboy, I. M. et al. Rejuvenation of aged progenitor cells by exposure to a young systemic environment. Nature 433, 760–764 (2005).

    Article  CAS  PubMed  Google Scholar 

  28. Nishiguchi, M. A., Spencer, C. A., Leung, D. H. & Leung, T. H. Aging suppresses skin-derived circulating SDF1 to promote full-thickness tissue regeneration. Cell Rep. 24, 3383–3392 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Sarbacher, C. A. & Halper, J. T. Connective tissue and age-related diseases. Subcell. Biochem. 91, 281–310 (2019).

    Article  CAS  PubMed  Google Scholar 

  30. Segel, M. et al. Niche stiffness underlies the ageing of central nervous system progenitor cells. Nature 573, 130–134 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Marinkovich, M. P., Lunstrum, G. P. & Burgeson, R. E. The anchoring filament protein kalinin is synthesized and secreted as a high molecular weight precursor. J. Biol. Chem. 267, 17900–17906 (1992).

    Article  CAS  PubMed  Google Scholar 

  32. Sorokin, L. M. et al. Laminin α4 and integrin α6 are upregulated in regenerating dy/dy skeletal muscle: comparative expression of laminin and integrin isoforms in muscles regenerating after crush injury. Exp. Cell Res. 256, 500–514 (2000).

    Article  CAS  PubMed  Google Scholar 

  33. Schindelin, J. et al. Fiji: an open-source platform for biological-image analysis. Nat. Methods 9, 676–682 (2012).

    Article  CAS  PubMed  Google Scholar 

  34. Liang, Y. et al. A cell-instructive hydrogel to regulate malignancy of 3D tumor spheroids with matrix rigidity. Biomaterials 32, 9308–9315 (2011).

    Article  CAS  PubMed  Google Scholar 

  35. Dodt, M., Roehr, J. T., Ahmed, R. & Dieterich, C. FLEXBAR-flexible barcode and adapter processing for next-generation sequencing platforms. Biology 1, 895–905 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  36. Li, H. Aligning sequence reads, clone sequences and assembly contigs with BWA-MEM. Preprint at https://arxiv.org/abs/1303.3997 (2013).

  37. Allhoff, M., Sere, K., J, F. P., Zenke, M. & I, G. C. Differential peak calling of ChIP-seq signals with replicates with THOR. Nucleic Acids Res. 44, e153 (2016).

    PubMed  PubMed Central  Google Scholar 

  38. Ramirez, F. et al. deepTools2: a next generation web server for deep-sequencing data analysis. Nucleic Acids Res. 44, W160–W165 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Robinson, J. T. et al. Integrative genomics viewer. Nat. Biotechnol. 29, 24–26 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. 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 

  41. 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 

  42. Yu, G., Wang, L. G. & He, Q. Y. ChIPseeker: an R/Bioconductor package for ChIP peak annotation, comparison and visualization. Bioinformatics 31, 2382–2383 (2015).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  44. Liao, Y., Smyth, G. K. & Shi, W. featureCounts: an efficient general purpose program for assigning sequence reads to genomic features. Bioinformatics 30, 923–930 (2014).

    Article  CAS  PubMed  Google Scholar 

  45. 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 

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

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  47. Ross-Innes, C. S. et al. Differential oestrogen receptor binding is associated with clinical outcome in breast cancer. Nature 481, 389–393 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Rappsilber, J., Ishihama, Y. & Mann, M. Stop and go extraction tips for matrix-assisted laser desorption/ionization, nanoelectrospray, and LC/MS sample pretreatment in proteomics. Anal. Chem. 75, 663–670 (2003).

    Article  CAS  PubMed  Google Scholar 

  49. Cox, J. & Mann, M. MaxQuant enables high peptide identification rates, individualized p.p.b.-range mass accuracies and proteome-wide protein quantification. Nat. Biotechnol. 26, 1367–1372 (2008).

    Article  CAS  PubMed  Google Scholar 

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

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  51. Perez-Riverol, Y. et al. The PRIDE database and related tools and resources in 2019: improving support for quantification data. Nucleic Acids Res. 47, D442–D450 (2019).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank A. M. Luoto and N. Hachenberg for expert technical assistance; A. Gyenis for help with experiments; M. Loparic and D. Schneider for initial atomic force microscopy measurements; L. Sorokin for laminin antibodies; the staff at the FACS & Imaging Facility of MPI for Biology of Ageing, the Imaging Facility of CECAD Cologne and the Biomedicum Helsinki Imaging Unit for imaging support; and C. Becker and E. Kirstat at the Cologne Center for Genomics for help with sequencing. This work was supported by the Sigrid Juselius Foundation, Helsinki Institute of Life Science, Wihuri Research Institute, Max Planck Society, the Max Planck Förderstiftung and the European Research Council (ERC) under the EU Horizon 2020 research and innovation programme (grant agreement 770877—STEMpop) (all to S.A.W.) and the Deutsche Forschungsgemeinschaft (DFG; project number 73111208—SFB 829 (to S.A.W., C.M.N. and A.R.-I.), and FOR2722 to M.K. Y.A.M. is the recipient of the EMBO Long-Term fellowship ALTF 728-2017 and Human Frontier Science Program fellowship LT000861/2018.

Author information

Authors and Affiliations

Authors

Contributions

J.K. designed and performed most of the experiments and analysed data. Y.A.M. performed atomic force microscopy, immunostainings, organoid experiments, designed experiments and analysed data. S.G. performed the HFSC quantification and BrdU feeding experiments. C.A.C.-M. performed cell sorting and depilation experiments with J.K. J.M. performed immunostainings. X.L. and I.A. performed proteomics experiments. W.B. performed electron microscopy. J.A. performed ChIP sequencing. D.E.B. and M.K. provided collagen XIV and Tnx mutant mice and other key reagents. M.B. and A.R.-I. supported the ATAC-seq experiments and analysis of sequencing data. C.M.N. and A.R.-I. designed experiments and analysed data. S.A.W. conceived and supervised the study, designed experiments, analysed data and wrote the paper. All of the authors commented and edited the manuscript.

Corresponding author

Correspondence to Sara A. Wickström.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Peer review information Nature Cell Biology thanks Karl Lenhard Rudolph and the other, anonymous, reviewer(s) for their contribution to the peer review of this work. Peer reviewer reports are available.

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

Extended data

Extended Data Fig. 1 Analyses of young and aged HFSCs using flow cytometry, ATACseq and RNAseq.

a, Representative images of young (6 month) and aged (24 month) old mice show no visible differences in haircoats. (b,c) Immunofluorescence imaging (b) and quantification (c) of hair follicle density from young and old mice (mean ±SD; n = 5 mice/group). Scale bars 100 µm. d, Quantification of CD34 + /α6 integrin+ HFSCs by flow cytometry from young and aged mice cohorts from two different animal facilities (facilities #2 and #3, facility #1 shown in main Fig. 1b), where the respective young and aged mice were housed in the same room. Note decreased HFSC levels in mice from both facilities (mean, n = 7 mice/group; *p = 0.0293 Student’s t-test (facility #2), n = 4 mice/group; *p = 0.0283, Mann-Whitney (facility #3)). e, Plotting relative abundance of CD34 + HFSCs as a function of anagen-to-telogen ratio shows no correlation between the two parameters in young or aged mice (n = 15 young/14 aged mice; R2 = 0.0001168, Pearson’s correlation). f, Fragment frequency distribution of the ATACseq reads shows expected distribution both in young and aged biological replicates (n = 4 mice/group). g, Motif enrichment analyses of differentially accessible ATACseq peaks shows enrichment of CpG islands and promoter regions in regions more accessible in young HFSCs, whereas more accessible peaks in aged HFSCs are enriched in transposable elements. (-log(p) values: CpG islands – yHFSCs: 670.48/ aHFSCs: 2.03; promoters - yHFSCs: 289.13, aHFSCs: 1.22; 5’ utr - yHFSCs: 203.36, aHFSCs: 2.69; IAPEz-int|LTR | ERVK - young HFSCs: -40.18, aged HFSCs: 83.66; LTR | ERVK - young HFSCs: -151.31, aged HFSCs: 12.56) (h) Volcano plot of significantly altered transcripts in young and aged HFSCs. Genes involved in differentiation (eg. Mef2c, Gata6, Sox21, Tcf24), proliferation and signaling (Ptch1, Fgfr2, Tgfa, Nptx1), actin cytoskeleton (Espn, Fhod3, Smoc2), and stemness (Peg3) are among the most regulated (n = 3 mice/group; padj<0.05, Wald test/Benjamini-Hochberg). (i) GO-term analyses of genes up- and down-regulated in aged HFSCs.

Source data

Extended Data Fig. 2 Histone methylation analyses of young and aged HFSCs.

a, Heatmap of differentially accessible intergenic regions clustered according to H3K4me1, H3K27ac, H3K27me3 as well as the transcripts of these regions quantified by RNAseq from young and aged HFSCs. Note that most regions have high H3K4me1, but low H3K27ac and no detectible transcripts, indicative of a primed enhancer state. b, GO term analyses of the regions in (a) implicate enhancers for genes involved processes such as cell-cell adhesion, tissue development, cell differentiation, and proliferation to be less accessible (upper panel), whereas enhancers for genes involved in actin cytoskeleton organization and regulation of cell adhesion are more accessible (lower panel) in aged HFSCs. c, Heatmap of H3K27me3 ChIP sequencing peaks from young (H3K27me3-Y) and aged (H3K27me3-A) HFSCs shows slightly increased peak intensity in aged HFSCs (n = 2 mice/group). d, Box plot of -log2 fold changes of H3K4me3 occupancy at promoters of aged HFSC compared to young (top, middle and bottom delimiters are the 75th, 50th, 25th percentiles; top and bottom whiskers are the 90th and 10th percentiles; n = 2 mice/group). e, Representative genes from cluster-4 with functions in stem cell (SC) fate, differentiation and self-renewal include key genes required for HFSC activation and differentiation.

Source data

Extended Data Fig. 3 Niche-dependent regulation of HFSC potency and epigenetic state.

a, Representative images and quantification of BrdU incorporation in bulge (Bu) HFSCs from mice fed with BrdU in drinking water for 4 weeks. Note reduced BrdU incorporation in aged HFSCs (n = 3 mice/group). Scale bars 25 µm. b, Quantitative RT-PCR of RNAi knockdowns for selected genes with bivalent promoter state (n = 3 independent experiments). (c-e) Representative images (c) and quantification of colony number (d) and size (e) from colony forming assays with HFSCs depleted of the indicated genes (n = 3 independent experiments; *p = 0.0263, RM-ANOVA/Fischer’s, **p = 0.0045, Friedman/Dunn’s). f, Representative images and quantification of H3K4me3 in hair follicles generated in young nude mice by transplanting young or aged HFSCs (n = 3 mice/group). Scale bars 25 µm. g, Quantitative RT-PCR of selected genes with bivalent promoter state and reduced accessibility in vivo from H3K4me3 ChIP of young and aged HFSCs in 24 h 3 C organoid cultures. No significant differences in expression are observed (n = 4 mice/group). h, Quantitative RT-PCR of selected genes with bivalent promoter state and reduced accessibility in vivo from young and aged HFSCs in 3 C organoid cultures. No significant differences in expression are observed (n = 4 mice/group). Bar graphs in g, h show mean ±SEM, others mean ±SD.

Source data

Extended Data Fig. 4 Basement membranes in young and aged skin and decellularized scaffolds.

a, Representative immunofluorescence images and quantification of Laminin 332 staining show increased levels in aged BMs (mean; n = 4 mice/group, *p = 0.0286, Mann-Whitney; Scale bars 50 µm). b, Representative immunofluorescence images and quantification of Laminin 511 staining show increased levels in aged BMs (mean; n = 4 mice/group, *p = 0.0286, Mann-Whitney; Scale bars 50 µm). c, Schematic illustration of decellularized basement membrane scaffold preparation from young and aged mice. d, Immunofluorescence images of decellularized basement membrane scaffolds showing absence of epidermal cell nuclei but intact basement membranes as marked by Collagen IV and Laminin 332 staining. A representative of 3 mice is shown. Scale bars 100 µm. e, Box and whiskers plot of atomic force microscopy force indentation measurements of 3 C organoid hydrogel stiffness (top, middle and bottom delimiters are the 75th, 50th, 25th percentiles; top and bottom whiskers are the maximum and minimum; n = 13 force curves pooled across 4 independent experiments; **p = 0.0012, Kolmogorov-Smirnov).

Source data

Extended Data Fig. 5 Analyses of Collagen-XIV- and Tenascin-X-deficient mice.

a, Hematoxylin/Eosin stainings from 1 year old Collagen-XIV-deficient (Col XIV -/-) and wild type (ColXIV + /+) control mice reveal no overt skin pathology. Scale bars 100 µm. Representative images from 3 mice/genotype are shown. b, Immunofluorescence images of Cd34 staining to mark the bulge (bu) niche from 1 year old Col XIV + / + and -/- mice show comparable hair follicle morphology. Representative images from 3 mice/genotype are shown. Scale bars 25 µm. c, Quantification of CD34 + /α6 integrin+ HFSCs by flow cytometry from 6 months old Col XIV + / + and -/- mice shows no major reduction in levels of HFSCs (n = 5 Col XIV + / + and n = 4 Col XIV -/- mice). d, Quantification of basement membrane (BM) thickness from skin electron micrographs of Tenascin-X-deficient (TnX -/-) and wild type (TnX + /+) control mice. Note increased BM thickness in TnX -/- mice (n = 4 mice/group; *p = 0.0286, Mann-Whitney). e, mHematoxylin/Eosin stainings from 1 year old TnX + /+ and TnX -/- mice show no overt skin pathology. Representative images from 3 mice/genotype are shown. Scale bars 100 µm. f, Immunofluorescence images of Keratin-15 staining to mark the bulge (bu) niche from 1 year old TnX + /+ and -/- mice show comparable hair follicle morphology. Representative images from 3 mice/genotype are shown. Scale bars 25 µm. g, Quantification of CD34 + /α6 integrin+ HFSCs by flow cytometry from 6 months old TnX + /+ and -/- mice shows slightly reduced levels of HFSCs in TnX -/- mice (n = 7 TnX + /+ and n = 8 TnX -/- mice; *p = 0.0301, Student’s t-test). Bar graphs show mean ±SD.

Source data

Extended Data Fig. 6 Analyses of contractility and transcription in hair follicles and organoid cultures.

a, Representative immunofluorescence images and quantification of phosphorylated myosin light chain 2 (pMLC2). Note increased pMLC2 indicating increased contractility in aged bulge (bu) HFSCs (n = 5 young and n = 4 aged mice; *p = 0.0451, Student’s t-test; Scale bar 50 µm). b, Quantification of the nuclear aspect ratio (major axis/minor axis) shows increased nuclear elongation of aged HFSCs (n = 5 mice/group with >100 nuclei per mouse; **p = 0.0031, Student’s t-test). c, Representative images and quantification of H3K4me3 intensity from HFSC organoids treated with Kdm5 inhibitor (Kdm5i) (n = 6 independent experiments; p = 0.0515, Student’s t-test; Scale bar 50 µm). d, Quantitative RT-PCR of selected genes with bivalent promoter state from HFSC organoids treated with Kdm5i (n = 4 independent experiments). Bar graphs show mean ±SD.

Source data

Supplementary information

Supplementary Information

Supplementary Fig. 1: the gating strategy for quantification and purification of integrin α6+CD34+ HFSCs by FACS.

Reporting Summary

Peer Review Information

Supplementary Tables 1–7

Supplementary Table 1: differential peak analysis of ATAC-seq data. Analysis of differentially accessible genomic regions in young and aged HFSCs using ATAC-seq. Supplementary Table 2: differential gene expression analysis of RNA-seq data. Analysis of differentially expressed genes in young and aged HFSCs using RNA-seq. Supplementary Table 3: differential peak analysis of H3K4me3 ChIP–seq data. Analysis of differential H3K4me3 peaks in young and aged HFSCs. Supplementary Table 4: differential peak analysis of H3K27me3 ChIP–seq data. Analysis of differential H3K27me3 peaks in young and aged HFSCs. Supplementary Table 5: differential protein abundance analysis of proteomics data. Analyses of differences in proteome in young and aged skin. Supplementary Table 6: differential peak analysis of RNAPII ChIP–seq data. Analysis of differential RNAPII peaks in young and aged HFSCs. Supplementary Table 7: primers used in this study. A list of primers used for ChIP PCR and PCR in this study.

Source data

Source Data Fig. 1

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.

Source Data Extended Data Fig. 3

Statistical source data.

Source Data Extended Data Fig. 4

Statistical source data.

Source Data Extended Data Fig. 5

Statistical source data.

Source Data Extended Data Fig. 6

Statistical source data.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Koester, J., Miroshnikova, Y.A., Ghatak, S. et al. Niche stiffening compromises hair follicle stem cell potential during ageing by reducing bivalent promoter accessibility. Nat Cell Biol 23, 771–781 (2021). https://doi.org/10.1038/s41556-021-00705-x

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41556-021-00705-x

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