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.

  • Letter
  • Published:

Vasculature atrophy causes a stiffened microenvironment that augments epidermal stem cell differentiation in aged skin

Abstract

Stem cell loss causes tissue deterioration associated with aging. The accumulation of genomic and oxidative stress-induced DNA damage is an intrinsic cue for stem cell loss1,2; however, whether there is an external microenvironmental cue that triggers stem cell loss remains unclear. Here we report that the involution of skin vasculature causes dermal stiffening that augments the differentiation and hemidesmosome fragility of interfollicular epidermal stem cells (IFESCs) in aged mouse skin. Aging-related IFESC dysregulation occurs in plantar and tail skin, and is correlated with prolonged calcium influx, which is contributed by the mechanoresponsive ion channel Piezo1 (ref. 3). Epidermal deletion of Piezo1 ameliorated IFESC dysregulation in aged skin, whereas Piezo1 activation augmented IFESC differentiation and hemidesmosome fragility in young mice. The dermis stiffened with age, which was accompanied by dermal vasculature atrophy. Conversely, induction of the dermal vasculature softened the dermis and ameliorated IFESC dysregulation in aged skin. Single-cell RNA sequencing of dermal fibroblasts identified an aging-associated anti-angiogenetic secretory molecule, pentraxin 3 (ref. 4), which caused dermal sclerotization and IFESC dysregulation in aged skin. Our findings show that the vasculature softens the microenvironment for stem cell maintenance and provide a potential mechanobiology-based therapeutic strategy against skin disorders in aging.

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: IFESCs are prone to differentiation and delamination with prolonged calcium influx in aged plantar skin.
Fig. 2: Dermal stiffening-induced Piezo1 activation causes IFESC dysregulation.
Fig. 3: Vascular atrophy induces dermal stiffening and IFESC dysregulation.
Fig. 4: Ptx3 induces vascular atrophy that leads to dermal stiffening and IFESC dysregulation.

Similar content being viewed by others

Data availability

The RNA-seq and scRNA-seq data were deposited in the Gene Expression Omnibus (GSE171035) and DNA Data Bank of Japan (DRA009978), respectively. Source data are provided with this study. All data needed to evaluate the conclusions in the study are present in the article and/or Supplementary Materials. All data are also available from the corresponding author upon reasonable request.

References

  1. Behrens, A., van Deursen, J. M., Rudolph, K. L. & Schumacher, B. Impact of genomic damage and ageing on stem cell function. Nat. Cell Biol. 16, 201–207 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Vitale, I. et al. DNA damage in stem cells. Mol Cell. 66, 306–319 (2017).

    Article  CAS  PubMed  Google Scholar 

  3. Coste, B. et al. Piezo1 and Piezo2 are essential components of distinct mechanically activated cation channels. Science 330, 55–60 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Garlanda, C. et al. Non-redundant role of the long pentraxin PTX3 in anti-fungal innate immune response. Nature 420, 182–186 (2002).

    Article  CAS  PubMed  Google Scholar 

  5. Kurban, R. S. & Bhawan, J. Histologic changes in skin associated with aging. J. Dermatol. Surg. Oncol. 16, 908–914 (1990).

    Article  CAS  PubMed  Google Scholar 

  6. Ashcroft, G. S. et al. Estrogen accelerates cutaneous wound healing associated with an increase in TGF-β1 levels. Nat. Med. 11, 1209–1215 (1997).

    Article  Google Scholar 

  7. Gosain, A. & DiPietro, L. A. Aging and wound healing. World J. Surg. 28, 321–326 (2004).

    Article  PubMed  Google Scholar 

  8. Liu, N. et al. Stem cell competition orchestrates skin homeostasis and ageing. Nature 568, 344–350 (2019).

    Article  CAS  PubMed  Google Scholar 

  9. Watanabe, M. et al. Type XVII collagen coordinates proliferation in the interfollicular epidermis. eLife 6, e26635 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  10. Vázquez, F., Palacios, S., Alemañ, N. & Guerrero, F. Changes of the basement membrane and type IV collagen in human skin during aging. Maturitas 25, 209–215 (1996).

    Article  PubMed  Google Scholar 

  11. Bikle, D. D., Xie, Z. & Tu, C. L. Calcium regulation of keratinocyte differentiation. Expert Rev. Endocrinol. Metab. 7, 461–472 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Song, J. G. et al. Structural insights into Ca2+-calmodulin regulation of plectin 1a-integrin β4 interaction in hemidesmosomes. Structure 23, 558–570 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Wells, R. G. The role of matrix stiffness in regulating cell behavior. Hepatology 47, 1394–1400 (2008).

    Article  CAS  PubMed  Google Scholar 

  14. Wu, J., Lewis, A. H. & Grandl, J. Touch, tension, and transduction—the function and regulation of piezo ion channels. Trends Biochem. Sci. 42, 57–71 (2017).

    Article  PubMed  CAS  Google Scholar 

  15. Keyes, B. E. et al. Impaired epidermal to dendritic T cell signaling slows wound repair in aged skin. Cell 167, 1323–1338 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Quan, T. & Fisher, G. J. Role of age-associated alterations of the dermal extracellular matrix microenvironment in human skin aging: a mini-review. Gerontology. 61, 427–434 (2015).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  18. Ichijo, R. et al. Vasculature-driven stem cell population coordinates tissue scaling in dynamic organs. Sci. Adv. 7, eabd2575 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Gunin, A. G. et al. Age-related changes in angiogenesis in human dermis. Exp. Gerontol. 55, 143–151 (2014).

    Article  CAS  PubMed  Google Scholar 

  20. Detmar, M. et al. Increased microvascular density and enhanced leukocyte rolling and adhesion in the skin of VEGF transgenic mice. J. Invest. Dermatol. 111, 1–6 (1998).

    Article  CAS  PubMed  Google Scholar 

  21. Efremova, M., Vento-Tormo, M., Teichmann, S. A. & Vento-Tormo, R. CellPhoneDB: inferring cell–cell communication from combined expression of multi-subunit ligand–receptor complexes. Nat. Protoc. 15, 1484–1506 (2020).

    Article  CAS  PubMed  Google Scholar 

  22. Rusnati, M. et al. Selective recognition of fibroblast growth factor-2 by the long pentraxin PTX3 inhibits angiogenesis. Blood 104, 92–99 (2004).

    Article  CAS  PubMed  Google Scholar 

  23. Doni, A. et al. The long pentraxin PTX3 as a link between innate immunity, tissue remodeling, and cancer. Front. Immunol. 10, 712 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Valavanidis, A., Vlachogianni, T. & Fiotakis, C. 8-hydroxy-2'-deoxyguanosine (8-OHdG): a critical biomarker of oxidative stress and carcinogenesis. J. Environ. Sci. Health C 27, 120–139 (2009).

    Article  CAS  Google Scholar 

  25. Jingpeng, G. et al. Architecture of the mammalian mechanosensitive Piezo1 channel. Nature 527, 64–69 (2015).

    Article  CAS  Google Scholar 

  26. Syeda, R. et al. Piezo1 channels are inherently mechanosensitive. Cell Rep. 17, 1739–1746 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Saotome, K. et al. Structure of the mechanically activated ion channel Piezo1. Nature 554, 481–486 (2018).

    Article  CAS  PubMed  Google Scholar 

  28. Fornai, F. et al. The inflammatory protein pentraxin 3 in cardiovascular disease. Immun. Ageing 13, 25 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  29. Vasioukhin, V., Degenstein, L., Wise, B. & Fuchs, E. The magical touch: genome targeting in epidermal stem cells induced by tamoxifen application to mouse skin. Proc. Natl Acad. Sci. USA 96, 8551–8556 (1999).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Chhalan, S. M. et al. Piezo1 links mechanical forces to red blood cell volume. eLife 4, e07370 (2015).

    Article  Google Scholar 

  31. Hooper, A. T. et al. Engraftment and reconstitution of hematopoiesis is dependent on VEGFR2-mediated regeneration of sinusoidal endothelial cells. Cell Stem Cell 4, 263–274 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Ebina, T. et al. 3D clustering of GABAergic neurons enhances inhibitory actions on excitatory neurons in the mouse visual cortex. Cell Rep. 9, 1896–1907 (2014).

    Article  CAS  PubMed  Google Scholar 

  33. Okabe, K. et al. Neurons limit angiogenesis by titrating VEGF in retina. Cell 159, 584–596 (2014).

    Article  CAS  PubMed  Google Scholar 

  34. Butt, H. J. & Jaschke, M. Calculation of thermal noise in atomic force microscopy. Nanotechnology 6, 1 (1995).

    Article  Google Scholar 

  35. Rodríguez-Franco, P. et al. Long-lived force patterns and deformation waves at repulsive epithelial boundaries. Nat. Mater. 16, 1029–1037 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  36. Hino, N. et al. ERK-mediated mechanochemical waves direct collective cell polarization. Dev. Cell 53, 646–660 (2020).

    Article  CAS  PubMed  Google Scholar 

  37. Lichti, U., Anders, J. & Yuspa, S. H. Isolation and short-term culture of primary keratinocytes, hair follicle populations and dermal cells from newborn mice and keratinocytes from adult mice for in vitro analysis and for grafting to immunodeficient mice. Nat. Protoc. 3, 799–810 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  39. Kechin, A., Boyarskikh, U., Kel, A. & Filipenko, M. cutPrimers: a new tool for accurate cutting of primers from reads of targeted next generation sequencing. J. Comput. Biol. 24, 1138–1143 (2017).

    Article  CAS  PubMed  Google Scholar 

  40. 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 

  41. 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 

  42. Butler, A. et al. Integrating single-cell transcriptomic data across different conditions, technologies, and species. Nat. Biotechnol. 36, 411–420 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. 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 

Download references

Acknowledgements

We thank S. Kitano, H. Miyachi, Y. Iizuka, M. Yoshikawa, A. Sada and N. Hino for support and technical advice and A. Enomoto, T. Miyata and K. Kabashima for support on AFM and calcium imaging analyses. We thank staff at the Single-Cell Genome Information Analysis Core in ASHBi for RNA-seq analysis. We thank A. Mantovani and C. Garlanda at IRCCS Humanitas for the Ptx3 KO mice and Y. Kubota at Keio University for the VE-cadherin-creER mice. This work was supported by JSPS KAKENHI 16H06280 (R.I.), 19K17796 (R.I.), 21K16209 (R.I.), 17H05640 (F.T.), 20K21601 (F.T.), 16H06279 (PAGS) (F.T.), AMED under grant no. 20gm5810029 (F.T.), CREST under grant no. JPMJCR2023 (F.T., T.Y. and K.M.), the Takeda Science Foundation (F.T.), KAO Foundation for Arts and Sciences (R.I.), Fujiwara Memorial Foundation (R.I.), ASHBi, supported by the World Premier International Research Center Initiative (WPI) and MEXT Japan. We thank J. Ludovic Croxford and M. Crawford from Edanz (https://jp.edanz.com/ac) for editing a draft of this manuscript.

Author information

Authors and Affiliations

Authors

Contributions

R.I. and F.T. conceived the project and designed the experiments. R.I. performed most experiments. K.M., A.N. and T.A. analyzed stiffness using AFM. M.K. and T.Y performed sequencing experiments and bioinformatics analyses. T.M. and T.H. contributed to live imaging experiments using two-photon microscopy. S.I. and H.H. contributed to hydrogel-related experiments for keratinocytes. R.I. and F.T. wrote the manuscript.

Corresponding authors

Correspondence to Ryo Ichijo or Fumiko Toyoshima.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature Aging thanks Salvador Benitah, Richard Cubbon and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

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 Aging causes stem cell dysregulation.

a, Representative immunofluorescence images and fluorescence intensities of COL17 in plantar skin from young (6 months) and aged (24–27 months) mice (n = 5). Data point; 30 cell average/mouse. b, Representative immunofluorescence images and fluorescence intensities of plectin in plantar skin from young (6 months) and aged (24–27 months) mice (n = 5). Each data point is the average of 30 cells per mouse. c, Representative immunofluorescence images of survivin in IFESCs from control (6 months) and aged (24–27 months) mice. White lines: BM. White-dotted lines: cell boundary. Radial histogram quantification of division angles (n = 5, >10 cells/mouse). d, Gating strategy for flow cytometric analysis of IFESCs derived from the plantar epidermis. Error bars show the s.e.m. Two-tailed t-test (a and b). One-sided Kolmogorov–Smirnov test (c). n = number of mice.

Source data

Extended Data Fig. 2 Prolonged calcium flux through Piezo1 causes keratinocyte differentiation and hemidesmosome fragility.

a, Piezo1 fluorescence in situ hybridization (FISH) in plantar skin. Similar results were obtained from three biological independent experiments. White-dotted lines: BM. b, qPCR for Piezo1 in plantar epidermis from aged Piezo1f/f (24 months) and aged Piezo1 cKO (24 months) mice (n = 5). c, Representative immunofluorescence images and fluorescence intensities of plectin in plantar skin from young Piezo1f/f (2 months), aged Piezo1f/f (24 months) and aged Piezo1 cKO mice (24 months) (n = 5). Data point; 30 cell average/mouse. d, Stiffness values for the dermis of young (6 months) and Piezo1 cKO aged (24 months) plantar skin (n = 5, >30 points/mouse). e, Stiffness values for the dermis of young (6 months), aged (24–27 months) plantar skin (n = 5, >30 points/mouse) and soft and stiff gels (n = 70 points on gels). f, Representative immunofluorescence images and quantification of K10+ cells in primary cultured keratinocytes (n = 3 biological replicates, >130 cells/experiment). g, Representative immunofluorescence images and fluorescence intensities of COL17 in primary cultured keratinocytes (n = 3 biological replicates). Data point; 20 cell average/mouse. h, Representative immunofluorescence images and quantification of K10+ cells in primary cultured Piezo1 cKO keratinocytes (n = 3 biological replicates, >120 cells/experiment). i, Representative immunofluorescence images and fluorescence intensities of COL17 in primary cultured Piezo1 cKO keratinocytes (n = 3 biological replicates). Data point; 20 cell average/experiment. Error bars show the s.e.m. Two-tailed t-test (b,d,f,g,h,i), Tukey’s multiple comparison tests (c). n = number of mice.

Source data

Extended Data Fig. 3 Piezo1 agonist Yoda1 induces IFESC dysregulation.

a, Representative immunofluorescence images and quantification of K10+ cells (arrowhead) in the basal layer of young (3 months) plantar skin injected with DMSO or Yoda1 (n = 5, >150 cells/mouse). Aged (24–27 months) plantar skin were injected with DMSO (n = 5, >170 cells/mouse). b, Representative immunofluorescence images and fluorescence intensities of COL17 in plantar skin from young (3 months) plantar skin injected with DMSO or Yoda1 (n = 5). Aged (24–27 months) plantar skin were injected with DMSO (n = 5). Data point; 30 cell average/mouse. c, Representative immunofluorescence images and fluorescence intensities of plectin in plantar skin from young (3 months) plantar skin injected with DMSO or Yoda1 (n = 5). Aged (24–27 months) plantar skin were injected with DMSO (n = 5). Data point; 30 cell average/mouse. d, Representative immunofluorescence images of survivin in IFESCs from young (3 months) plantar skin injected with DMSO or Yoda1. Aged (24–27 months) plantar skin were injected with DMSO. White lines: BM. White-dotted lines: cell boundary. Radial histogram quantification of division angles (n = 5, >10 cells/mouse). e, Representative immunofluorescence images and quantification of K10+ cells in primary keratinocytes after treatment with DMSO or Yoda1 (n = 3 biological replicates, >120 cells/experiment). f, Representative immunofluorescence images and fluorescence intensities of COL17 in primary keratinocytes after treatment with DMSO or Yoda1 (n = 3 biological replicates). Data point; 20 cell average/experiment. Error bars show the s.e.m. Tukey’s multiple comparison tests (a,b,c), One-sided Kolmogorov–Smirnov test (d), and two-tailed t-test (e,f). n = number of mice.

Source data

Extended Data Fig. 4 Regional variability in aging-associated IFESC dysregulation.

a, Representative immunofluorescence images and fluorescence intensities of COL17 in back skin from young (6 months) and aged (24–27 months) mice (n = 5). Data point; 30 cell average/mouse. b, Representative immunofluorescence images and fluorescence intensities of COL17 in tail skin from young (6 months) and aged (24–27 months) mice (n = 5). Data point; 30 cell average/mouse c, Representative immunofluorescence images and quantification of K10+ cells (arrowhead) in the basal layer of tail skin from young (6 months) and aged (24–27 months) mice (n = 5, >150 cells/mouse). d, Representative immunofluorescence images and fluorescence intensities of COL17 in tail skin from young Piezo1f/f (6 months), aged Piezo1f/f (24–27 months) and aged Piezo1 cKO (24–27 months) mice (n = 5). Data point; 30 cell average/mouse e, Representative immunofluorescence images and quantification of K10 + cells (arrowhead) in the basal layer of tail skin from young Piezo1f/f (6 months), aged Piezo1f/f (24–27 months) and aged Piezo1 cKO (24–27 months) (n = 5, >150 cells/mouse). White-dotted lines: BM (c and e). Error bars show the s.e.m. Two-tailed t-test (a,b,c) and Tukey’s multiple comparison tests (d,e). n = number of mice.

Source data

Extended Data Fig. 5 Skin vasculatures regulate dermal ECM and epidermal hemidesmosome fragility.

a, Representative immunofluorescence images and quantification of the length of elastin fibers in plantar skin from young (6 months) and aged (24–27 months) mice (n = 5, 10 points/mouse). White-dotted lines: BM. b, qPCR for VEGFA in plantar skin from aged WT (24–27 months) and aged K14VEGF (22 months) mice (n = 5). c, Representative immunofluorescence images and fluorescence intensities of plectin in plantar skin from young WT (2 months), aged WT (22 months), and K14VEGF mice (22 months). Data point; 30 cell average/mouse. d, qPCR for VEGFR2 in the plantar dermis from VEGFR2f/f (6 months) and VEGFR2 cKO (6 months) mice (n = 5). e, Representative immunofluorescence images and fluorescence intensities of plectin in plantar skin from VEGFR2f/f (6 months) and VEGFR2 cKO (6 months) mice (n = 5). Data point; 30 cell average/mouse. Error bars show the s.e.m. Two-tailed t-test (a,b,d,e), and Tukey’s multiple comparison tests (c). n = number of mice.

Source data

Extended Data Fig. 6 scRNA-seq analysis of mouse plantar dermis from young and aged mice.

a, Whole plantar dermal cell transcriptomes (n = 9,839) from two young (6 months) and two aged (24–27 months) mice plotted on a UMAP plot. The 15 clusters identified by unsupervised clustering are separated by color. Identity and marker genes of each cluster are indicated below the plots. b, Expression of cluster-specific marker genes projected onto UMAP plots.

Extended Data Fig. 7 Cell–cell communication analysis between endothelial cells and fibroblasts during aging.

a, Ligand–receptor interactions inferred by CellPhoneDB were shown. P values calculated in CellPhoneDB were indicated by circle size (scale on top). The means of the average expression level of interacting molecules are indicated by color. b, Violin plots showing the expression of ECM-related genes (Col1a1, Col1a2, Col3a1, Eln and Sparc), angiogenic genes (Sparc and Thy1) and anti-angiogenic genes (Thbs1 and Egr1) in fibroblast clusters (CL-4 and CL-12) from young and aged mice (two mice each). Wilcox test.

Extended Data Fig. 8 Feature plots for each fibroblast cluster.

Expression of cluster-specific marker genes projected onto UMAP plots. Samples were collected from two young and two aged mice.

Extended Data Fig. 9 Aging-induced IFESC dysregulation is ameliorated in Ptx3 KO plantar skin.

a, qPCR for Ptx3 in the plantar dermis from aged WT (24–27 months) and aged Ptx3 KO (24–27 months) mice (n = 5). b, Representative immunofluorescence images and quantification of the length of elastin fibers in plantar skin from young WT (2 months), aged WT (24–27 months), and aged Ptx3 KO (24–27 months) mice (n = 5, 10 points/mouse). White-dotted lines: BM. c, Representative immunofluorescence images and fluorescence intensities of plectin in plantar skin from young WT (2 months), aged WT (24–27 months), and aged Ptx3 KO (24–27 months) mice (n = 5). Data point; 30 cell average/mouse Error bars show the s.e.m. Two-tailed t-test (a), Tukey’s multiple comparison tests (b,c). n = number of mice.

Source data

Extended Data Fig. 10 Cellular senescence is partially rescued in Piezo1 cKO, K14VEGF, and Ptx3 KO mice.

a, Representative immunofluorescence images and fluorescence intensities of 8-OHdG in tail skin from young Piezo1f/f (6 months), aged Piezo1f/f (24–27 months) and aged Piezo1 cKO (24–27 months) mice (n = 5). Data point; 30 cell average/mouse. b, Representative immunofluorescence images and fluorescence intensities of 8-OHdG in plantar skin from young WT (2 months), aged WT (22 months) and aged K14VEGF mice (22 months) (n = 5). Data point; 30 cell average/mouse c, Representative immunofluorescence images and fluorescence intensities of 8-OHdG in tail skin from young WT (6 months), aged WT (24–27 months) and aged Ptx3 KO (24–27 months) mice (n = 5). Data point; 30 cell average/mouse. d, Schematic of the mechanism of age-associated IFESC dysregulation. White-dotted lines: BM (a,b,c). Error bars show the s.e.m. Tukey’s multiple comparison tests (a,b,c). n = number of mice.

Source data

Supplementary information

Reporting Summary

Supplementary Table 1

Top 50 DEGs in each dermal cluster.

Supplementary Table 2

Top 50 DEGs in each fibroblast cluster.

Source data

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Ichijo, R., Maki, K., Kabata, M. et al. Vasculature atrophy causes a stiffened microenvironment that augments epidermal stem cell differentiation in aged skin. Nat Aging 2, 592–600 (2022). https://doi.org/10.1038/s43587-022-00244-6

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s43587-022-00244-6

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