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:

Skin chronological aging drives age-related bone loss via secretion of cystatin-A

An Author Correction to this article was published on 26 June 2023

This article has been updated

Abstract

Although clinical evidence has indicated an association between skin atrophy and bone loss during aging, their causal relationship and the underlying mechanisms are unknown. Here we show that premature skin aging drives bone loss in mice. We further identify that cystatin-A (Csta), a keratinocyte-enriched secreted factor, mediates the effect of skin on bone. Keratinocyte-derived Csta binds the receptor for activated C-kinase 1 in osteoblast and osteoclast progenitors, thus promoting their proliferation but inhibiting osteoclast differentiation. Csta secretion decreases with skin aging in both mice and humans, thereby causing senile osteoporosis by differentially decreasing the numbers of osteoblasts and osteoclasts. In contrast, topical application of calcipotriol stimulates Csta production in the epidermis and alleviates osteoporosis. These results reveal a mode of endocrine regulation of bone metabolism in the skin, and identify Csta as an epidermally derived hormone linking skin aging to age-related bone loss. Enhancers of skin Csta levels could serve as a potential topical drug for treatment of senile osteoporosis.

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: Premature skin aging drives bone loss in young mice.
Fig. 2: Csta expression and secretion decrease in aged human skin.
Fig. 3: Csta ablation in the epidermis results in low-turnover-osteopenia in mice.
Fig. 4: Csta overexpression in epidermis alleviates bone loss in aged mice.
Fig. 5: Topical calcipotriol alleviates age-related bone loss in mice through increasing Csta production in keratinocytes.
Fig. 6: Csta stimulates osteoblast proliferation via Rack1.
Fig. 7: Csta inhibits osteoclast formation and function via Rack1.
Fig. 8: Csta competes with Src for binding Rack1.

Similar content being viewed by others

Data availability

Cytokine array data and mass spectrometry data are provided in Supplementary Data 1 and 2 and Supplementary Information. All other data are provided within the article, Source Data and Supplementary Information or are available from the corresponding author upon reasonable request.

Change history

References

  1. Manolagas, S. C. & Jilka, R. L. Bone marrow, cytokines, and bone remodeling. Emerging insights into the pathophysiology of osteoporosis. N. Engl. J. Med. 332, 305–311 (1995).

    CAS  PubMed  Google Scholar 

  2. Kawaguchi, H. et al. Independent impairment of osteoblast and osteoclast differentiation in klotho mouse exhibiting low-turnover osteopenia. J. Clin. Invest. 104, 229–237 (1999).

    CAS  PubMed  PubMed Central  Google Scholar 

  3. Russell-Goldman, E. & Murphy, G. F. The pathobiology of skin aging: new insights into an old dilemma. Am. J. Pathol. 190, 1356–1369 (2020).

    PubMed  PubMed Central  Google Scholar 

  4. McConkey, B., Fraser, G. M., Bligh, A. S. & Whiteley, H. Transparent skin and osteoporosis. Lancet 281, 693–695 (1963).

    Google Scholar 

  5. Black, M. M., Shuster, S. & Bottoms, E. Osteoporosis, skin collagen, and androgen. Br. Med. J. 4, 773–774 (1970).

    CAS  PubMed  PubMed Central  Google Scholar 

  6. Chappard, D., Alexandre, C., Robert, J. M. & Riffat, G. Relationships between bone and skin atrophies during aging. Acta Anat. (Basel) 141, 239–244 (1991).

    CAS  PubMed  Google Scholar 

  7. Whitmore, S. E. & Levine, M. A. Risk factors for reduced skin thickness and bone density: possible clues regarding pathophysiology, prevention, and treatment. J. Am. Acad. Dermatol. 38, 248–255 (1998).

    CAS  PubMed  Google Scholar 

  8. Villeneuve, D., Lidove, O., Chazerain, P., Ziza, J. M. & Sené, T. Association between dermatoporosis and history of major osteoporotic fractures: a French prospective observational study in a general practice population. Joint Bone Spine 87, 511–512 (2020).

    PubMed  Google Scholar 

  9. Shuster, S. Osteoporosis, like skin ageing, is caused by collagen loss which is reversible. J. R. Soc. Med. 113, 158–160 (2020).

    PubMed  PubMed Central  Google Scholar 

  10. Sirufo, M. M., De Pietro, F., Bassino, E. M., Ginaldi, L. & De Martinis, M. Osteoporosis in skin diseases. Int. J. Mol. Sci. 21, 4749 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  11. Dayoub, J. C., Cortese, F., Anžič, A., Grum, T. & de Magalhães, J. P. The effects of donor age on organ transplants: a review and implications for aging research. Exp. Gerontol. 110, 230–240 (2018).

    PubMed  PubMed Central  Google Scholar 

  12. Nelson, G. et al. A senescent cell bystander effect: senescence-induced senescence. Aging Cell 11, 345–349 (2012).

    CAS  PubMed  Google Scholar 

  13. Acosta, J. C. et al. A complex secretory program orchestrated by the inflammasome controls paracrine senescence. Nat. Cell Biol. 15, 978–990 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  14. Yousefzadeh, M. J. et al. An aged immune system drives senescence and ageing of solid organs. Nature 594, 100–105 (2021).

  15. Giangreco, A., Goldie, S. J., Failla, V., Saintigny, G. & Watt, F. M. Human skin aging is associated with reduced expression of the stem cell markers beta1 integrin and MCSP. J. Invest. Dermatol. 130, 604–608 (2010).

    CAS  PubMed  Google Scholar 

  16. Umbayev, B. et al. Galactose-induced skin aging: the role of oxidative stress. Oxid. Med. Cell. Longev. 2020, 7145656 (2020).

  17. Riggs, B. L. & Melton, L. J. 3rd Involutional osteoporosis. N. Engl. J. Med. 314, 1676–1686 (1986).

    CAS  PubMed  Google Scholar 

  18. Hu, L. et al. Epidermal dysfunction leads to an age-associated increase in levels of serum inflammatory cytokines. J. Invest. Dermatol. 137, 1277–1285 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  19. Moschen, A. R. et al. The RANKL/OPG system is activated in inflammatory bowel disease and relates to the state of bone loss. Gut 54, 479–487 (2005).

    CAS  PubMed  PubMed Central  Google Scholar 

  20. Redlich, K. & Smolen, J. S. Inflammatory bone loss: pathogenesis and therapeutic intervention. Nat. Rev. Drug Discov. 11, 234–250 (2012).

    CAS  PubMed  Google Scholar 

  21. Kato, T., Takai, T., Mitsuishi, K., Okumura, K. & Ogawa, H. Cystatin A inhibits IL-8 production by keratinocytes stimulated with Der p 1 and Der f 1: biochemical skin barrier against mite cysteine proteases. J. Allergy Clin. Immunol. 116, 169–176 (2005).

    CAS  PubMed  Google Scholar 

  22. Hall, G. & Phillips, T. J. Estrogen and skin: the effects of estrogen, menopause, and hormone replacement therapy on the skin. J. Am. Acad. Dermatol. 53, 555–568 (2005).

    PubMed  Google Scholar 

  23. Sallach, J. et al. Tropism-modified AAV vectors overcome barriers to successful cutaneous therapy. Mol. Ther. 22, 929–939 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  24. Tohmonda, T. et al. IRE1α/XBP1-mediated branch of the unfolded protein response regulates osteoclastogenesis. J. Clin. Invest. 125, 3269–3279 (2015).

    PubMed  PubMed Central  Google Scholar 

  25. Steven, A. C. & Steinert, P. M. Protein composition of cornified cell envelopes of epidermal keratinocytes. J. Cell Sci. 107, 693–700 (1994).

    CAS  PubMed  Google Scholar 

  26. Blaydon, D. C. et al. Mutations in CSTA, encoding Cystatin A, underlie exfoliative ichthyosis and reveal a role for this protease inhibitor in cell-cell adhesion. Am. J. Hum. Genet. 89, 564–571 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  27. Walters, M. R. Newly identified actions of the vitamin D endocrine system. Endocr. Rev. 13, 719–764 (1992).

    CAS  PubMed  Google Scholar 

  28. Hosomi, J., Hosoi, J., Abe, E., Suda, T. & Kuroki, T. Regulation of terminal differentiation of cultured mouse epidermal cells by 1 alpha,25-dihydroxyvitamin D3. Endocrinology 113, 1950–1957 (1983).

    CAS  PubMed  Google Scholar 

  29. Smith, E. L., Walworth, N. C. & Holick, M. F. Effect of 1 alpha,25-dihydroxyvitamin D3 on the morphologic and biochemical differentiation of cultured human epidermal keratinocytes grown in serum-free conditions. J. Invest. Dermatol. 86, 709–714 (1986).

    CAS  PubMed  Google Scholar 

  30. Takahashi, H. et al. 1, 25-Dihydroxyvitamin D 3 increases human cystatin A expression by inhibiting the Raf-1/MEK1/ERK signaling pathway of keratinocytes. Arch. Dermatol. Res. 295, 80–87 (2003).

    CAS  PubMed  Google Scholar 

  31. Cunningham, T. J. et al. Randomized trial of calcipotriol combined with 5-fluorouracil for skin cancer precursor immunotherapy. J. Clin. Invest. 127, 106–116 (2017).

    PubMed  Google Scholar 

  32. Kragballe, K. & Iversen, L. Calcipotriol. A new topical antipsoriatic. Dermatol. Clin. 11, 137–141 (1993).

    CAS  PubMed  Google Scholar 

  33. Li, M. et al. Topical vitamin D3 and low-calcemic analogs induce thymic stromal lymphopoietin in mouse keratinocytes and trigger an atopic dermatitis. Proc. Natl Acad. Sci. USA 103, 11736–11741 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

  34. Chandler, P. D. et al. Reduction of parathyroid hormone with vitamin D supplementation in blacks: a randomized controlled trial. BMC Nutr. 1, 26 (2015).

    PubMed  Google Scholar 

  35. Bianco, P. et al. The meaning, the sense and the significance: translating the science of mesenchymal stem cells into medicine. Nat. Med. 19, 35–42 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  36. Boyle, W. J., Simonet, W. S. & Lacey, D. L. Osteoclast differentiation and activation. Nature 423, 337–342 (2003).

    CAS  PubMed  Google Scholar 

  37. Teitelbaum, S. L. Bone resorption by osteoclasts. Science 289, 1504–1508 (2000).

    CAS  PubMed  Google Scholar 

  38. Adams, D. R., Ron, D. & Kiely, P. A. RACK1, a multifaceted scaffolding protein: structure and function. Cell Commun. Signal. 9, 22 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  39. Mamidipudi, V. et al. RACK1 inhibits colonic cell growth by regulating Src activity at cell cycle checkpoints. Oncogene 26, 2914–2924 (2007).

    CAS  PubMed  Google Scholar 

  40. Lin, J., Lee, D., Choi, Y. & Lee, S. Y. The scaffold protein RACK1 mediates the RANKL-dependent activation of p38 MAPK in osteoclast precursors. Sci. Signal. 8, ra54 (2015).

    PubMed  PubMed Central  Google Scholar 

  41. Park, J. H. et al. RACK1 interaction with c-Src is essential for osteoclast function. Exp. Mol. Med. 51, 86 (2019).

    PubMed  PubMed Central  Google Scholar 

  42. Chang, B. Y., Chiang, M. & Cartwright, C. A. The interaction of Src and RACK1 is enhanced by activation of protein kinase C and tyrosine phosphorylation of RACK1. J. Biol. Chem. 276, 20346–20356 (2001).

    CAS  PubMed  Google Scholar 

  43. Chang, B. Y., Harte, R. A. & Cartwright, C. A. RACK1: a novel substrate for the Src protein-tyrosine kinase. Oncogene 21, 7619–7629 (2002).

    CAS  PubMed  Google Scholar 

  44. Tong, L. et al. Crystal structures of the human p56lck SH2 domain in complex with two short phosphotyrosyl peptides at 1.0 A and 1.8 A resolution. J. Mol. Biol. 256, 601–610 (1996).

    CAS  PubMed  Google Scholar 

  45. Lause, M., Kamboj, A. & Fernandez Faith, E. Dermatologic manifestations of endocrine disorders. Transl. Pediatr. 6, 300–312 (2017).

    PubMed  PubMed Central  Google Scholar 

  46. Robinson, R. J., Al-Azzawi, F., Iqbal, J. S., Abrams, K. & Mayberry, J. F. The relation of hand skin-fold thickness to bone mineral density in patients with Crohn’s disease. Eur. J. Gastroenterol. Hepatol. 9, 945–949 (1997).

    CAS  PubMed  Google Scholar 

  47. Savvas, M. et al. The effect of anorexia nervosa on skin thickness, skin collagen and bone density. Br. J. Obstet. Gynaecol. 96, 1392–1394 (1989).

    CAS  PubMed  Google Scholar 

  48. Van Dijk, F. S. & Sillence, D. O. Osteogenesis imperfecta: clinical diagnosis, nomenclature and severity assessment. Am. J. Med. Genet. A 164, 1470–1481 (2014).

    PubMed Central  Google Scholar 

  49. Stevenson, S. & Thornton, J. Effect of estrogens on skin aging and the potential role of SERMs. Clin. Interv. Aging 2, 283–297 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  50. Denu, R. A. et al. Fibroblasts and mesenchymal stromal/stem cells are phenotypically indistinguishable. Acta Haematol. 136, 85–97 (2016).

    CAS  PubMed  Google Scholar 

  51. Gil, A., Plaza-Diaz, J. & Mesa, M. D. Vitamin D: classic and novel actions. Ann. Nutr. Metab. 72, 87–95 (2018).

    CAS  PubMed  Google Scholar 

  52. Zenz, R. et al. Psoriasis-like skin disease and arthritis caused by inducible epidermal deletion of Jun proteins. Nature 437, 369–375 (2005).

    CAS  PubMed  Google Scholar 

  53. Vasilopoulos, Y. et al. A nonsynonymous substitution of cystatin A, a cysteine protease inhibitor of house dust mite protease, leads to decreased mRNA stability and shows a significant association with atopic dermatitis. Allergy 62, 514–519 (2007).

    CAS  PubMed  Google Scholar 

  54. Krunic, A. L., Stone, K. L., Simpson, M. A. & McGrath, J. A. Acral peeling skin syndrome resulting from a homozygous nonsense mutation in the CSTA gene encoding cystatin A. Pediatr. Dermatol. 30, e87–e88 (2013).

    PubMed  Google Scholar 

  55. Moosbrugger-Martinz, V. et al. Epidermal barrier abnormalities in exfoliative ichthyosis with a novel homozygous loss-of-function mutation in CSTA. Br. J. Dermatol. 172, 1628–1632 (2015).

    CAS  PubMed  Google Scholar 

  56. Vasilopoulos, Y. et al. Association analysis of the skin barrier gene cystatin A at the PSORS5 locus in psoriatic patients: evidence for interaction between PSORS1 and PSORS5. Eur. J. Hum. Genet. 16, 1002–1009 (2008).

    CAS  PubMed  Google Scholar 

  57. Ritchlin, C. T., Haas-Smith, S. A., Li, P., Hicks, D. G. & Schwarz, E. M. Mechanisms of TNF-alpha- and RANKL-mediated osteoclastogenesis and bone resorption in psoriatic arthritis. J. Clin. Invest. 111, 821–831 (2003).

    CAS  PubMed  PubMed Central  Google Scholar 

  58. Ekström, U. et al. Internalization of cystatin C in human cell lines. FEBS J. 275, 4571–4582 (2008).

    PubMed  PubMed Central  Google Scholar 

  59. Di Giaimo, R. et al. New insights into the molecular basis of progressive myoclonus epilepsy: a multiprotein complex with cystatin B. Hum. Mol. Genet. 11, 2941–2950 (2002).

    PubMed  Google Scholar 

  60. Horne, W. C., Sanjay, A., Bruzzaniti, A. & Baron, R. The role(s) of Src kinase and Cbl proteins in the regulation of osteoclast differentiation and function. Immunol. Rev. 208, 106–125 (2005).

    CAS  PubMed  Google Scholar 

  61. Zaidi, S. K. et al. Tyrosine phosphorylation controls Runx2-mediated subnuclear targeting of YAP to repress transcription. EMBO J. 23, 790–799 (2004).

    CAS  PubMed  PubMed Central  Google Scholar 

  62. Nakashima, K. et al. The novel zinc finger-containing transcription factor osterix is required for osteoblast differentiation and bone formation. Cell 108, 17–29 (2002).

    CAS  PubMed  Google Scholar 

  63. Choi, Y. H. et al. Src enhances osteogenic differentiation through phosphorylation of Osterix. Mol. Cell. Endocrinol. 407, 85–97 (2015).

    CAS  PubMed  Google Scholar 

  64. Muzumdar, M. D., Tasic, B., Miyamichi, K., Li, L. & Luo, L. A global double-fluorescent Cre reporter mouse. Genesis 45, 593–605 (2007).

    CAS  PubMed  Google Scholar 

  65. Opie, S. R., Warrington, K. H. Jr., Agbandje-McKenna, M., Zolotukhin, S. & Muzyczka, N. Identification of amino acid residues in the capsid proteins of adeno-associated virus type 2 that contribute to heparan sulfate proteoglycan binding. J. Virol. 77, 6995–7006 (2003).

    CAS  PubMed  PubMed Central  Google Scholar 

  66. Acosta, J. C. et al. Chemokine signaling via the CXCR2 receptor reinforces senescence. Cell 133, 1006–1018 (2008).

    CAS  PubMed  Google Scholar 

  67. Huang, M. J. et al. Enhancement of the synthesis of n-3 PUFAs in fat-1 transgenic mice inhibits mTORC1 signalling and delays surgically induced osteoarthritis in comparison with wild-type mice. Ann. Rheum. Dis. 73, 1719–1727 (2014).

    CAS  PubMed  Google Scholar 

  68. Czekanska, E. M., Stoddart, M. J., Richards, R. G. & Hayes, J. S. In search of an osteoblast cell model for in vitro research. Eur. Cell. Mater. 24, 1–17 (2012).

    CAS  PubMed  Google Scholar 

  69. Li, F., Adase, C. A. & Zhang, L. J. Isolation and culture of primary mouse keratinocytes from neonatal and adult mouse skin. J. Vis. Exp. https://doi.org/10.3791/56027 (2017).

  70. Deng, P. et al. Loss of KDM4B exacerbates bone-fat imbalance and mesenchymal stromal cell exhaustion in skeletal aging. Cell Stem Cell https://doi.org/10.1016/j.stem.2021.01.010 (2021).

  71. Ouyang, Z. et al. DEPTOR exacerbates bone-fat imbalance in osteoporosis by transcriptionally modulating BMSC differentiation. Biomed. Pharmacother. 151, 113164 (2022).

    CAS  PubMed  Google Scholar 

  72. Yi, Z. et al. A novel role for c-Src and STAT3 in apoptotic cell-mediated MerTK-dependent immunoregulation of dendritic cells. Blood 114, 3191–3198 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  73. Chen, A. et al. mTORC1 induces plasma membrane depolarization and promotes preosteoblast senescence by regulating the sodium channel Scn1a. Bone Res. 10, 25 (2022).

    CAS  PubMed  PubMed Central  Google Scholar 

  74. Huang, B. et al. Osteoblasts secrete Cxcl9 to regulate angiogenesis in bone. Nat. Commun. 7, 13885 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  75. Rosenfeld, L. et al. Nanobodies targeting prostate-specific membrane antigen for the imaging and therapy of prostate cancer. J. Med. Chem. 63, 7601–7615 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

This research was supported by grant nos. 82172507 (B.H.), 81700783 (B.H.) and 81991511 (X.B.) from the National Natural Science Foundation of China, nos. 2019A1515011876 (B.H.) and 2018A030313937 (Z.L.) from Guangdong Natural Science Fund Management Committee and no. 202002030176 (B.H.) from Guangzhou Municipal Science and Technology Bureau. The funders had no role in study design, data collection and analysis, decision to publish or preparation of the manuscript.

Author information

Authors and Affiliations

Authors

Contributions

B.H. conceived the ideas for experimental designs and wrote the manuscript. W.L., Q.C. and S.C. conducted the majority of the experiments, analyzed data and helped with manuscript preparation. R.W. and Y.L. maintained mice, collected tissue samples and performed micro-CT analyses. C.Y. and Z.O. conducted cell cultures and immunoblot experiments. D.K., A.C., Z.L. and K.L. performed immunohistochemistry, immunofluorescence and confocal imaging. X.B. and Q.L. developed the concept, supervised the project and critically reviewed the manuscript.

Corresponding author

Correspondence to Bin Huang.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature Aging thanks Thea Mauro, Marc Wein 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 Chemokines and cytokines detected in the serum from young and older male patients with a cytokine antibody array.

a, Scatter plot showing the distribution levels of the 640 cytokines detected in the serum of the patients. Red dots show cytokines up-regulated ≥1.2-fold from older relative to young patients; blue dots show cytokines down-regulated ≥1.2-fold. b, Volcano plot illustrating the magnitude and significance of the differences in cytokine serum concentrations in the patients. Green dots are cytokines with a significantly different serum concentration between groups (True, fold change ≥1.2, p < 0.05), red dots were not different (False).

Extended Data Fig. 2 Skin presents the most abundant Csta expression among tissues in the body.

a, Immunofluorescence staining of Csta in the indicated tissues of adult mice. Dashed lines, basement membrane (skin) or boundary between growth plate and bone marrow (bone). Arrowheads, positively stained cells. BM, bone marrow. Scale bars, 100 μm. Experiments were repeated six times with similar results. b, Real-time PCR analysis of Csta expression levels in the indicated tissues from wild-type mice. Data are presented as mean ± s.d. The number of biological replicates per group is indicated.

Source data

Extended Data Fig. 3 Csta expression and secretion decreases in the skin of aged mice.

a, Immunohistochemical staining of Csta in the skin of young (3 months old) and aged (20 months old) wild-type male mice. Bottom, magnified views of dashed boxed areas. Scale bar, 100 μm. b, c, Protein and mRNA levels of Csta in the mouse skin were analysed by western blotting and qPCR, respectively. d, ELISA analysis of Csta levels in the serum and bone marrow in the mice. e, f, H&E staining in sections of the mouse skin, and quantification of epidermal thickness and cell numbers. Bottom, magnified views of boxed areas (e). Scale bar, 100 μm. g-j, Immunostaining of p16 (cell senescence markers) (g) and Ki-67 (i) (cell proliferation marker) in sections of mouse skin. Arrowheads, positively stained cells. Epidermal cells positively stained by the markers are quantified as their numbers as a percentage of total cells (h, j). Scale bar, 100 μm. k, l, Representative images of micro-CT analyses and quantitative analysis of trabecular and cortical bone microstructure of metaphyseal in the distal femurs from the mice. Scale bar, 500 μm. m, Individual correlations between serum Csta levels and trabecular and cortical bone mass in 21 aged (20-month-old) wild-type mice. Dashed lines, basement membrane. P values were calculatedwith two-tailed Student’s t-test, and all data are presented as mean ± s.d. The number of biological replicates per group and P values are indicated. Experiments were repeated six times (a, e, k), three times (b), nine times (g), and six or seven times (i), with similar results.

Source data

Extended Data Fig. 4 Diminished Csta levels in ovariectomized mice.

a, Fatty body of ovariectomized (OVX) mice relative to their Sham controls. b, Atrophic uterus due to oestrogen deficiency in OVX mice. c, d, Representative images of micro-CT analyses and quantitative analysis of trabecular and cortical bone microstructure of metaphyseal in the distal femurs from the mice. Scale bar, 500 μm. e, f, H&E staining in sections of mice skin, and quantification of epidermal thickness and cell numbers. Scale bar, 100 μm. g, h, Immunohistochemical staining and quantification of the staining intensity of Csta in mice skin in the two groups. Scale bar, 100 μm. i, ELISA analysis of Csta levels in mice serum and bone marrow supernatants. P values were calculated with two-tailed Student’s t-test, and all data are presented as mean ± s.d. The number of biological replicates per group and P values are indicated. Experiments were repeated five times (c) and six times (e, g) with similar results.

Source data

Extended Data Fig. 5 Establishment of epidermis specific and inducible Csta knockout mice.

a, Structure G/Rflox transgenes and schematic diagram of the G/Rflox construct before and after Cre-mediated recombination. G/R consists of a CAG promoter driving a loxP flanked coding sequence of green fluorescent protein (ZsGreen). After Cre-mediated intra-chromosomal recombination, the ZsGreen sequence is excised, thus allowing the promoter to drive expression of the membrane-targeted tandem dimer Tomato (tdTomato). b, G/Rflox transgenic mice were topically treated with AAV2-Cre (G/RAAV2-Cre) or AAV2 (G/RAAV2, control) vector on a patch of dorsal skin once per 7 days for 1 month. The fluorescence of the indicated tissues was observed to monitor AAV2-Cre recombinase activity. AAV2-Cre recombinase was activated in the skin of G/Rflox mice but not in other tissues. Dashed lines, basement membrane; arrowheads, tdTomato-positive regions. Right images are magnified views of the boxed areas (bone). Scale bars, 100 μm. Experiments were repeated five times with similar results.

Extended Data Fig. 6 Csta knockout causes epidermal thinning in mice.

Two-month-old male Cstafl/fl mice were topically treated with AAV2-Cre (CstaAAV2-Cre) or AAV2 (CstaAAV2, control) on shaved patches on dorsal skin once per 7 days for 1 month. The appearance of patches on dorsal skin was photographed (a). b, c, H&E staining in sections of the skin patches, and quantification of epidermal thickness and cell numbers. Bottom, magnified views of boxed areas (b). d, e, Immunostaining of Ki-67 and quantification of Ki-67 positive cells as a percentage of total epidermal cells. Bottom, magnified views of boxed areas (d). f, g, Immunostaining of K10 (a marker for keratinocyte differentiation) and K14 and quantification of K10 positive cells as a percentage of total epidermal cells. Scale bars, 50 μm. P values were calculated with two-tailed Student’s t-test, and all data are presented as mean ± s.d. The number of biological replicates per group and P values are indicated. Experiments were repeated nine times (b), six times (d), and eight or nine (f), with similar results.

Source data

Extended Data Fig. 7 Representative images of FACS isolation of murine osteoclast precursor cells.

Cells were first gated by forward scatter (FSC) and side scatter (SSC) to remove doublets. Osteoclast precursor cells were identified by negative gating for CD11b, and positive gating for CD115 and CD117.

Extended Data Fig. 8 Calcipotriol increases Csta production in keratinocytes.

a-d, Representative images of K10 staining, and quantification of K10 positive cells in dorsal skin from young and older male patients (a, b) and mice (c, d). Right, magnified views of dashed boxed areas (a). Dashed lines, basement membrane. Arrowheads, positively stained cells (c). e-g, Primary mouse keratinocytes were induced to differentiate with 0.2 mM CaCl2 and treated with calcipotriol or left untreated for the indicated times. Csta mRNA, protein, and secretion levels in the cells were analysed by qPCR (e), western blotting (f), and ELISA (g), respectively. h, CSTA-luciferase reporter plasmid or empty vector were transfected into HaCaT cells under low Ca2+ (0.06 mM) conditions. After incubation for 24 h, the transfected cells were incubated with and 10–9 M calcipotriol and 50 µmol/L ceramide C6 (ERK activator) for 12 h, and subjected to detection of ERK phosphorylation and luciferase activity. i, Serum calcium levels and body weights in mice receiving topical calcipotriol or vehicle (Control). j, k, Primary mouse keratinocytes were treated with vehicle (Control) or 10-9 M calcipotriol in the presence of the indicated concentrations of CaCl2 and subjected to detection of cell proliferation rates with EdU assays. Scale bars: 50 μm in a, 20 μm in c, and 200 μm in i. P values were calculated with ANOVA or two-tailed Student’s t-test, and all data are presented as mean ± s.d. The number of biological replicates per group and P values are indicated. Experiments were repeated eight times (a), eight or nine times (c), three times (f, h), and 10 times (j), with similar results.

Source data

Extended Data Fig. 9 Csta promotes proliferation of osteoclast precursors via Rack1.

a, b, CstaAAV2-Cre and CstaAAV2 mice were injected intraperitoneally with EdU. After 4 hours, bone marrow cells were isolated freshly from bilateral femurs and tibias, and subjected to flow cytometric analysis of osteoclast precursor proliferation. Representative flow cytometry plots (a) and the frequencies of proliferative osteoclast precursors (EdU+ CD11bdull) in CD117+- and CD115+-gated bone marrow cells (b) are presented. c-i, Raw264.7 cells stably transfected with Flag-Rack1 or lentiviral vector were treated with recombinant Csta or vehicle. c, d, Representative images and quantification of cell proliferation with EdU assays. Scale bar, 100 μm. e-g, Cells were analysed for cell cycle distribution by flow cytometry (e, f) and for expression and activity of G1 cell cycle regulators (g). h, i, Cell apoptosis, assessed by flow cytometry with Annexin V/PI staining. j, Flow cytometric analysis of osteoclast precursor apoptosis in bone marrow cells freshly isolated from bilateral femurs and tibias of CstaAAV2-Cre and CstaAAV2 mice. Frequencies of Annexin V+ apoptotic cells in CD117+-, CD115+- and CD11bdull- gated bone marrow cells are quantified as apoptosis rates of osteoclast precursors. P values were calculated with ANOVA or two-tailed Student’s t-test, and all data are presented as mean ± s.d. The number of biological replicates per group and P values are indicated. Experiments were repeated three times (a, e, g, h, j), and six times (c), with similar results.

Source data

Extended Data Fig. 10 Csta competes with Src for binding Rack1 in osteoblasts.

MC3T3-E1 cells stably transfected with Flag-Rack1 or lentiviral vector were serum-starved for 5 hours, then treated with recombinant GST-Csta, GST or vehicle (Veh) plus 10%FBS for 1 hour. Proteins in cell lysates were precipitated with anti-Rack1 and subjected to immunoblot analysis with anti-Src and anti-GST (a), or precipitated with anti-Src and analyzed for in vitro protein kinase activity against recombinant Stat3 (b). Experiments were repeated three times (a, b) with similar results.

Source data

Supplementary information

Supplementary Information

Supplementary Tables 1–6.

Reporting Summary

Supplementary Data 1

Cytokine array data.

Supplementary Data 2

Mass spectrometry data.

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 Fig. 6

Unprocessed immunoblots.

Source Data Fig. 7

Statistical Source Data.

Source Data Fig. 7

Unprocessed immunoblots.

Source Data Fig. 8

Unprocessed immunoblots.

Source Data Extended Data Fig. 2

Statistical Source Data.

Source Data Extended Data Fig. 3

Statistical Source Data.

Source Data Extended Data Fig. 3

Unprocessed immunoblots.

Source Data Extended Data Fig. 4

Statistical Source Data.

Source Data Extended Data Fig. 6

Statistical Source Data.

Source Data Extended Data Fig. 8

Statistical Source Data.

Source Data Extended Data Fig. 8

Unprocessed immunoblots.

Source Data Extended Data Fig. 9

Statistical Source Data.

Source Data Extended Data Fig. 9

Unprocessed immunoblots.

Source Data Extended Data Fig. 10

Unprocessed immunoblots.

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Liang, W., Chen, Q., Cheng, S. et al. Skin chronological aging drives age-related bone loss via secretion of cystatin-A. Nat Aging 2, 906–922 (2022). https://doi.org/10.1038/s43587-022-00285-x

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s43587-022-00285-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