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:

Distinct fibroblast lineages determine dermal architecture in skin development and repair

Subjects

Abstract

Fibroblasts are the major mesenchymal cell type in connective tissue and deposit the collagen and elastic fibres of the extracellular matrix (ECM)1. Even within a single tissue, fibroblasts exhibit considerable functional diversity, but it is not known whether this reflects the existence of a differentiation hierarchy or is a response to different environmental factors. Here we show, using transplantation assays and lineage tracing in mice, that the fibroblasts of skin connective tissue arise from two distinct lineages. One forms the upper dermis, including the dermal papilla that regulates hair growth and the arrector pili muscle, which controls piloerection. The other forms the lower dermis, including the reticular fibroblasts that synthesize the bulk of the fibrillar ECM, and the preadipocytes and adipocytes of the hypodermis. The upper lineage is required for hair follicle formation. In wounded adult skin, the initial wave of dermal repair is mediated by the lower lineage and upper dermal fibroblasts are recruited only during re-epithelialization. Epidermal β-catenin activation stimulates the expansion of the upper dermal lineage, rendering wounds permissive for hair follicle formation. Our findings explain why wounding is linked to formation of ECM-rich scar tissue that lacks hair follicles2,3,4. They also form a platform for discovering fibroblast lineages in other tissues and for examining fibroblast changes in ageing and disease.

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

Figure 1: Morphological and molecular markers of embryonic and postnatal fibroblasts.
Figure 2: Skin-reconstitution assays.
Figure 3: Fibroblast lineage commitment during skin development.
Figure 4: Contribution of fibroblast lineages in adult skin.

Similar content being viewed by others

References

  1. Kalluri, R. & Zeisberg, M. Fibroblasts in cancer. Nature Rev. Cancer 6, 392–401 (2006)

    Article  CAS  Google Scholar 

  2. Gurtner, G. C., Werner, S., Barrandon, Y. & Longaker, M. T. Wound repair and regeneration. Nature 453, 314–321 (2008)

    Article  ADS  CAS  Google Scholar 

  3. Martin, P. Wound healing–aiming for perfect skin regeneration. Science 276, 75–81 (1997)

    Article  CAS  Google Scholar 

  4. Shah, M., Foreman, D. M. & Ferguson, M. W. Neutralising antibody to TGF-β1,2 reduces cutaneous scarring in adult rodents. J. Cell Sci. 107, 1137–1157 (1994)

    CAS  PubMed  Google Scholar 

  5. Van Exan, R. J. & Hardy, M. H. The differentiation of the dermis in the laboratory mouse. Am. J. Anat. 169, 149–164 (1984)

    Article  CAS  Google Scholar 

  6. Dulauroy, S., Di Carlo, S. E., Langa, F., Eberl, G. & Peduto, L. Lineage tracing and genetic ablation of ADAM12+ perivascular cells identify a major source of profibrotic cells during acute tissue injury. Nature Med. 18, 1262–1270 (2012)

    Article  CAS  Google Scholar 

  7. Janson, D. G., Saintigny, G., van Adrichem, A., Mahe, C. & El Ghalbzouri, A. Different gene expression patterns in human papillary and reticular fibroblasts. J. Invest. Dermatol. 132, 2565–2572 (2012)

    Article  CAS  Google Scholar 

  8. Collins, C. A., Kretzschmar, K. & Watt, F. M. Reprogramming adult dermis to a neonatal state through epidermal activation of β-catenin. Development 138, 5189–5199 (2011)

    Article  CAS  Google Scholar 

  9. Jensen, K. B., Driskell, R. R. & Watt, F. M. Assaying proliferation and differentiation capacity of stem cells using disaggregated adult mouse epidermis. Nature Protocols 5, 898–911 (2010)

    Article  CAS  Google Scholar 

  10. Festa, E. et al. Adipocyte lineage cells contribute to the skin stem cell niche to drive hair cycling. Cell 146, 761–771 (2011)

    Article  CAS  Google Scholar 

  11. Driskell, R. R. et al. Clonal growth of dermal papilla cells in hydrogels reveals intrinsic differences between Sox2-positive and -negative cells in vitro and in vivo. J. Invest. Dermatol. 132, 1084–1093 (2012)

    Article  CAS  Google Scholar 

  12. Lesko, M. H., Driskell, R. R., Kretzschmar, K., Goldie, S. J. & Watt, F. M. Sox2 modulates the function of two distinct cell lineages in mouse skin. Dev. Biol. 382, 15–26 (2013)

    Article  CAS  Google Scholar 

  13. Robertson, E. J. et al. Blimp1 regulates development of the posterior forelimb, caudal pharyngeal arches, heart and sensory vibrissae in mice. Development 134, 4335–4345 (2007)

    Article  CAS  Google Scholar 

  14. Horsley, V. et al. Blimp1 defines a progenitor population that governs cellular input to the sebaceous gland. Cell 126, 597–609 (2006)

    Article  CAS  Google Scholar 

  15. Magnúsdóttir, E. et al. Epidermal terminal differentiation depends on B lymphocyte-induced maturation protein-1. Proc. Natl Acad. Sci. USA 104, 14988–14993 (2007)

    Article  ADS  Google Scholar 

  16. Gomez, C. et al. The interfollicular epidermis of adult mouse tail comprises two distinct cell lineages that are differentially regulated by Wnt, Edaradd, and Lrig1. Stem Cell Reports 1, 19–27 (2013)

    Article  CAS  Google Scholar 

  17. Page, M. E., Lombard, P., Ng, F., Gottgens, B. & Jensen, K. B. The epidermis comprises autonomous compartments maintained by distinct stem cell populations. Cell Stem Cell 13, 471–482 (2013)

    Article  CAS  Google Scholar 

  18. Egawa, G., Osawa, M., Uemura, A., Miyachi, Y. & Nishikawa, S. Transient expression of ephrin b2 in perinatal skin is required for maintenance of keratinocyte homeostasis. J. Invest. Dermatol. 129, 2386–2395 (2009)

    Article  CAS  Google Scholar 

  19. Schmidt, B. A. & Horsley, V. Intradermal adipocytes mediate fibroblast recruitment during skin wound healing. Development 140, 1517–1527 (2013)

    Article  CAS  Google Scholar 

  20. Ito, M. et al. Wnt-dependent de novo hair follicle regeneration in adult mouse skin after wounding. Nature 447, 316–320 (2007)

    Article  ADS  CAS  Google Scholar 

  21. Silva-Vargas, V. et al. β-catenin and Hedgehog signal strength can specify number and location of hair follicles in adult epidermis without recruitment of bulge stem cells. Dev. Cell 9, 121–131 (2005)

    Article  CAS  Google Scholar 

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

    Article  ADS  CAS  Google Scholar 

  23. Staal, F. J., Luis, T. C. & Tiemessen, M. M. WNT signalling in the immune system: WNT is spreading its wings. Nature Rev. Immunol. 8, 581–593 (2008)

    Article  CAS  Google Scholar 

  24. Gay, D. et al. Fgf9 from dermal γδ T cells induces hair follicle neogenesis after wounding. Nature Med. 19, 916–923 (2013)

    Article  CAS  Google Scholar 

  25. 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  Google Scholar 

  26. Arwert, E. N. et al. Tumor formation initiated by nondividing epidermal cells via an inflammatory infiltrate. Proc. Natl Acad. Sci. USA 107, 19903–19908 (2010)

    Article  ADS  CAS  Google Scholar 

  27. Driskell, R. R., Giangreco, A., Jensen, K. B., Mulder, K. W. & Watt, F. M. Sox2-positive dermal papilla cells specify hair follicle type in mammalian epidermis. Development 136, 2815–2823 (2009)

    Article  CAS  Google Scholar 

  28. Rivers, L. E. et al. PDGFRA/NG2 glia generate myelinating oligodendrocytes and piriform projection neurons in adult mice. Nature Neurosci. 11, 1392–1401 (2008)

    Article  CAS  Google Scholar 

  29. Kawamoto, S. et al. A novel reporter mouse strain that expresses enhanced green fluorescent protein upon Cre-mediated recombination. FEBS Lett. 470, 263–268 (2000)

    Article  CAS  Google Scholar 

  30. Hamilton, T. G., Klinghoffer, R. A., Corrin, P. D. & Soriano, P. Evolutionary divergence of platelet-derived growth factor alpha receptor signaling mechanisms. Mol. Cell. Biol. 23, 4013–4025 (2003)

    Article  CAS  Google Scholar 

Download references

Acknowledgements

This work was funded by the Wellcome Trust (F.M.W., A.C.F.-S.), the Medical Research Council (MRC) (F.M.W., A.C.F.-S.) and the European Union FP7 programme: TUMIC (F.M.W.), HEALING (F.M.W.) and EpigeneSys (A.C.F.-S.). B.M.L. is the recipient of a FEBS long-term fellowship. K.K. is the recipient of a MRC PhD Studentship. The authors acknowledge financial support from the Department of Health via the National Institute for Health Research (NIHR) comprehensive Biomedical Research Centre award to Guy’s & St Thomas’ NHS Foundation Trust in partnership with King’s College London (KCL) and King’s College Hospital NHS Foundation Trust. Input from M. Mastrogiannaki, A. Reimer and B. Trappmann is gratefully acknowledged. The authors also thank J. Connelly and G. Theocharidis for providing tissue, KCL core staff, in particular J. Castle, C. Pearce and D. Cooper, for technical support, and J. Harris and the Nikon Imaging Centre at KCL for expert assistance.

Author information

Authors and Affiliations

Authors

Contributions

R.R.D. and F.M.W. designed the experiments, performed data analysis, interpreted the results and wrote the manuscript. B.M.L., E.H., K.K., A.C.F.-S., S.R.F., B.D.S. and M.C. assisted in performing and designing experiments, analysing data and interpreting results. Y.H. and G.P. generated the Dlk1-CreER transgenic mouse.

Corresponding author

Correspondence to Fiona M. Watt.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Extended data figures and tables

Extended Data Figure 1 Expression of panel of markers during back skin development.

Sections were counterstained with DAPI (blue). Scale bars, 50 μm. Photomicrographs of 10-μm-thick sections are representative of three biological replicates.

Extended Data Figure 2 Expression of markers during back skin development.

a, Expression of additional markers. Counterstained with DAPI (blue). b, Co-expression of CD31, CD45, CD26, Dlk1 and Sca1. Counterstained with DAPI (blue). Scale bars, 50 μm. Photomicrographs of 10-μm-thick sections are representative of three biological replicates.

Extended Data Figure 3 Isolation and in vitro properties of dermal subpopulations.

ac, Flow cytometry of E12.5, E14.5, E16.5 and P2 Pdgfr-α-H2B–eGFP mouse dermal cells labelled with antibodies to CD26 (a), Dlk1 (b) and Sca1 (c). Graphs show average percentage of cells that co-express individual markers and GFP. Data from three biological replicates at each time point. dg, Clonal growth of fibroblast subpopulations from E16.5 (d, e) and P2 (f, g) dermis. d, f, Representative phase contrast and LipidTOX fluorescent images of clones encapsulated in hydrogels and cultured in DMEM plus 10% bovine serum or adipogenic differentiation medium. e, g, Quantification of lipid-positive colonies. n = 3 biological replicates. One-way ANOVA parametric tests of significance were used to determine P values: *P < 0.05; **P < 0.005; ***P < 0.0005. Error bars are ± s.e.m. Scale bars, 10 μm.

Extended Data Figure 4 Fibroblast marker expression in human skin.

a, c, eh, Neonatal human foreskin. b, d, Adult human abdomen. a, b, PDGFR-α was detected in mesenchymal (vimentin+) cells throughout the dermis. DLK1 (c, d, f), CD26 (e, g) and PDPN (h) were differentially expressed in upper (CD26) or lower (DLK1, PDPN) neonatal dermis. DLK1 was not expressed by adult dermal fibroblasts (d). Scale bars, 50 μm. n = 2 biological replicates.

Extended Data Figure 5 Further characterization of P2 mouse dermal fibroblast subpopulations.

a, b, 10-μm sections of P2 skin stained for markers indicated, with DAPI counterstain (white). c, Flow-sorting strategy to isolate cells from P2 papillary dermis (CD26+Sca1), reticular/hypodermis (Dlk1+/Sca1) and hypodermis (Dlk1+Sca1+ and Dlk1Sca1+). d, Heat map of relative messenger RNA levels of the genes indicated that are represented in histograms, summarizing data in e. e, Characterization of flow-sorted fibroblasts from P2 dermis. Relative mRNA levels of the genes indicated, normalized to Gapdh. Data are means of three biological replicates (± s.e.m.). One-way ANOVA parametric tests of significance were used to determine P values: *P < 0.05; **P < 0.005; ***P < 0.0005. fi, Preadipocyte precursors (LinCD34+CD29+Sca1+) express Pdgfr-α. Single-cell suspension of P2 Pdgfr-α-H2B–eGFP mouse dermis was gated for Lin(f), CD29+CD34+(g), Sca1+ (h) and GFP+(i). Scale bars, 50 μm. All data are representative of three biological replicates.

Extended Data Figure 6 Skin reconstitution with different fibroblast subpopulations.

ai, See Fig. 2a for experimental set-up. ad, Horizontal whole-mount micrographs of grafts immunostained for eGFP and LipidTOX with DAPI counterstain (blue). Boundary between dermis and hypodermis/adipocytes is indicated by dotted line. eh, Representative macroscopic views of skin grafts. i, Number of hair follicles per graft. Error bars are ± s.e.m. n = 4 grafts per fibroblast population. One-way ANOVA parametric tests were used to determine significance. *P < 0.05. Scale bars, 100 μm (ad) and 400 μm (eh). jl, Representative flow-cytometry plots from exclusion sorts of Pdgfr-α-H2B–eGFP dermis for grafting assays in Fig. 2g, k. j, Positive control: all dermal cells collected. k, No reticular/hypodermis: Dlk1+ and Sca1+ cells were not collected. l, No papillary/reticular dermal sorts: Dlk+ Sca1 and CD26+ Sca1 cells were not collected.

Extended Data Figure 7 Lineage tracing controls.

a, Dlk1-CreER and LSL-tdTomato mice were bred and at 16.5 days of gestation were injected with 5 mg ml−1 tamoxifen in corn oil. b, c, Embryos were collected at E17.5 and single-cell suspensions of labelled embryonic dermis were analysed for overlapping tdTomato expression and endogenous Dlk1 expression. b, Representative flow cytometry plot. c, Graph is representative of n = 4 embryos. Error bars are s.e.m. dg, Blimp1 expression in embryonic and postnatal skin of wild-type (d) and Blimp1–GFP (eg) mice. Horizontal whole mounts were stained for endogenous Blimp1 (d, f), CD26 (e) and Itga8 (g). h, i, Horizontal whole mounts of Bimp1–GFP (h) and Blimp1-Cre/LSL-GFP (i) skin co-stained for CD31. j, k, E18.5 single-cell suspensions of Blimp1–GFP (j) and Blimp1-Cre/LSL-GFP (k) cells labelled with CD26. l, Experimental approach for Lrig1-CreER labelling of papillary dermis. m, Horizontal whole-mount stain of Lrig1-CreER/LSL-tdTomato mouse skin labelled at E16.5 and collected at P2. np, Flow-cytometric analysis of labelling specificity of tdTomato compared to Lrig1 (n), Pdgfr-α (o) and Sca1 (p). q, Flow-cytometric analysis of overlapping expression of Lrig1–GFP with CD26. Data are representative of three biological replicates. Scale bars, 100 μm.

Extended Data Figure 8 Clonal analysis of papillary dermis.

a, Experimental strategy and flow cytometry of dermal cells isolated from mice treated with increasing concentrations of tamoxifen. b, Percentage of Pdgfr-α+ cells 4 days after treatment with different concentrations of tamoxifen (n = 7 per concentration). 25 mg g−1 tamoxifen resulted in less than 1% of fibroblasts being labelled. c, d, Whole-mount analysis of GFP-labelled cells in E16.5 skin. e, f, j, Histological analysis of the fate of papillary clones from Pdgfr-α-CreER × LSL-GFP mice injected with 25 mg g−1 tamoxifen at E12.5 and harvested at P2 (e), P10 (j) and P21 (f). gi, Quantification of the fate of papillary fibroblast clones derived from E12.5 embryonic mesenchyme. g, Number of cells per clone. h, i, Number of cells per cross-compartmental clone of papillary and arrector pili cells (h) and pure arrector pili cells (i). n = 3 biological replicates (± s.e.m.). Scale bars, 50 μm. One-way ANOVA parametric tests were used to test for significance. ***P < 0.0005.

Extended Data Figure 9 Bone marrow transplantation, wound healing and β-catenin activation.

ak, Bone marrow (BM) reconstitution experiments. a, Schematic representation of the experiments. Bone marrow of mice that express eGFP under the control of the CAG promoter (β-actin CMV promoter (ACP)–GFP)) or Pdgfr-α-H2B–eGFP mice was transplanted into sublethally irradiated wild-type mice. One month later, 8-mm diameter full-thickness circular wounds were made in the back skin with a biopsy punch. Skin was analysed 5 and 10 days after wounding. b, c, Y chromosome staining of reconstituted spleen from ACP–GFP (b) and Pdgfr-α-H2B–eGFP mice (c). d, e, Horizontal whole mounts of wild-type mouse back skin reconstituted with bone marrow from ACP–GFP (d) or Pdgfr-α-H2B–eGFP (e) mice, stained for GFP and Pdgfr-α with DAPI counterstain (blue). fi, Horizontal whole mounts of wild-type mouse back skin reconstituted with bone marrow from ACP–GFP (f, h) and Pdgfr-α-H2B–eGFP (g, i) mice 5 (f, g) and 10 (h, i) days after wounding, stained for GFP and Pdgfr-α with DAPI counterstain (blue). Arrows indicate wounded regions of skin. Scale bars, 40 μm (b, c), 100 μm (di). j, k, Flow cytometry of GFP+ dermal cells. j, representative plots. k, Quantification of GFP+cells. ACP–GFP, n = 6 biological replicates; Pdgfr-α-H2B–eGFP, n = 5 biological replicates. One-way ANOVA parametric tests were used to test for significance. ***P < 0.0005. lp, Fibroblast responses to wounding and epidermal β-catenin activation. ln, Fibroblast lineages during full thickness wound repair. Pdgfr-α-CreER/LSL-tdTomato lineage tracing was performed as indicated (l) and wound beds were stained for tdTomato (green) and Lrig1 (red) (m, n). Scale bars, 150 and 50 μm. Micrographs are representative of n = 3 female biological replicates. o, p, Experimental strategy (o) and histological analysis (p) of wild-type and K14ΔNβ-cateninER transgenic mice co-stained with Itga8 (green) and Sca1 (red). n = 3 biological replicates. Scale bars, 100 μm. Number of tamoxifen (4OHT) treatments is indicated.

Extended Data Figure 10 Fibroblast lineages during full thickness wound repair.

ad, Dlk1-CreER/LSL-tdTomato (a, b) and Blimp1/LSL-GFP (c, d) lineage tracing was performed as indicated (a, c) and wound beds were stained for PDGFRα, Itga8, Acta2, CD45 and CD31. Scale bars, 50 μm. Micrographs are representative of n = 3 female biological replicates. eg, Quantitative analysis of single-cell suspensions of Pdgfr-α-H2B–eGFP single (control) and K14ΔNβ-cateninER/Pdgfr-α-H2B–eGFP double-transgenic mice co-stained with Lrig1-PE (e) or Itga8-APC (f) or Sca1-PE (g). n = 3 biological replicates. h, Horizontal whole mounts of control and K14bcatER skin immunstained for Lrig1 (green) and Sca1 (red). n = 6 female biological replicates. Scale bars, 100 μm. Number of tamoxifen (4OHT) treatments is indicated.

Supplementary information

Supplementary Information

This file contains Supplementary Text and additional references. (PDF 176 kb)

PowerPoint slides

Rights and permissions

Reprints and permissions

About this article

Cite this article

Driskell, R., Lichtenberger, B., Hoste, E. et al. Distinct fibroblast lineages determine dermal architecture in skin development and repair. Nature 504, 277–281 (2013). https://doi.org/10.1038/nature12783

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nature12783

This article is cited by

Comments

By submitting a comment you agree to abide by our Terms and Community Guidelines. If you find something abusive or that does not comply with our terms or guidelines please flag it as inappropriate.

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