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:

MicroRNA-205 controls neonatal expansion of skin stem cells by modulating the PI(3)K pathway

Abstract

Skin stem cells (SCs) are specified and rapidly expanded to fuel body growth during early development. However, the molecular mechanisms that govern the amplification of skin SCs remain unclear. Here we report an essential role for miR-205, one of the most highly expressed microRNAs in skin SCs, in promoting neonatal expansion of these cells. Unlike most mammalian miRNAs, genetic deletion of miR-205 causes neonatal lethality with severely compromised epidermal and hair follicle growth. In the miR-205 knockout skin SCs, phospho-Akt is significantly downregulated, and the SCs prematurely exit the cell cycle. In the hair follicle, this accelerates the transition of the neonatal SCs towards quiescence. We identify multiple miR-205-targeted negative regulators of PI(3)K signalling that mediate the repression of phospho-Akt and restrict the proliferation of SCs. Our findings reveal an essential role for miR-205 in maintaining the expansion of skin SCs by antagonizing negative regulators of PI(3)K signalling.

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: miR-205 is highly expressed in skin progenitors and SCs.
Figure 2: Generation of miR-205 KO mice.
Figure 3: Genetic deletion of miR-205 causes neonatal lethality with severe skin defects.
Figure 4: Ablation of miR-205 compromises the proliferation of interfollicular progenitors.
Figure 5: miR-205 is required for the expansion of HFSCs during neonatal development.
Figure 6: Ablation of miR-205 impairs the proliferation of K5+ basal cells in stratified epithelium.
Figure 7: miR-205 directly targets multiple negative regulators of the PI(3)K/Akt pathway.
Figure 8: miR-205 is required to maintain pAkt levels by targeting multiple negative regulators in skin progenitors and SCs.

Similar content being viewed by others

Accession codes

Primary accessions

Gene Expression Omnibus

References

  1. Nowak, J. A., Polak, L., Pasolli, H. A. & Fuchs, E. Hair follicle stem cells are specified and function in early skin morphogenesis. Cell Stem Cell 3, 33–43 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Cotsarelis, G., Sun, T. T. & Lavker, R. M. Label-retaining cells reside in the bulge area of pilosebaceous unit: implications for follicular stem cells, hair cycle, and skin carcinogenesis. Cell 61, 1329–1337 (1990).

    Article  CAS  PubMed  Google Scholar 

  3. Tumbar, T. et al. Defining the epithelial stem cell niche in skin. Science 303, 359–363 (2004).

    Article  CAS  PubMed  Google Scholar 

  4. He, S., Nakada, D. & Morrison, S. J. Mechanisms of stem cell self-renewal. Annu. Rev. Cell Dev. Biol. 25, 377–406 (2009).

    Article  CAS  PubMed  Google Scholar 

  5. Hsu, Y. C. & Fuchs, E. A family business: stem cell progeny join the niche to regulate homeostasis. Nat. Rev. Mol. Cell Biol. 13, 103–114 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Bartel, D. P. MicroRNAs: target recognition and regulatory functions. Cell 136, 215–233 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Yi, R. & Fuchs, E. MicroRNAs and their roles in mammalian stem cells. J. Cell Sci. 124, 1775–1783 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Murchison, E. P., Partridge, J. F., Tam, O. H., Cheloufi, S. & Hannon, G. J. Characterization of Dicer-deficient murine embryonic stem cells. Proc. Natl Acad. Sci. USA 102, 12135–12140 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Andl, T. et al. The miRNA-processing enzyme dicer is essential for the morphogenesis and maintenance of hair follicles. Curr. Biol. 16, 1041–1049 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Yi, R. et al. Morphogenesis in skin is governed by discrete sets of differentially expressed microRNAs. Nat. Genet. 38, 356–362 (2006).

    Article  CAS  PubMed  Google Scholar 

  11. Wang, Y., Medvid, R., Melton, C., Jaenisch, R. & Blelloch, R. DGCR8 is essential for microRNA biogenesis and silencing of embryonic stem cell self-renewal. Nat. Genet. 39, 380–385 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Melton, C., Judson, R. L. & Blelloch, R. Opposing microRNA families regulate self-renewal in mouse embryonic stem cells. Nature 463, 621–626 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Yi, R. et al. DGCR8-dependent microRNA biogenesis is essential for skin development. Proc. Natl Acad. Sci. USA 106, 498–502 (2009).

    Article  CAS  PubMed  Google Scholar 

  14. Liu, N. & Olson, E. N. MicroRNA regulatory networks in cardiovascular development. Dev. Cell 18, 510–525 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Small, E. M. & Olson, E. N. Pervasive roles of microRNAs in cardiovascular biology. Nature 469, 336–342 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Horsley, V., Aliprantis, A. O., Polak, L., Glimcher, L. H. & Fuchs, E. NFATc1 balances quiescence and proliferation of skin stem cells. Cell 132, 299–310 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Hsu, Y. C., Pasolli, H. A. & Fuchs, E. Dynamics between stem cells, niche, and progeny in the hair follicle. Cell 144, 92–105 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Senoo, M., Pinto, F., Crum, C. P. & McKeon, F. p63 Is essential for the proliferative potential of stem cells in stratified epithelia. Cell 129, 523–536 (2007).

    Article  CAS  PubMed  Google Scholar 

  19. Zhang, L., Stokes, N., Polak, L. & Fuchs, E. Specific MicroRNAs Are preferentially expressed by skin stem cells to balance self-renewal and early lineage commitment. Cell Stem Cell 8, 294–308 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Blanpain, C., Lowry, W. E., Geoghegan, A., Polak, L. & Fuchs, E. Self-renewal, multipotency, and the existence of two cell populations within an epithelial stem cell niche. Cell 118, 635–648 (2004).

    Article  CAS  PubMed  Google Scholar 

  21. Landgraf, P. et al. A mammalian microRNA expression atlas based on small RNA library sequencing. Cell 129, 1401–1414 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Guttman, M. et al. Chromatin signature reveals over a thousand highly conserved large non-coding RNAs in mammals. Nature 458, 223–227 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Wang, L., Dowell, R. D. & Yi, R. Genome-wide maps of polyadenylation reveal dynamic mRNA 3’-end formation in mammalian cell lineages. RNA 19, 413–425 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Blanpain, C. & Fuchs, E. Epidermal stem cells of the skin. Annu. Rev. Cell Dev. Biol. 22, 339–373 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Ito, M. et al. Stem cells in the hair follicle bulge contribute to wound repair but not to homeostasis of the epidermis. Nat. Med. 11, 1351–1354 (2005).

    Article  CAS  PubMed  Google Scholar 

  26. Huang da, W., Sherman, B. T. & Lempicki, R. A. Bioinformatics enrichment tools: paths toward the comprehensive functional analysis of large gene lists. Nucleic Acids Res. 37, 1–13 (2009).

    Article  PubMed  Google Scholar 

  27. Park, I. K. et al. Bmi-1 is required for maintenance of adult self-renewing haematopoietic stem cells. Nature 423, 302–305 (2003).

    Article  CAS  PubMed  Google Scholar 

  28. Ezhkova, E. et al. EZH1 and EZH2 cogovern histone H3K27 trimethylation and are essential for hair follicle homeostasis and wound repair. Genes Dev. 25, 485–498 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Cheng, T. et al. Hematopoietic stem cell quiescence maintained by p21cip1/waf1. Science 287, 1804–1808 (2000).

    Article  CAS  PubMed  Google Scholar 

  30. Gewinner, C. et al. Evidence that inositol polyphosphate 4-phosphatase type IIis a tumor suppressor that inhibits PI3K signaling. Cancer Cell 16, 115–125 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Yim, E. K. et al. Rak functions as a tumor suppressor by regulating PTEN protein stability and function. Cancer Cell 15, 304–314 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Kawase, T. et al. PH domain-only protein PHLDA3 is a p53-regulated repressor of Akt. Cell 136, 535–550 (2009).

    Article  CAS  PubMed  Google Scholar 

  33. Dyson, J. M. et al. The SH2 domain containing inositol polyphosphate 5-phosphatase-2: SHIP2. Int. J. Biochem. Cell Biol. 37, 2260–2265 (2005).

    Article  CAS  PubMed  Google Scholar 

  34. Yu, J. et al. MicroRNA-184 antagonizes microRNA-205 to maintain SHIP2 levels in epithelia. Proc. Natl Acad. Sci. USA 105, 19300–19305 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Yu, J. et al. MicroRNA-205 promotes keratinocyte migration via the lipid phosphatase SHIP2. FASEB J. 24, 3950–3959 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Alonso, L. et al. Sgk3 links growth factor signaling to maintenance of progenitor cells in the hair follicle. J. Cell Biol. 170, 559–570 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Murayama, K. et al. Akt activation induces epidermal hyperplasia and proliferation of epidermal progenitors. Oncogene 26, 4882–4888 (2007).

    Article  CAS  PubMed  Google Scholar 

  38. Kobielak, K., Stokes, N., de la Cruz, J., Polak, L. & Fuchs, E. Loss of a quiescent niche but not follicle stem cells in the absence of bone morphogenetic protein signaling. Proc. Natl Acad. Sci. USA 104, 10063–10068 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Gangaraju, V. K. & Lin, H. MicroRNAs: key regulators of stem cells. Nat. Rev. Mol. Cell Biol. 10, 116–125 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Ivey, K. N. & Srivastava, D. MicroRNAs as regulators of differentiation and cell fate decisions. Cell Stem Cell 7, 36–41 (2010).

    Article  CAS  PubMed  Google Scholar 

  41. Martinez, N. J. & Gregory, R. I. MicroRNA gene regulatory pathways in the establishment and maintenance of ESC identity. Cell Stem Cell 7, 31–35 (2010).

    Article  CAS  PubMed  Google Scholar 

  42. Miska, E. A. et al. Most Caenorhabditis elegans microRNAs are individually not essential for development or viability. PLoS Genet. 3, 2395–2403 (2007).

    Article  CAS  Google Scholar 

  43. Gregory, P.A. et al. The miR-200 family and miR-205 regulate epithelial to mesenchymal transition by targeting ZEB1 and SIP1. Nat. Cell Biol. 10, 593–601 (2008).

    Article  CAS  PubMed  Google Scholar 

  44. Li, C., Finkelstein, D. & Sherr, C. J. Arf tumor suppressor and miR-205 regulate cell adhesion and formation of extraembryonic endoderm from pluripotent stem cells. Proc. Natl Acad. Sci. USA 110, E1112–E1121 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Rendl, M., Lewis, L. & Fuchs, E. Molecular dissection of mesenchymal-epithelial interactions in the hair follicle. PLoS Biol. 3, e331 (2005).

    Article  PubMed  PubMed Central  Google Scholar 

  46. Rhee, H., Polak, L. & Fuchs, E. Lhx2 maintains stem cell character in hair follicles. Science 312, 1946–1949 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Yi, R., Poy, M. N., Stoffel, M. & Fuchs, E. A skin microRNA promotes differentiation by repressing ‘stemness’. Nature 452, 225–229 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We are grateful to E. Fuchs for K14–RFP mice. We thank T. Blumenthal, T. Cech, B. Cullen, M. Han, M. Winey and X-J. Wang for comments on the manuscript. We thank C. Yang, J. Gao and D. Feng for the generation of miR-205 KO; S. Ha and L. Greiner for assistance in the animal facility; Y. Han for FACS; and G. Voeltz for confocal microscopy. We also thank members of the Yi laboratory for their critical discussions. This publication was made possible by a start-up fund provided by the University of Colorado and Grant Number R00AR054704 and R01AR059697 (R.Y.) and R01GM083300 (E.C.L.).

Author information

Authors and Affiliations

Authors

Contributions

R.Y. conceived the study. D.W. carried out most experiments and analysed the data with assistance from Z.Z. (bioinformatic analysis), E.O. (in situ hybridization), L.W. (ChIP-seq and RNA-seq) and X.F. (target validation). E.C.L. provided critical resources. R.Y. and D.W. wrote the manuscript with input from all authors.

Corresponding author

Correspondence to Rui Yi.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Integrated supplementary information

Supplementary Figure 1 miRNAs are required for the expansion and maintenance of epidermal progenitor and hair follicle stem cells.

(a) At P4.5, the Dicer cKO pup is smaller and shows less developed hair coat and dehydrated skin, compared to the WT littermate. (b) At birth (P0.5), HFSCs are similarly specified and detected in both WT and Dicer cKO as indicated by Nfatc1 staining. (c and d) Loss of HFSCs in the bulge of Dicer cKO skin by P4.5. Nfatc1+ HFSCs are completely absent in Dicer cKO (c). Sox9+ HFSCs are also significantly reduced in the bulge (d). White dotted lines mark the epidermal/dermal boundary; brackets denote the bulge region. IFE, interfollicular epidermis; Epi, epidermis; ORS, outer root sheath; Mx, matrix; Bu, bulge. Scale bars, 20 μm.

Supplementary Figure 2 Expression pattern of miR-205 in adult skin and FACS purification strategy to specifically isolate HFSCs using K14-RFP/Sox9-GFP mice.

(a) By in situ hybridization, expression of miR-205 is universally high in telogen adult skin, while its expression in anagen adult skin shows the highest expression in the bulge region, mimic its expression pattern in neonatal stage. (b) miR-205 expresses at lower level in active anagen HFSCs comparing to quiescent telogen HFSCs. LinCD34+α6hi HFSCs were FACS purified from anagen and telogen HFs and miR-205 level was measured by qRT-PCR. Data shown are mean ± s.d. from 5 independent experiments. ***, P<0.001 by Student’s t-test. (c) FACS purification strategy to specifically isolate HFSCs using K14-RFP/Sox9-GFP mice. P4.5 total back skin is first separated into epidermal sheet and HF/dermis part by dispase treatment. One dissected K14-RFP/Sox9-GFP hair follicle is shown. All K14-RFP positive hair follicle cells are marked by red fluorescence, while hair follicle stem cells, which express Sox9, are marked by green fluorescence. HFSC is purified as K14RFPhi/Sox9GFP+/α6integrin+, ORS is sorted as K14RFPhi/Sox9GFP/α6integrin+, matrix as K14RFPlow/Sox9GFP. White dotted lines mark the epidermal/dermal boundary. Scale bars, 20 μm.

Supplementary Figure 3 miR-205 is expressed in organs with stratified epithelial tissue.

(a) miR-205 is co-expressed with K5 in stratified epithelial tissue. Using in situ hybridization, miR-205 is detected in the epithelial tissue of skin, tongue, oesophagus, stomach and bladder where K5 is expressed. White dotted lines mark the epidermal/dermal boundary. (b) Most vital organs are negative for miR-205. Expression of miR-205 or K5 is not detected in intestine, heart, lung, brown fat, spleen, liver or kidney by in situ hybridization and immunofluorescence staining. Scale bars, 20 μm.

Supplementary Figure 4 miR-205 is conserved in vertebrates.

(a) The only highly conserved region in the primary transcript of miR-205 is the miR-205 hairpin. (b) The mature sequences of miR-205 are conserved in vertebrates but not in lower eukaryotes e.g. worm and fly.

Supplementary Figure 5 Terminal differentiation in both epidermal and hair follicle lineages is not altered in miR-205 KO.

(a) K5 marks the basal layer; K1 marks the spinous layer. (b) Loricrin (Lori) marks the granular layer. (c) AE13 marks the hair shaft. White asterisks denote autofluoscence of stratum corneum; white dotted lines mark the epidermal/dermal boundary. Scale bars are 50 μm.

Supplementary Figure 6 Full-thickness grafting. Back skin from P0.5 littermates of WT and KO was grafted onto nude mice.

(a) Images show the skin right after putting the donor skin onto the host nude mouse. (b) Bandage was removed at 14 days after grafting. At day 15, hair shaft could be seen from the grafted skin and the grafts were taken for analysis.

Supplementary Figure 7 Characterization of the transition from proliferation to quiescence of bulge stem cells in early stage HFs.

(a and b) In E17.5 hair germ (a) and P0.5 rudimentary HFs (b), all the Nfatc1+ bulge stem cells are positive for Ki67, indicating they are actively proliferating. (c) In as early as P1.5 HF, some Nfatc1 cells have already exited cell cycle, become quiescent, marked by Ki67 negative staining. White dotted lines mark the epidermal/dermal boundary; arrowheads indicate Ki67 negative/Nfatc1 positive bulge stem cells in P1.5 HF. Scale bars, 20 μm.

Supplementary Figure 8 miR-205 directly regulates negative regulators of the PI3K/Akt pathway.

(a-c) The scan for 7mer motifs reveals the enrichment of miR-205 seed (1-7, 2-8 and 3-9) matches in the 3′ UTRs of upregulated genes. All 7mer matches to the miR-205 sequences (1-22nt) are determined in the 3′ UTR, 5′ UTR and coding sequences (CDS) of three gene categories: genes that are upregulated more than 20% in the miR-205 KO HFSCs; genes that are downregulated more than 20% in the miR-205 KO HFSCs; and all genes. The percentage of genes from each category that contains any given 7mer match is calculated and plotted in the Y-axis. Note that only 1-7, 2-8 and 3-9 motifs show enrichment in the 3′UTR of the upregulated genes. (d) The same scan for 7mer motifs of miR-1 in the upregulated genes fails to show any enrichment, indicating the specificity of the upregulated genes for miR-205. (e) Base-pairing between the miR-205 seed (red) and its target sites in the 3′ UTR of candidate genes and the mutated target sites (green). (f) Phospho-Akt level is not affected by miR-205 KO in other vital organs. Numbers between panels represent the densitometry values of phosphorylated protein normalized to total levels. (g) Inhibition of the PI3K pathway abolishes the growth of keratinocytes in colony formation assay. Primary keratinocytes were treated with DMSO (control) or the PI3K small molecule inhibitor LY294002. The treatment of LY294002 completely abolishes cell growth.

Supplementary Figure 9 Uncropped images of immunoblots.

Supplementary information

Supplementary Information

Supplementary Information (PDF 2668 kb)

Supplementary Table 1

Supplementary Information (XLSX 49 kb)

Supplementary Table 2

Supplementary Information (XLSX 12 kb)

Rights and permissions

Reprints and permissions

About this article

Cite this article

Wang, D., Zhang, Z., O’Loughlin, E. et al. MicroRNA-205 controls neonatal expansion of skin stem cells by modulating the PI(3)K pathway. Nat Cell Biol 15, 1153–1163 (2013). https://doi.org/10.1038/ncb2827

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/ncb2827

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