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:

Dominant role of the niche in melanocyte stem-cell fate determination

Abstract

Stem cells—which have the capacity to self-renew and generate differentiated progeny—are thought to be maintained in a specific environment known as a niche1,2,3. The localization of the niche, however, remains largely obscure for most stem-cell systems. Melanocytes (pigment cells) in hair follicles proliferate and differentiate closely coupled to the hair regeneration cycle4. Here we report that stem cells of the melanocyte lineage can be identified, using Dct-lacZ transgenic mice5,6, in the lower permanent portion of mouse hair follicles throughout the hair cycle. It is only the population in this region that fulfils the criteria for stem cells, being immature, slow cycling, self-maintaining and fully competent in regenerating progeny on activation at early anagen (the growing phase of hair follicles). Induction of the re-pigmentation process in K14-steel factor transgenic mice7 demonstrates that a portion of amplifying stem-cell progeny can migrate out from the niche and retain sufficient self-renewing capability to function as stem cells after repopulation into vacant niches. Our data indicate that the niche has a dominant role in the fate determination of melanocyte stem-cell progeny.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Purchase on Springer Link

Instant access to full article PDF

Prices may be subject to local taxes which are calculated during checkout

Figure 1: Dct-lacZ+ cells in the bulge area survive in a Kit-independent manner.
Figure 2: Bulge melanoblasts retain BrdU during hair cycling.
Figure 3: Regeneration of functional melanocytes from an implanted lower permanent portion.
Figure 4: LacZ+ melanoblast (arrows) distribution during hair cycling.
Figure 5: Stem cells recolonize empty niches.

Similar content being viewed by others

References

  1. Schofield, R. The relationship between the spleen colony-forming cell and the haemopoietic stem cell. Blood Cells 4, 7–25 (1978)

    CAS  Google Scholar 

  2. Watt, F. M. & Hogan, B. L. Out of Eden: stem cells and their niches. Science 287, 1427–1430 (2000)

    Article  ADS  CAS  Google Scholar 

  3. Xie, T. & Spradling, A. C. A niche maintaining germ line stem cells in the Drosophila ovary. Science 290, 328–330 (2000)

    Article  ADS  CAS  PubMed  Google Scholar 

  4. Slominski, A. & Paus, R. Melanogenesis is coupled to murine anagen: toward new concepts for the role of melanocytes and the regulation of melanogenesis in hair growth. J. Invest. Dermatol. 101, 90s–97s (1993)

    Article  CAS  PubMed  Google Scholar 

  5. Mackenzie, M. A., Jordan, S. A., Budd, P. S. & Jackson, I. J. Activation of the receptor tyrosine kinase Kit is required for the proliferation of melanoblasts in the mouse embryo. Dev. Biol. 192, 99–107 (1997)

    Article  CAS  PubMed  Google Scholar 

  6. Jordan, S. A. & Jackson, I. J. A late wave of melanoblast differentiation and rostrocaudal migration revealed in patch and rump-white embryos. Mech. Dev. 92, 135–143 (2000)

    Article  CAS  PubMed  Google Scholar 

  7. Kunisada, T. et al. Murine cutaneous mastocytosis and epidermal melanocytosis induced by keratinocyte expression of transgenic stem cell factor. J. Exp. Med. 187, 1565–1573 (1998)

    Article  CAS  PubMed  Google Scholar 

  8. 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  Google Scholar 

  9. Oshima, H., Rochat, A., Kedzia, C., Kobayashi, K. & Barrandon, Y. Morphogenesis and renewal of hair follicles from adult multipotent stem cells. Cell 104, 233–245 (2001)

    Article  CAS  PubMed  Google Scholar 

  10. Lavker, R. M. et al. Hair follicle stem cells: their location, role in hair cycle, and involvement in skin tumour formation. J. Invest. Dermatol. 101, 16s–26s (1993)

    Article  CAS  PubMed  Google Scholar 

  11. Chase, H. B. Critical stages of hair development and pigmentation in the mouse. Phisiol. Zool. 24, 1–8 (1951)

    Article  CAS  Google Scholar 

  12. Tobin, D. J., Hagen, E., Botchkarev, V. A. & Paus, R. Do hair bulb melanocytes undergo apoptosis during hair follicle regression (catagen)? J. Invest. Dermatol. 111, 941–947 (1998)

    Article  CAS  PubMed  Google Scholar 

  13. Loeffler, M. & Potten, C. S. in Stem Cells (ed. Potten, S. C.) 5–6 (Academic, London, 1997)

    Google Scholar 

  14. Silver, A. F., Chase, H. B. & Potten, C. S. Melanocyte precursor cells in the hair follicle germ during the dormant stage (telogen). Experientia 25, 299–301 (1968)

    Article  Google Scholar 

  15. Cock, A. G. & Cohen, J. The melanoblast reservoir available to a feather papilla. Embryology 6, 530–545 (1958)

    CAS  Google Scholar 

  16. Sugiyama, S. Mode of redifferentiation and melanogenesis of melanocytes in mouse hair follicles. An ultrastructural and cytochemical study. J. Ultrastruct. Res. 67, 40–54 (1979)

    Article  CAS  PubMed  Google Scholar 

  17. Slominski, A. et al. Pharmacological disruption of hair follicle pigmentation by cyclophosphamide as a model for studying the melanocyte response to and recovery from cytotoxic drug damage in situ. J. Invest. Dermatol. 106, 1203–1211 (1996)

    Article  CAS  PubMed  Google Scholar 

  18. Botchkareva, N. V., Khlgatian, M., Longley, B. J., Botchkarev, V. A. & Gilchrest, B. A. SCF/c-kit signalling is required for cyclic regeneration of the hair pigmentation unit. FASEB J. 15, 645–658 (2001)

    Article  CAS  PubMed  Google Scholar 

  19. Nishikawa, S. et al. In utero manipulation of coat colour formation by a monoclonal anti-c-kit antibody: two distinct waves of c-kit-dependency during melanocyte development. EMBO J. 10, 2111–2118 (1991)

    Article  CAS  PubMed  Google Scholar 

  20. Okura, M., Maeda, H., Nishikawa, S. & Mizoguchi, M. Effects of monoclonal anti-c-kit antibody (ACK2) on melanocytes in newborn mice. J. Invest. Dermatol. 105, 322–328 (1995)

    Article  CAS  PubMed  Google Scholar 

  21. Ogawa, M. et al. Expression and function of c-Kit in fetal hemopoietic progenitor cells: transition from the early c-Kit-independent to the late c-Kit-dependent wave of hemopoiesis in the murine embryo. Development 117, 1089–1098 (1993)

    CAS  PubMed  Google Scholar 

  22. Yoshinaga, K. et al. Role of c-kit in mouse spermatogenesis: identification of spermatogonia as a specific site of c-kit expression and function. Development 113, 689–699 (1991)

    CAS  PubMed  Google Scholar 

  23. Mayer, T. C. The migratory pathway of neural crest cells into the skin of mouse embryos. Dev. Biol. 34, 39–46 (1973)

    Article  CAS  PubMed  Google Scholar 

  24. Nishimura, E. K., Yoshida, H., Kunisada, T. & Nishikawa, S. I. Regulation of E- and P-cadherin expression correlated with melanocyte migration and diversification. Dev. Biol. 215, 155–166 (1999)

    Article  CAS  PubMed  Google Scholar 

  25. Silvers, W. K. The Coat Colours of Mice. A Model for Gene Action and Interaction (Springer, New York, 1979)

    Book  Google Scholar 

  26. Mann, S. J. Prenatal formation of hair follicle types. Anat. Rec. 144, 135–142 (1962)

    Article  Google Scholar 

  27. Friedlich, G. & Soriano, P. Promoter traps in embryonic stem cells: a genetic screen to identify and mutate developmental genes in mice. Genes Dev. 5, 1513–1523 (1991)

    Article  Google Scholar 

  28. Yoshida, H. et al. Neural and skin cell-specific expression pattern conferred by steel factor regulatory sequence in transgenic mice. Dev. Dyn. 207, 222–232 (1996)

    Article  CAS  PubMed  Google Scholar 

  29. Kunisada, T. et al. Transgene expression of steel factor in the basal layer of epidermis promotes survival, proliferation, differentiation and migration of melanocyte precursors. Development 125, 2915–2923 (1998)

    CAS  PubMed  Google Scholar 

  30. Fitzpatrick, T. B., Elsen, A. Z. & Wolff, K. Vitiligo. in Dermatology in General Medicine 3rd edn (ed. Fitzpatrick, T. B.) 810 (McGraw Hill, New York, 1987)

    Google Scholar 

Download references

Acknowledgements

We thank G. Motyckova, R. Yu and A. M. Kenny for critical reading of the manuscript, and E. Nishioka for scientific support. This study was partly supported by grants from the Japanese Ministry of Education, Science and Culture. Work in the MRC Human Genetics Unit is supported by the UK Medical Research Council. E.K.N. was supported by a Research Fellowship from the Japan Society for the Promotion of Science.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Shin-Ichi Nishikawa.

Ethics declarations

Competing interests

The authors declare that they have no competing financial interests

Rights and permissions

Reprints and permissions

About this article

Cite this article

Nishimura, E., Jordan, S., Oshima, H. et al. Dominant role of the niche in melanocyte stem-cell fate determination. Nature 416, 854–860 (2002). https://doi.org/10.1038/416854a

Download citation

  • Received:

  • Accepted:

  • Issue Date:

  • DOI: https://doi.org/10.1038/416854a

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