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:

NFIB is a governor of epithelial–melanocyte stem cell behaviour in a shared niche

Subjects

Abstract

Adult stem cells reside in specialized niches where they receive environmental cues to maintain tissue homeostasis. In mammals, the stem cell niche within hair follicles is home to epithelial hair follicle stem cells and melanocyte stem cells, which sustain cyclical bouts of hair regeneration and pigmentation1,2,3,4. To generate pigmented hairs, synchrony is achieved such that upon initiation of a new hair cycle, stem cells of each type activate lineage commitment2,5. Dissecting the inter-stem-cell crosstalk governing this intricate coordination has been difficult, because mutations affecting one lineage often affect the other. Here we identify transcription factor NFIB as an unanticipated coordinator of stem cell behaviour. Hair follicle stem-cell-specific conditional targeting of Nfib in mice uncouples stem cell synchrony. Remarkably, this happens not by perturbing hair cycle and follicle architecture, but rather by promoting melanocyte stem cell proliferation and differentiation. The early production of melanin is restricted to melanocyte stem cells at the niche base. Melanocyte stem cells more distant from the dermal papilla are unscathed, thereby preventing hair greying typical of melanocyte stem cell differentiation mutants. Furthermore, we pinpoint KIT-ligand as a dermal papilla signal promoting melanocyte stem cell differentiation. Additionally, through chromatin-immunoprecipitation with high-throughput-sequencing and transcriptional profiling, we identify endothelin 2 (Edn2) as an NFIB target aberrantly activated in NFIB-deficient hair follicle stem cells. Ectopically induced Edn2 recapitulates NFIB-deficient phenotypes in wild-type mice. Conversely, endothelin receptor antagonists and/or KIT blocking antibodies prevent precocious melanocyte stem cell differentiation in the NFIB-deficient niche. Our findings reveal how melanocyte and hair follicle stem cell behaviours maintain reliance upon cooperative factors within the niche, and how this can be uncoupled in injury, stress and disease states.

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

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

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

Figure 1: Conditional Nfib targeting in hair follicle stem cells does not perturb hair cycle or follicle architecture.
Figure 2: NFIB loss enhances melanocyte stem-cell self-renewal and perturbs melanocyte stem-cells activity in the hair follicle stem cell niche.
Figure 3: Premature transfer of pigment promotes apoptotic cell death in hair follicle stem cells in the NFIB-deficient niche.
Figure 4: RNA-seq and ChIP-seq analyses identify Edn2 as a direct NFIB-regulated gene mediating inter-stem cell crosstalk.

Similar content being viewed by others

Accession codes

Primary accessions

Gene Expression Omnibus

Data deposits

ChIP-seq data have been deposited in the Gene Expression Omnibus database under accession number GSE42900.

References

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

    Article  ADS  CAS  Google Scholar 

  2. Nishimura, E. K. et al. Dominant role of the niche in melanocyte stem cell fate determination. Nature 416, 854–860 (2002)

    Article  ADS  CAS  Google Scholar 

  3. Blanpain, C. & Fuchs, E. Epidermal homeostasis: a balancing act of stem cells in the skin. Nature Rev. Mol. Cell Biol. 10, 207–217 (2009)

    Article  CAS  Google Scholar 

  4. Cotsarelis, G. Gene expression profiling gets to the root of human hair follicle stem cells. J. Clin. Invest. 116, 19–22 (2006)

    Article  CAS  Google Scholar 

  5. Hirobe, T. How are proliferation and differentiation of melanocytes regulated? Pigment Cell Melanoma Res. 24, 462–478 (2011)

    Article  CAS  Google Scholar 

  6. Rabbani, P. et al. Coordinated activation of Wnt in epithelial and melanocyte stem cells initiates pigmented hair regeneration. Cell 145, 941–955 (2011)

    Article  CAS  Google Scholar 

  7. Nishimura, E. K. et al. Key roles for transforming growth factor β in melanocyte stem cell maintenance. Cell Stem Cell 6, 130–140 (2010)

    Article  CAS  Google Scholar 

  8. Greco, V. et al. A two-step mechanism for stem cell activation during hair regeneration. Cell Stem Cell 4, 155–169 (2009)

    Article  CAS  Google Scholar 

  9. Tanimura, S. et al. Hair follicle stem cells provide a functional niche for melanocyte stem cells. Cell Stem Cell 8, 177–187 (2011)

    Article  CAS  Google Scholar 

  10. Weiner, L. et al. Dedicated epithelial recipient cells determine pigmentation patterns. Cell 130, 932–942 (2007)

    Article  CAS  Google Scholar 

  11. Inomata, K. et al. Genotoxic stress abrogates renewal of melanocyte stem cells by triggering their differentiation. Cell 137, 1088–1099 (2009)

    Article  CAS  Google Scholar 

  12. Fitch, K. R. et al. Genetics of dark skin in mice. Genes Dev. 17, 214–228 (2003)

    Article  CAS  Google Scholar 

  13. Gründer, A. et al. Nuclear factor I-B (Nfib) deficient mice have severe lung hypoplasia. Mech. Dev. 112, 69–77 (2002)

    Article  Google Scholar 

  14. Dooley, A. L. et al. Nuclear factor I/B is an oncogene in small cell lung cancer. Genes Dev. 25, 1470–1475 (2011)

    Article  CAS  Google Scholar 

  15. Steele-Perkins, G. et al. The transcription factor gene Nfib is essential for both lung maturation and brain development. Mol. Cell. Biol. 25, 685–698 (2005)

    Article  CAS  Google Scholar 

  16. 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  Google Scholar 

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

    Article  CAS  Google Scholar 

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

    Article  Google Scholar 

  19. Lien, W. H. et al. Genome-wide maps of histone modifications unwind in vivo chromatin states of the hair follicle lineage. Cell Stem Cell 9, 219–232 (2011)

    Article  CAS  Google Scholar 

  20. Aoki, H., Yamada, Y., Hara, A. & Kunisada, T. Two distinct types of mouse melanocyte: differential signaling requirement for the maintenance of non-cutaneous and dermal versus epidermal melanocytes. Development 136, 2511–2521 (2009)

    Article  CAS  Google Scholar 

  21. D’Orazio, J. A. et al. Topical drug rescue strategy and skin protection based on the role of Mc1r in UV-induced tanning. Nature 443, 340–344 (2006)

    Article  ADS  Google Scholar 

  22. McDade, S. S. et al. Genome-wide analysis of p63 binding sites identifies AP-2 factors as co-regulators of epidermal differentiation. Nucleic Acids Res. 40, 7190–7206 (2012)

    Article  CAS  Google Scholar 

  23. Pjanic, M. et al. Nuclear factor I revealed as family of promoter binding transcription activators. BMC Genomics 12, 181 (2011)

    Article  CAS  Google Scholar 

  24. Pla, P. & Larue, L. Involvement of endothelin receptors in normal and pathological development of neural crest cells. Int. J. Dev. Biol. 47, 315–325 (2003)

    CAS  PubMed  Google Scholar 

  25. Nishikawa, S. et al. In utero manipulation of coat color 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  Google Scholar 

  26. Beronja, S., Livshits, G., Williams, S. & Fuchs, E. Rapid functional dissection of genetic networks via tissue-specific transduction and RNAi in mouse embryos. Nature Med. 16, 821–827 (2010)

    Article  CAS  Google Scholar 

  27. Baynash, A. G. et al. Interaction of endothelin-3 with endothelin-B receptor is essential for development of epidermal melanocytes and enteric neurons. Cell 79, 1277–1285 (1994)

    Article  CAS  Google Scholar 

  28. Reid, K. et al. Multiple roles for endothelin in melanocyte development: regulation of progenitor number and stimulation of differentiation. Development 122, 3911–3919 (1996)

    CAS  PubMed  Google Scholar 

  29. Adur, J., Takizawa, S., Uchide, T., Casco, V. & Saida, K. High doses of ultraviolet-C irradiation increases vasoactive intestinal contractor/endothelin-2 expression in keratinocytes of the newborn mouse epidermis. Peptides 28, 1083–1094 (2007)

    Article  CAS  Google Scholar 

  30. Klipper, E. et al. Induction of endothelin-2 expression by luteinizing hormone and hypoxia: possible role in bovine corpus luteum formation. Endocrinology 151, 1914–1922 (2010)

    Article  CAS  Google Scholar 

  31. Hsu, Y. C. et al. Mesenchymal nuclear factor I B regulates cell proliferation and epithelial differentiation during lung maturation. Dev. Biol. 354, 242–252 (2011)

    Article  CAS  Google Scholar 

  32. Soeda, T. et al. Sox9-expressing precursors are the cellular origin of the cruciate ligament of the knee joint and the limb tendons. Genesis 48, 635–644 (2010)

    Article  CAS  Google Scholar 

  33. Morris, R. J. et al. Capturing and profiling adult hair follicle stem cells. Nature Biotechnol. 22, 411–417 (2004)

    Article  CAS  Google Scholar 

  34. Srinivas, S. et al. Cre reporter strains produced by targeted insertion of EYFP and ECFP into the ROSA26 locus. BMC Dev. Biol. 1, 4 (2001)

    Article  CAS  Google Scholar 

  35. Heintz, N. Gene expression nervous system atlas (GENSAT). Nature Neurosci. 7, 483 (2004)

    Article  CAS  Google Scholar 

  36. Govender, D., Davids, L. M. & Kidson, S. H. Immunofluorescent identification of melanocytes in murine hair follicles. J. Mol. Histol. 37, 1–3 (2006)

    Article  Google Scholar 

  37. Trapnell, C., Pachter, L. & Salzberg, S. L. TopHat: discovering splice junctions with RNA-Seq. Bioinformatics 25, 1105–1111 (2009)

    Article  CAS  Google Scholar 

  38. Trapnell, C. et al. Transcript assembly and quantification by RNA-Seq reveals unannotated transcripts and isoform switching during cell differentiation. Nature Biotechnol. 28, 511–515 (2010)

    Article  CAS  Google Scholar 

  39. Goodarzi, H., Elemento, O. & Tavazoie, S. Revealing global regulatory perturbations across human cancers. Mol. Cell 36, 900–911 (2009)

    Article  CAS  Google Scholar 

  40. Giannopoulou, E. G. & Elemento, O. An integrated ChIP-seq analysis platform with customizable workflows. BMC Bioinformatics 12, 277 (2011)

    Article  Google Scholar 

  41. Langmead, B., Trapnell, C., Pop, M. & Salzberg, S. L. Ultrafast and memory-efficient alignment of short DNA sequences to the human genome. Genome Biol. 10, R25 (2009)

    Article  Google Scholar 

  42. Elemento, O., Slonim, N. & Tavazoie, S. A universal framework for regulatory element discovery across all genomes and data types. Mol. Cell 28, 337–350 (2007)

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank Y.-C. Hsu, T. Chen, B. Keyes, S. E. Williams, A. Rodriguez-Folgueras and other Fuchs laboratory members for intellectual input and suggestions; L. Polak and N. Stokes for breeding of mouse lines and conducting in utero lentiviral injections; V. J. Hearing for providing anti-DCT, TYRP1 and TYR antibodies. We also thank Rockefeller facilities: Comparative Bioscience Center (AAALAC accredited) for care and husbandry care of mice in accordance with National Institutes of Health (NIH) guidelines; Bioimaging Center for advice on image acquisition; Flow Cytometry facility for FACS sorting. E.F. is an investigator of the Howard Hughes Medical Institute. This work was supported by grants from the NIH to E.F. (R01-AR050452 and R01-AR31737) and R.M.G. (R01-HL080624), and a CAREER grant to O.E. from the National Science Foundation (DB1054964).

Author information

Authors and Affiliations

Authors

Contributions

C.-Y.C. performed all experiments; H.A.P. performed the ultrastructural analyses; E.G.G. and O.E. performed the bioinformatic analyses; G.G. carried out the initial characterization of NFIB expression during mouse development; R.M.G. provided the conditional Nfibfl/fl mice; E.F. supervised the project; E.F. and C.-Y.C. wrote the manuscript.

Corresponding author

Correspondence to Elaine Fuchs.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Supplementary information

Supplementary Information

This file contains Supplementary Figures 1-15 and Supplementary Tables 1-2. (PDF 2637 kb)

PowerPoint slides

Rights and permissions

Reprints and permissions

About this article

Cite this article

Chang, CY., Pasolli, H., Giannopoulou, E. et al. NFIB is a governor of epithelial–melanocyte stem cell behaviour in a shared niche. Nature 495, 98–102 (2013). https://doi.org/10.1038/nature11847

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nature11847

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